WO2002096359A2 - Piperazine pentanamide hiv protease inhibitors - Google Patents

Piperazine pentanamide hiv protease inhibitors Download PDF

Info

Publication number
WO2002096359A2
WO2002096359A2 PCT/US2002/016739 US0216739W WO02096359A2 WO 2002096359 A2 WO2002096359 A2 WO 2002096359A2 US 0216739 W US0216739 W US 0216739W WO 02096359 A2 WO02096359 A2 WO 02096359A2
Authority
WO
WIPO (PCT)
Prior art keywords
htv
mmol
solution
compound
pharmaceutically acceptable
Prior art date
Application number
PCT/US2002/016739
Other languages
French (fr)
Other versions
WO2002096359A3 (en
Inventor
James R. Tata
Subharekha Raghavan
Zhijian Lu
Fengqi Zhang
Yuan Cheng
Jiang Chang
Ronald M. Kim
Joann M. Bohn
Thomas Rano
Dong-Ming Shen
Min Shu
Original Assignee
Merck & Co., Inc.
Huening, Tracy, T.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck & Co., Inc., Huening, Tracy, T. filed Critical Merck & Co., Inc.
Priority to AU2002305720A priority Critical patent/AU2002305720A1/en
Publication of WO2002096359A2 publication Critical patent/WO2002096359A2/en
Publication of WO2002096359A3 publication Critical patent/WO2002096359A3/en

Links

Classifications

    • 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
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • the present invention is directed to a class of N-[(3,4-dihydro-3- hydroxy-2H- 1 -benzopyran-4-yl] - ⁇ -hydroxy-4- [ [5-(halophenyl)-2-(f uranyl or oxazolyl)] alkyl] - -(arylmethyl or heteroarylmethyl)-2-[[(2,2,2- trifluoroethyl)amino]carbonyl]-l-piperazinepentanamides which are ⁇ protease inhibitors.
  • the present invention is also directed to pharmaceutical compositions containing these protease inhibitors and methods of using the protease inhibitors.
  • the HTV retrovirus is the causative agent for AIDS.
  • the HIV-1 retrovirus primarily uses the CD4 receptor (a 58 kDa transmembrane protein) to gain entry into cells, through high- affinity interactions between the viral envelope glycoprotein (gp 120) and a specific region of the CD4 molecule found in
  • HTV infection is characterized by an asymptomatic period immediately following infection that is devoid of clinical manifestations in the patient. Progressive HTV-induced destruction of the immune system then leads to increased susceptibility to opportunistic infections, which eventually produces a syndrome called AJDS- related complex (ARC) characterized by symptoms such as persistent generalized lymphadenopathy, fever, and weight loss, followed itself by full blown AIDS.
  • AJDS- related complex ARC
  • RNA-dependent DNA polymerase activity which catalyzes the synthesis of the minus strand DNA from viral RNA
  • RNase H ribonuclease H
  • DNA-dependent DNA polymerase activity which catalyzes the synthesis of a second DNA strand from the minus strand DNA template
  • the viral genome in the form of DNA is integrated into host genomic DNA and serves as a template for viral gene expression by the host transcription system, which leads eventually to virus replication (Sakai, H al., J. Virol. 1993, 67: 1169-1174).
  • the preintegration complex consists of integrase, reverse transcriptase, pl7 and proviral DNA (Bukrinsky et al., Proc. Nat. Acad. Sci. USA 1992, 89: 6580-6584).
  • the phosphorylated pl7 protein plays a key role in targeting the preintegration complex into the nucleus of host cell (Gallay et al., Cell 1995, 80:, 379-388).
  • HTV encodes the production of a protease which carries out post-translational cleavage of precursor polypeptides in a process necessary for the formation of infectious virions
  • These gene products include pol — which encodes the virion RNA-dependent DNA polymerase (reverse transcriptase), an endonuclease, and HTV protease — and gag — which encodes the core-proteins of the virion.
  • HTV HTV cellular fusion
  • reverse transcriptase inhibitors e.g., didanosine, zidovudine (AZT), and efavirenz
  • integrase inhibitors e.g., didanosine, zidovudine (AZT), and efavirenz
  • protease inhibitors inhibit the formation of infectious virions by interfering with the processing of viral polyprotein precursors.
  • HTV protease inhibitors are presently in clinical use for the treatment of AIDS and HTV infection, including indinavir (see US 5413999), nelfinavir (US 5484926), saquinavir (US 5196438), and ritonavir (US 5484801).
  • indinavir see US 5413999
  • nelfinavir US 5484926
  • saquinavir US 51964308
  • ritonavir US 5484801
  • Each of these protease inhibitors is a peptidomimetic, competitive inhibitor of the viral protease which prevents cleavage of the HTV gag-pol polyprotein precursor.
  • Indinavir for example, has been found to be highly effective in reducing HTV viral loads and increasing CD4 cell counts in HTV-infected patients, when used in combination with nucleoside reverse transcriptase inhibitors. See, for example, Hammer et al, New England J. Med. 1997, 337: 725-733 and Gulick et al., New England J. Med. 1997, 337: 734-739.
  • a substantial and persistent problem in the treatment of AIDS has been the ability of the HTV virus to develop resistance to the therapeutic agents employed to treat the disease.
  • Resistance to HTV-1 protease inhibitors has been associated with 25 or more amino acid substitutions in both the protease and the cleavage sites.
  • the present invention provides a novel class of N-[(3,4-dihydro-3- hydroxy-2H-l-benzopyran-4-yl]- ⁇ -hydroxy-4-[[5-(halophenyl)-2-(furanyl or oxazolyl)] alkyl] - ⁇ -(arylmethyl or heteroarylmethyl)-2-[[(2,2,2- trifluoroethyl)amino]carbonyl]-l-piperazinepentanamides which are potent inhibitors of ⁇ TV protease including mutant forms thereof that are resistant to known protease inhibitors.
  • a feature of the piperazinepentanamides in this class is that they exhibit little or no inhibition of either or both the CYP 2D6 and 2C9 enzymes relative to their inhibition of CYP 3A4.
  • These compounds are useful in the inhibition of ⁇ TV protease, the prevention of infection by ⁇ TV, the treatment of infection by ⁇ TV and in the treatment of AIDS and/or ARC, when employed as compounds or pharmaceutically acceptable salts or hydrates (when appropriate) thereof, optionally as pharmaceutical composition ingredients, and optionally in combination with other antivirals, anti-infectives, immunomodulators, antibiotics or vaccines. More particularly, the present invention includes a compound of Formula (I):
  • A is CH or N
  • R2 and R3 are each independently -H or methyl
  • X is O or S
  • Y is pyridyl which is optionally substituted with from 1 to 3 substituents each of which is independently halogen, cyano, C ⁇ -C6 alkyl, or C1-C6 alkoxy;
  • each Z is independently hydrogen, halogen, cyano, C1-C6 alkyl, or C1-C6 alkoxy;
  • q is an integer from zero to 2; and with the proviso that when A is N and R4 is:
  • the present invention also includes pharmaceutical compositions containing a compound of the present invention and methods of preparing such pharmaceutical compositions.
  • the present invention further includes processes for preparing the compounds of the invention, and also includes methods of inhibiting HIV protease, delaying the onset of AIDS, treating AIDS, preventing infection by HTV, and treating infection by HTV.
  • the present invention includes the compounds of Formula (I) above. These compounds and their pharmaceutically acceptable salts are HTV protease inhibitors.
  • each of the variables is as originally defined above; or a pharmaceutically acceptable salt thereof.
  • a second embodiment of the invention is a compound of Formula (I), wherein
  • Rl is -F or -Cl
  • R2 and R are either both -H or both methyl
  • Y is pyridyl which is optionally substituted with from 1 to 3 substituents each of which is independently halogen, cyano, C1-C4 alkyl, or C1-C4 alkoxy;
  • An aspect of the second embodiment is a compound of Formula (I), wherein
  • R2 and R3 are either both -H or both methyl
  • Q is -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy;
  • a third embodiment of the present invention is a compound of Formula (I), wherein
  • A is CH or N
  • Rl is -F or -Cl
  • R2 and R are either both -H or both methyl
  • X is S or O
  • each Z is independently -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy;
  • q is an integer from zero to 2;
  • An aspect of the third embodiment is a compound of Formula (I), wherein
  • A is CH or N
  • Rl is -F or -Cl
  • R2 and R3 are either both -H or both methyl
  • Z is -H, C1-C4 alkyl, or C1-C4 alkoxy
  • a fourth embodiment of the present invention is a compound of Formula (I), wherein
  • A is CH or N
  • Rl is -F or -Cl
  • R2 and R3 are either both -H or both methyl
  • each Z is independently -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy;
  • q is an integer from zero to 2; and with the proviso that when A is N and R4 is:
  • An aspect of the fourth embodiment is a compound of Formula (I), wherein
  • A is CH or N
  • Rl is -F or -Cl
  • R2 and R3 are either both -H or both methyl
  • R2 and R3 are both -H;
  • a fifth embodiment of the present invention is a compound of Formula (I), wherein
  • A is CH or N
  • Rl is -F or -Cl;
  • R2 and R3 are either both -H or both methyl;
  • each Z is independently -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy;
  • q is an integer from zero to 2;
  • An aspect of the fifth embodiment is a compound of Formula (I), wherein
  • A is CH or N
  • Rl is -F or -Cl
  • R and R3 are either both -H or both methyl
  • Z is -H, C 1-C4 alkyl, or C1-C4 alkoxy
  • Additional embodiments and aspects of the present invention include a compound of Formula (II) whose variables are as defined in any one of the second, third, fourth, and fifth embodiments set forth above, or in any one of the aspects thereof as set forth above.
  • Additional embodiments and aspects of the present invention include a compound of Formula (I) or of Formula (II), in which A is CH and all other variables are as originally defined above or as defined in any one of the foregoing embodiments and aspects. Still other embodiments and aspects of the present invention include a compound of Formula (I) or of Formula (II), in which A is N and all other variables are as originally defined above or as defined in any one of the foregoing embodiments and aspects.
  • Exemplifying the invention are compounds selected from the group consisting of
  • compositions comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition which comprises the product prepared (i.e., made) by combining (e.g., mixing) a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier.
  • AIDS treatment agent selected from the group consisting of AIDS antiviral agents, immunomodulators, and anti-infective agents.
  • at least one antiviral agent selected from the group consisting of non-nucleoside HTV reverse transcriptase inhibitors, nucleoside HTV reverse transcriptase inhibitors, HTV integrase inhibitors, and CCR5 receptor antagonists.
  • composition further comprises a therapeutically effective amount of at least one antiviral agent which is a CCR5 receptor antagonist.
  • a cytochrome P450 monooxygenase inhibitor e.g., ritonavir or a pharmaceutically acceptable salt thereof
  • a combination which comprises a therapeutically effective amount of a compound of Formula (I) and a therapeutically effective amount of an HTV infection/ AIDS treatment agent selected from the group consisting of HTV/ AIDS antiviral agents, immunomodulators, and anti-infective agents.
  • (k) The combination of (j), wherein the HTV infection/ AIDS treatment agent is an antiviral selected from the group consisting of non-nucleoside HTV reverse transcriptase inhibitors, nucleoside HTV reverse transcriptase inhibitors, HTV integrase inhibitors, and CCR5 receptor antagonists.
  • the HTV infection/ AIDS treatment agent is an antiviral selected from the group consisting of non-nucleoside HTV reverse transcriptase inhibitors, nucleoside HTV reverse transcriptase inhibitors, HTV integrase inhibitors, and CCR5 receptor antagonists.
  • a method of inhibiting HTV protease in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of a compound of Formula (I).
  • (m) A method of preventing or treating infection by HTV in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of a compound of Formula (I).
  • Additional embodiments of the invention include the pharmaceutical compositions, combinations and methods set forth in (a)-(t) above, wherein the compound employed therein is a compound of one of the embodiments or aspects of the compounds described above. It is to be understood that, unless expressly stated to the contrary, the compound of the present invention can be employed in the above- described pharmaceutical compositions, combinations, and methods in its free form or as a pharmaceutically acceptable salt.
  • C1-C6 alkyl refers to a linear or branched chain alkyl group having from 1 to 6 carbon atoms, and is selected from the hexyl alkyl and pentyl alkyl isomers, n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl.
  • C1-C4 alkyl refers to a linear or branched chain alkyl group having from 1 to 4 carbon atoms, and is selected from n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl.
  • C ⁇ -C6 alkoxy means an -O-alkyl group wherein alkyl is Cl to C6 alkyl as defined above.
  • C1-C4 alkoxy has an analogous meaning; i.e., it is an alkoxy group selected from methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, and sec-butoxy.
  • C1-C3 alkoxy is selected from methoxy, ethoxy, n-propoxy, and isopropoxy.
  • aryl refers to aromatic mono- and poly-carbocyclic ring systems, wherein the carbocyclic rings in the polyring systems may be fused or attached to each other via a single ring carbon.
  • exemplary aryl groups are phenyl and naphthyl.
  • heteroaryl refers to (i) a 5- or 6-membered aromatic ring consisting of carbon atoms and from 1 to 3 heteroatoms selected from N, S, and O or (ii) an 8- to 10-membered bicyclic ring system consisting of carbon atoms and from 1 to 3 heteroatoms selected from N, S, and O, wherein at least one of the rings in the bicyclic system is an aromatic ring.
  • substituted refers to mono- and poly-substitution by a named substituent to the extent such single and multiple substitution is chemically allowed and results in a chemically stable compound.
  • the compounds of the present invention are potent inhibitors of HTV protease including mutant forms thereof that are resistant to known protease inhibitors, as may be seen by reference to Examples 12 - 14 below.
  • the compounds of the instant invention are significantly less potent inhibitors of CYP 2D6 and CYP 2C9 than structurally related piperazinepentanamide protease inhibitors. Accordingly, the compounds of the invention can be expected to avoid and/or have fewer drug-drug interactions compared to their structurally related counterparts. Inhibition of CYP 2D6 and 2C9 can be determined by many methods such as those detailed in Chiba et al., Drug metabolism and Disposition 1997, 25, No. 9: 1022-1031.
  • the present invention includes pharmaceutical compositions useful for inhibiting HTV protease, comprising an effective amount of a compound of this invention, and a pharmaceutically acceptable carrier.
  • Pharmaceutical compositions useful for preventing or treating infection by HTV, for delaying the onset of AIDS, or for treating AIDS are also encompassed by the present invention, as well as a method of inhibiting HTV protease, and a method of preventing or treating infection by HTV, of delaying the onset of AIDS, or of treating AIDS.
  • An aspect of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention in combination with a therapeutically effective amount of an agent useful for treating HTV infection and/or AIDS (alternatively referred to as an HTV/ AIDS treatment agent) selected from: (1) an HTV/ATDS antiviral agent,
  • the present invention also includes the use of a compound of the present invention as described above as a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, or (c) treating AIDS.
  • the present invention further includes the use of a compound of the present invention as described above in the preparation of a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, (c) delaying the onset of AIDS, or (d) treating AIDS.
  • the present invention also includes the use of any of the HTV protease inhibiting compounds of the present invention as described above in combination with one or more HTV/ATDS treatment agents selected from an HTV/ATDS antiviral agent, an anti-infective agent, and an immunomodulator for use as a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, (c) delaying the onset of AIDS, or (d) treating AIDS, said medicament comprising an effective amount of the HTV protease inhibitor compound and an effective amount of the one or more treatment agents.
  • HTV/ATDS treatment agents selected from an HTV/ATDS antiviral agent, an anti-infective agent, and an immunomodulator for use as a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, (c) delaying the onset of AIDS, or (d) treating AIDS, said medicament comprising an effective amount of the HTV protease inhibitor compound and an effective amount of
  • the present invention further includes the use of any of the HTV protease inhibiting compounds of the present invention as described above in combination with one or more HTV/ AIDS treatment agents selected from an HTV/ATDS antiviral agent, an anti-infective agent, and an immunomodulator for the manufacture of a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, (c) delaying the onset of ADDS, or (d) treating ADDS, said medicament comprising an effective amount of the HTV protease inhibitor compound and an effective amount of the one or more treatment agents.
  • HTV/ATDS antiviral agent selected from an HTV/ATDS antiviral agent, an anti-infective agent, and an immunomodulator for the manufacture of a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, (c) delaying the onset of ADDS, or (d) treating ADDS, said medicament comprising an effective amount of the HTV protease inhibitor compound and
  • the present invention also includes a compound of the present invention for use in (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, or (c) preventing, treating or delaying the onset of ADDS.
  • the compound of the present invention can optionally be employed in combination with one or more HTV/ATDS treatment agents selected from HTV/ATDS antiviral agents, anti-infective agents, and immunomodulators.
  • the compounds of the present invention have asymmetric centers and can have additional centers (e.g., where one of R and R3 is -H and the other is methyl), in which event the compound of the present invention may occur as a mixture of stereoisomers or as individual diastereomers, with all isomeric forms being included in the present invention.
  • a therapeutically effective amount of the compounds of the present invention are useful in the inhibition of HTV protease, the prevention or treatment of infection by HTV and the delay in the onset of or treatment of consequent pathological conditions such as ADDS.
  • Treating or delaying the onset of ADDS or preventing or treating infection by HTV is defined as including, but not limited to, treating a wide range of states of HTV infection: ADDS, ARC, both symptomatic and asymptomatic, and actual or potential exposure to HTV.
  • the compounds of this invention are useful in treating infection by HTV after suspected past exposure to HTV by e.g., blood transfusion, exchange of body fluids, bites, accidental needle stick, or exposure to patient blood during surgery.
  • the compounds of the invention can also be used in "salvage" therapy; i.e., the compounds can be used to treat HTV infection, ADDS, or ARC in HIV-positive subjects whose viral load achieved undetectable levels via conventional therapies employing known protease inhibitors, and then rebounded due to the emergence of HTV mutants resistant to the known inhibitors.
  • the compounds of this invention are useful in the preparation and execution of screening assays for antiviral compounds.
  • the compounds of this invention are useful for isolating enzyme mutants, which are excellent screening tools for more powerful antiviral compounds.
  • the compounds of this invention are useful in establishing or determining the binding site of other antivirals to HTV protease, e.g., by competitive inhibition.
  • the compounds of this invention are commercial products to be sold for these purposes.
  • the present invention also provides for the use of a compound of structural formula (I) to make a pharmaceutical composition useful for inhibiting HTV protease, treating or preventing HTV infection, delaying the onset of ADDS, and for treating ADDS.
  • the compounds of the present invention may be administered in the form of pharmaceutically acceptable salts.
  • pharmaceutically acceptable salt refers to all acceptable salts of the compounds of Formula (I) (in the form of water- or oil-soluble or dispersible products) and includes the conventional non-toxic salts or the quaternary ammonium salts which are formed, e.g., from inorganic or organic acids or bases.
  • the salt can be used as a dosage form for modifying the solubility or hydrolysis characteristics of the compound or can be used in sustained release or pro-drug formulations.
  • the compounds of the present invention may be administered orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, in dosage unit formulations containing conventional non-toxic pharmaceutically- acceptable carriers, adjuvants and vehicles.
  • administration and variants thereof (e.g., “administering” a compound) in reference to a compound of the invention each mean providing the compound or a prodrug of the compound to the individual in need of treatment.
  • a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., ADDS antivirals)
  • administration and its variants are each understood to include concunent and sequential provision of the compound or prodrug thereof and other agents.
  • the present invention further provides for preventing or treating HTV infection and for delaying the onset of or treating ADDS by administering to a subject in need of such treatment a pharmaceutical composition comprising a pharmaceutical carrier and a therapeutically effective amount of a compound of the present invention.
  • 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.
  • pharmaceutically acceptable means that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • subject refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • terapéuticaally effective amount means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease being treated.
  • compositions may be in the form of orally- administrable suspensions or tablets or capsules, nasal sprays, sterile injectible preparations, for example, as sterile injectible aqueous or oleagenous suspensions or suppositories.
  • these compositions When administered orally as a suspension, these compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may contain microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and 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.
  • compositions When administered by nasal aerosol or inhalation, these compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • the injectible 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.
  • compositions When rectally administered in the form of suppositories, these compositions may be prepared by mixing the drug with a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters of polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • a suitable non-irritating excipient such as cocoa butter, synthetic glyceride esters of polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • the compounds of this invention can be administered orally to humans in a dosage range of 0.01 to 1000 mg/kg body weight per day in a single dose or in divided doses.
  • One preferred dosage range is 0.1 to 200 mg/kg body weight per day orally in a single dose or in divided doses.
  • Another preferred dosage range is 0.5 to 100 mg/kg body weight per day orally in single or divided doses.
  • the compositions are preferably provided in the form of tablets containing 1 to 1000 milligrams of the active ingredient, particularly 1, 5, 10, 15. 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • the present invention is also directed to combinations of the HTV protease inhibitor compounds with one or more agents useful in the treatment of HTV infection and/or ADDS.
  • the compounds of this invention may be effectively administered, whether at periods of pre-exposure and/or post-exposure, in combination with effective amounts of the HIV/ ADDS antivirals, imunomodulators, antiinfectives, or vaccines, such as those in Table 1 as follows:
  • Cidofovir Gilead Science CMV retinitis, herpes, papillomavirus
  • mozenavir AVDD (Camden, NJ) HTV infection, ADDS, ARC (DMP-450) (protease inhibitor)
  • GS 840 Gilead HTV infection ADDS, ARC (reverse transcriptase inhibitor) HBY097 Hoechst Marion Roussel HTV infection, ADDS, ARC (non-nucleoside reverse transcriptase inhibitor)
  • ISIS 2922 ISIS Pharmaceuticals CMV retinitis KNI-272 Nat'l Cancer Institute HTV-assoc. diseases Lamivudine, 3TC Glaxo Wellcome HTV infection, ADDS,
  • Virazole Viratek/ICN (Costa asymptomatic HTV positive, Ribavirin Mesa, CA) LAS, ARC
  • Lopinavir (ABT-378) Abbott HTV infection, ADDS, ARC (protease inhibitor)
  • Lopinavir + ritonavir Abbott (KALETRA®) HTV infection, ADDS, ARC (ABT-378/r); Kaletra (protease inhibitor)
  • PRO 542 Progenies HIV infection, ADDS, ARC (attachment inhibitor)
  • PRO 140 Progenies HTV infection, ADDS, ARC (CCR5 co-receptor inhibitor) TAK-779 Takeda HTV infection, ADDS, ARC (injectable CCR5 receptor antagonist)
  • DPC 681 & DPC 684 DuPont HTV infection, ADDS, ARC (protease inhibitors)
  • DPC 961 & DPC 083 DuPont HTV infection ADDS, ARC (nonnucleoside reverse transcriptase inhibitors) abacavir + lamivudine + GlaxoSmithKline HTV infection, ADDS, ARC zidovudine (TRIZTVTR®) (reverse transcriptase inhibitors) tipranavir (PNU-140690) Boehringer Ingelheim HTV infection, ADDS, ARC (protease inhibitor) tenofovir Gilead (VTREAD®) HTV infection, ADDS, ARC (nucleotide reverse transcriptase inhibitor)
  • TMC-120 & TMC-125 Tibotec HTV infections ADDS, ARC (non-nucleoside reverse transcriptase inhibitors)
  • Interleukin-2 (aldeslukin) counts
  • Pentamidine Isethionate LyphoMed PCP treatment (TM & TV) (Rosemont, TL)
  • HTV/ATDS antivirals antivirals
  • immunomodulators anti-infectives or vaccines
  • any pharmaceutical composition useful for the treatment of HTV infection and/or ADDS are typically employed in their conventional dosage ranges and regimens as reported in the art, including the dosages described in the Physicians' Desk Reference, 54 th edition, Medical Economics Company, 2000.
  • the dosage ranges for a compound of the invention in these combinations are the same as those set forth above just before Table 1.
  • One suitable combination is a compound of the present mvention and a nucleoside inhibitor of HTV reverse transcriptase such as AZT, 3TC, ddC, or ddl.
  • Another suitable combination is a compound of the present invention and a non- nucleoside inhibitor of HTV reverse transcriptase, such as efavirenz, and optionally a nucleoside inhibitor of HTV reverse transcriptase, such as AZT, 3TC, ddC or ddl.
  • Still another suitable combination is any one of the combinations in the preceding paragraph, further comprising an additional HTV protease inhibitor such as indinavir, Compound A, nelfinavir, ritonavir, saquinavir, amprenavir, or abacavir.
  • An aspect of this combination is the combination wherein the additional inhibitor of HTV protease is the sulfate salt of indinavir.
  • Another aspect of this combination is the combination in which the additional protease inhibitor is selected from nelfinavir and ritonavir.
  • Still another aspect of this combination is the combination in which the additional inhibitor of HTV protease is saquinavir, which is typically administered in a dosage of 600 or 1200 mg tid.
  • a compound of the present invention with the following (1) efavirenz, optionally with AZT and/or 3TC and/or ddl and/or ddC, and optionally with indinavir; (2) any of AZT and or ddl and/or ddC and/or 3TC, and optionally with indinavir; (3) d4T and 3TC and/or AZT; (4) AZT and 3TC; and (5) AZT and d4T.
  • Another aspect of the present invention is co-administration of a compound of the present invention with an inhibitor of cytochrome P450 monooxygenase in an amount effective to improve the pharmacokinetics of the compound.
  • Compounds of the invention can be metabolized, at least in part, by cytochrome P450 (CYP3A4).
  • Co-administration of compounds of the invention with a cytcochrome P450 inhibitor can improve the pharmacokinetic profile of the compound in subjects (e.g., humans); i.e., co-administration can increase C a ⁇ (the maximum plasma concentration of the compound), AUC (area under the curve of plasma concentration of the compound versus time), and/or the half -life of the compound.
  • a suitable P450 inhibitor is ritonavir.
  • ritonavir in this circumstance is as a pharmacokinetic modulator and not as a protease inhibitor; i.e., an amount of ritonavir which is effective for improving the pharmacokinetics of the compound can provide a secondary or even negligible contribution to the antiviral effect.
  • a compound of the present invention can also be administered in combination with an HTV integrase inhibitor such as a compound described in WO 99/62520, WO 99/62513, or WO 99/62897.
  • a compound of the present invention can also be administered in combination with a CCR5 receptor antagonist, such as a compound described in WO 00/59502, WO 00/59503, WO 00/59497, WO 00/59498, WO 00/76511, WO 00/76512, WO 00/76513, WO 00/76514, WO 00/76972, or WO 00/76793.
  • the compound of the present invention and other active agents may be administered together or separately.
  • the administration of one agent may be prior to, concunent with, or subsequent to the administration of other agent(s).
  • Efavirenz is (-)-6-chloro-4-cyclopropylethynyl-4-trifluoromethyl-l ,4- dihydro-2H-3,l-benzoxazin-2-one, also known as DMP-266 or SUSTTVA® (DuPont) or STOCRTN® (Merck).
  • DMP-266 or SUSTTVA® (DuPont) or STOCRTN® (Merck).
  • Efavirenz and its utility as an HTV reverse transcriptase inhibitor is described in US 5519021 and in the corresponding PCT published application, WO 95/20389. Efavirenz can be synthesized by the protocol of US 5633405.
  • AZT is 3'-azido-3'-deoxythymidine, is also known as zidovudine, and is available from Burroughs-Wellcome under the tradename RETROVTR®.
  • Stavudine is 2',3'-didehydro-3'-deoxythymidine, is also known as 2',3'-dihydro-3'- deoxythymidine and d4T, and is available from Bristol-Myers Squibb under the tradename ZERIT®.
  • 3TC is (2R-cis)-4-amino-l-[2-(hydroxymethyl)-l,3-oxathiolan- 5-yl]-2(lH)-pyrimidinone, is also known as (-)-l-[(2R,5S)-2-(hydroxymethyl)-l,3- oxathiolan-5-yl]cytosine and lamivudine, and is available from Glaxo Wellcome under the tradename EPTVTR®.
  • ddC is 2',3'-dideoxycytidine, is also known as zalcitabine, and is available from Hoffman LaRoche under the tradename H ⁇ VDD®.
  • ddl is 2',3 '-dideoxyinosine, is also known as didanosine, and is available from Bristol- Myers-Squibb under the tradename VDDEX®.
  • VDDEX® The preparation of ddC, ddl and AZT are also described in EPO 0,484,071.
  • Indinavir is 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, and can be prepared as described in US 5413999.
  • Indinavir is generally administered as the sulfate salt at a dosage of 800 mg three times a day.
  • Indinavir sulfate is available from Merck under the tradename CRTXTVAN®.
  • Compound A is N-(2(R)-hydroxy-l(S)-indanyl)-2(R)-phenylmethyl- 4(S)-hydroxy-5-(l-(4-(2-benzo[b]furanylmethyl)-2(S)-N'-(t-butylcarboxamido)- piperazinyl))pentaneamide, preferably administered as the sulfate salt.
  • Compound A can be prepared as described in US 5646148.
  • Ritonavir is [5S-(5R*,8R*,10R*, HR*)]-10-hydroxy-2-methyl-5-(l- methylethyl)-l -[2-( 1 -methylethyl)-4-thiazolyl]-3 ,6-dioxo-8 , 11 -bis(phenylmethyl)-2, 4, 7, 12-tetraazatridecan-13-oic acid 5-thiazolylmethyl ester, also known as 5- thiazolylmethyl [(aS)-a-[(lS,3S)-l-hydroxy-3-[(2S)-2-[3-[(2-isopropyl-4- thiazolyl)methyl]-3-methylureido]-3-methylbutyramido]-4- phenylbutyl]phenethyl]carbamate.
  • Nelfinavir is [3S-[2(2S*,3S*),3a,4ab,8ab]]-N-(l,l- dimethylethyl)decahydro-2- [2-hydroxy-3 - [(3 -hydroxy-2-methylbenzoyl)amino] -4- (phenylthio)butyl]-3-isoquinolinecarboxamide, also known as (3S,4aS,8aS)-N-tert- Butyl-2-[(2R,3R)-3-(3,2-crestoamido)-2-hydroxy-4-(phenylthio)butyl]decahydro-3- isoquinolinecarboxamide.
  • VERACEPT® the monomethanesulfonate salt of nelfinavir (nelfinavir mesylate) is commerically available from Agouron. Nelfinavir
  • Saquinavir is N-tert-butyl-decahydro-2-[2(R)-hydroxy-4-phenyl- 3(S)-[[N-(2-quinolylcarbonyl)-L-asparaginyl]amino]butyl]-(4aS,8aS)- isoquinoline-3(S)-carboxamide.
  • Saquinavir can be prepared in accordance with procedures disclosed in US 5196438.
  • TNVTRASE® (saquinavir mesylate) is available from Roche Laboratories.
  • Amprenavir is 4-amino-N-((2 syn,3S)-2-hydroxy-4-phenyl-3-((S)- tetrahydrofuran-3-yloxycarbonylamino)-butyl)-N-isobutyl-benzenesulfonamide, also known as Compound 168 and 141 W94.
  • Amprenavir is an aspartyl protease inhibitor that can be prepared by following the procedures described in
  • Amprenavir is available under the tradename AGENERASE® from Glaxo Wellcome. Amprenavir can be prepared as described in US 5783701.
  • Abacavir is (lS,4R)-cis-4-[2-amino-6-(cyclopropylamino)-9H- purin-9-yl]-2-cyclopentene-l-methanol, also known as 1592U89.
  • Abacavir can be prepared by following the protocol of EP 0434450.
  • ACN acetonitrile
  • AcOH acetic acid
  • ADDS acquired immune deficiency syndrome
  • Alloc allyloxycarbonyl
  • CBZ carbobenzoxy (alternatively, benzyloxycarbonyl)
  • DTEA diisopropylethylamine
  • EDC l-ethyl-3-(3-dimethylaminopropyl) carbodiimide
  • HOBT 1-hydroxy benzotriazole hydrate
  • Ph phenyl
  • TB AF tetrabutylammonium fluoride
  • TBSOTf t-butyldimethylsilyl triflate
  • TEA triethylamine
  • TFA trifluoroacetic acid
  • TFEA trifluoroethylamine
  • Tf2 ⁇ triflic anhydride
  • THF tetrahydrofuran
  • TLC thin layer chromatgraphy
  • TMSCN trimethylsilyl cyanide
  • TsOH p-toluenesulfonic acid
  • the compounds of the present invention can be readily prepared according to the following reaction schemes and examples, or modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants which are themselves known to those of ordinary skill in this art, but are not mentioned in greater detail. Furthermore, other methods for preparing compounds of the invention will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples. Unless otherwise indicated, all variables are as defined above.
  • a compound of Formula (I) can be prepared in accordance with Scheme 1, wherein Compound I is readily prepared via literature procedures described in Dorsey et al., J. Med. Chem. 1994, 37: 3443-3451, and also in US 5413999.
  • Treatment of the hydroxyl compound 1 with triflic anhydride and lutidine in an inert solvent such as dichloromethane provides triflate 2.
  • Displacement of the triflate with piperazine 3 occurs on heating in an inert solvent such as isopropanol to give lactone 4.
  • Amide coupling of compound 5 with aminochromanol to obtain 6 is typically performed by the carbodiimide method with reagents such as l-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) and HOBT in an inert solvent such as dichloromethane.
  • reagents such as l-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) and HOBT in an inert solvent such as dichloromethane.
  • EDC l-ethyl-3-(3-dimethylaminopropyl) carbodiimide
  • HOBT inert solvent such as dichloromethane.
  • Other methods of forming the amide or peptide bond include, but are not limited to, the synthetic routes via an acid chloride, azide, mixed anhydride or activated ester.
  • the silyl protecting group is removed with fluoride to arrive at compound 7.
  • the BOC protecting group on the amine is then removed with a strong acid such as trifluoroacetic acid or hydrochloric acid in an alcoholic solvent such as methanol to give the penultimate intermediate 8.
  • Penultimate 8 is then reacted with the desired aldehyde 9 and a reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride in an inert solvent such as dichloromethane to give compound 10.
  • the epoxide opening can be carried out by heating piperazine 3 and the epoxide in an inert solvent. Acidic removal of the protecting groups can be accomplished by treatment with hydrochloric acid in an alcoholic solvent such as methanol, ethanol or isopropanol. The resulting intermediate 18 is then reductively aminated as in Scheme 1 to provide the compounds of the present invention.
  • the epoxide opening can be preformed with fully elaborated piperazine 14 to give 20. Once again the protecting group is removed with strong acid to give 19.
  • Piperazine intermediates are readily prepared from the known piperazine carboxylic acid 21, which can be prepared as described in He/. Chem. Acta. 1960, 43: 888-896. Selective monoprotection of the piperazine is carried out using BOC anhydride as described in Tetrahedron Letters 1989, 30: 5193-5196. The remaining unprotected amine can then be protected with any number of chloroformates including allyl chloroformate or benzyl chloroformate to give 23. Amide couplings of 23 with trifluoroethylamine to give 24 are performed using standard amide coupling reactions as described above. Acidic removal of the BOC protecting group as before gives 25. The Alloc group can be removed as before.
  • the CBZ group is readily removed by hydrogenolysis with a palladium catalyst under a hydrogen atmosphere in an alcoholic solvent such as methanol or ethanol. Removal of the protecting groups can also be accomplished by a number of methods known in the art, such as those described in Greene, Protective Groups in Organic Synthesis, John Wiley and Sons, New York, 1991. These deprotected intermediates are then carried onto compounds of the instant invention via the synthetic routes shown in Schemes 1, 2 and 3.
  • aldehyde intermediates are, in many cases, commercially available (e.g., Aldrich Chemical).
  • Other aldehydes of interest can be prepared by literature methods including classical methods familiar to those skilled in the art. Stille and Suzuki coupling of commercially and readily available aryl and heteroaryl halides, aryl trialkylstannanes, and arylboronic acids also provides the desired aldehydes as exemplified for bromofuran in Scheme 5 below.
  • Aldehyde 27 can be reacted with trialkylarylstannane 26 in the presence of a palladium catalyst by the method of Gronowitz et al. , J. Heterocyclic Chem. 1995, 35: 771, to give 28.
  • trialkylstannane 30 can be coupled with arylhalides such as 29 to give 31 which can be deprotected under mild conditions with dilute hydrochloric acid to give aldehyde 28.
  • arylhalides such as 29
  • Other aldehydes are available via metal halogen exchange followed by anion quenching with DMF as described by Vogel et al., J. Chem. Soc. Perkin Trans I, 1974, 37.
  • Metalation of a biaryl or heterobiaryl compound such as 32 with a strong base such as n-butyllithium at low temperature in an inert solvent such as THF followed by anion trapping with DMF also provides aldehydes such as 28.
  • Oxazolyl piperazine intermediates such as 40 are available via the route shown in Scheme 7 below. Alkylation of piperazine 25 with bromo acid 36 in the presence of silver triflate in an inert solvent such as THF, according to methods detailed in J. Org. Chem. 1995, 60: 4013-4016, provides 37. Amide coupling of amine 38 to acid 37 to provide 39 can be carried out by any of the methods described above including the EDC /HOBT method. Amines such as 38 are prepared via chemistry described in Org. Synth. 1986, 64: 19-26 and Tetrahedron Letters 1999, 40: 6739-6743.
  • Oxazole formation is accomplished by the action of a strong acid such as sulfuric acid on 39 in an inert solvent at elevated temperature, or as described in J. Med. Chem. 1996, 39: 2753-2763, to give intermediate 40.
  • a strong acid such as sulfuric acid
  • intermediate 40 can be transformed into compounds of the instant invention via synthetic routes shown in Schemes 1,2, and 3.
  • Oxazolyl piperazine intermediate 40 can also be obtained via the route shown in Scheme 8 below.
  • Treatment of piperazine amide 41 with acetone at an elevated temperature will afford the acetonide which can be treated with acid HL (wherein L" is a non-nucleophilic anion such as OTf-) to afford the acid salt 42.
  • Reaction of 42 with a dimethoxyalkane (e.g., 2,2-dimethoxypropane) will provide iminium salt 43, which can be coupled with a suitably metallated oxazole to provide coupled product 44.
  • a dimethoxyalkane e.g., 2,2-dimethoxypropane
  • Suitable metallated oxazoles include those obtained by metallating the oxaole with an alkyllithium (e.g., n-butyllithium) or an alkylmagnesium halide (e.g., isopropylmagnesium bromide) in an inert solvent (e.g., an ether such as THF).
  • Oxazolyl piperazine intermediate 40 can be obtained from 44 by treating 44 with acid (e.g., HCI, TFA, or an arylsulfonic acid).
  • MetaChem 21X150mm Polaris C18-A5 micron column The gradient used was 35% acetonitrile-water with 0.1% TFA to 60% acetonitrile-water. The product containing fractions were combined and concentrated in vacuo. The residue was dissolved in ethyl acetate and washed with saturated sodium carbonate solution to give the desired compound as a white solid.
  • the reaction mixture was stined at room temperature for 1.5 h, then it was poured into saturated NaHCO 3 solution and extracted with ethyl acetate three times. The combined organic layer was dried over Na ⁇ O,,, filtered and concentrated. The crude product was purified by flash column chromatography on silica gel with 4: 1 ethyl acetate/ hexane as eluant to give the title compound.
  • reaction mixture was poured into 20mL ice/brine and stined 30 minutes. This mixture was poured into CH 2 C1 2 and washed with 2x H O and 2x brine. After drying (MgSO 4 ), filtration, and removal of solvent in vacuo, the residue was purified by Biotage column chromatography (40M; 20% EtOAc/hexane) to provide the desired intermediate.
  • MeOH was hydrogenated at 1 atm using Pd/C as the catalyst. The mixture was stined for 2 h and filtered through Celite. The filtrate was concentrated via vacuum. The residue was dissolved in MeOH (5 mL). An aqueous of NaOH (0.036 g, 0.91 mmol) was added. The solution was stined at room temperature for 2 h. The solvents were removed. Toluene was added to the solid and removed (2 x 5 mL). 1 mL of IN HCI solution was added and removed. The above process was repeated with toluene (2 x).
  • NTS N-iodosuccinimide
  • the titled compound from Step J (0.04 g, 0.045 mmol) was mixed with 1 mL of MeO ⁇ and 2 mL of IM ⁇ C1 in ether. The solution was stined at room temperature for 2 h. The solvent was removed. The residue was mixed with saturated ⁇ a ⁇ CO 3 (2 mL) and extracted with EtOAc (3x20 mL). The combined EtOAc layers were washed with brine, and dried over sodium sulfate. The titled compound was obtained as a white solid after flash chromatography using EtO Ac/acetone (1:1) as the elute.
  • Step J To a solution of the intermediate obtained from Example 11, Step J (51.6 mg, 0.12 mmol) in 2-propanol (1.5 mL) was added the intermediate from
  • Example 1 Step N (55.5 mg, 0.12 mmol) and the resulting mixture refluxed overnight.
  • the reaction mixture was purified on RP HPLC using 30-60% MeCN gradient over 15 minutes at 6.0 mL per minute with 0.1%) TFA on a 9.4x250 mm Zorbax SB-C18 column. Pure product fractions were pooled and evaporated in vacuo to give the title compound.
  • LC-MS (El) 863.3 (M+l) and 885.3 (M+Na).
  • Synthetic oligonucleotide cassettes of 444 base pairs were designed according to the wild-type sequence of pET-3b-HTVPR. Point mutations were incorporated into the DNA with a bias toward optimal codon usage in E. Coli to yield amino acid mutations listed in Table 2 below.
  • the oligonucleotides were annealed and ligated into pUC-18 or pUC-19 by Midland Certified Reagent Company. The primary sequence was verified before subcloning into a pET-3b expression vector via Nde I and Bpull021 sites and reconfirmed by automated double-stranded DNA sequencing. Clones carrying the mutant DNA were transformed and expressed as previously described in Schock et al., 7. Biol. Chem.
  • the reaction is initiated by the addition of 28 ⁇ L of 14.3 picomolar (wild type, K-60, Q-60) and 28.6 pM (V-18) protease in a solution of 50mM Na acetate pH 5.5 and 0.1% bovine serum albumin.
  • the reaction was quenched with 120 ⁇ L of 10% phosphoric acid.
  • Products of the reaction were separated by HPLC (VYDAC wide pore 5 cm C-18 reverse phase, acetonitrile gradient, 0.1% phosphoric acid). The extent of inhibition of the reaction was determined from the peak heights of the products. HPLC of the products, independently synthesized, proved quantitation standards and confirmation of the product composition.
  • the compounds of the invention prepared in Examples 1-12 exhibited IC50 values ranging from about 0.05 to about 1 nM against the wild-type enzyme.
  • the indinavir IC50 value against the wild type enzyme is 0.6 nM (average).
  • the compounds of the invention prepared in Examples 1-12 exhibited IC50 values in the range of about 0.02 to about 5 nM against the mutant enzymes Q-60, K-60, and V-18. These IC50 values range from about 4-fold to greater than about 100-fold more potent than the values of of 20 to 50 nM obtained for indinavir against these same mutant enzymes.
  • HTV-1 p24 accumulation was directly conelated with virus spread.
  • the cell culture inhibitory concentration was defined as the inhibitor concentration in nanomoles/liter which reduced the spread of infection by at least 95%, or CIC95.
  • the compounds of the invention prepared in Examples 1-12 exhibited CIC95 values in the range of from less than about 8 to about 50 nM against the wild-type viral construct.
  • the CIC95 of indinavir against the wild-type viral construct is from 50 to 100 nM.
  • the compounds of the invention prepared in Examples 1-12 exhibited CIC95 values in the range of about 8 to about 125 nM against the viral constructs Q60, K-60, and V-18. These CIC95 values range from about 4-fold to more than about 100-fold more potent than the values of greater than 1000 nM obtained for indinavir against these same viral constructs.
  • HTV-infected MT-4 cell Suspension MT cells are infected at Day 0 at a concentration of 250,000 per ml with a 1:1000 dilution of HTV-1 strain Tub stock (final 125 pg p24/ml; sufficient to yield ⁇ 1% infected cells on day 1 and 25-100% on day 4).
  • Cells are infected and grown in the following medium: RPMI 1640 (Whittaker BioProducts), 10% inactivated fetal bovine serum, 4 mM glutamine (Gibco Labs) and 1:100 Penicillin- Streptomycin (Gibco Labs). The mixture is incubated overnight at 37°C in 5% CO2 atmosphere.
  • a matrix of nanomolar range concentrations of the pairwise combinations is prepared.
  • aliquots of 125 ml of inhibitors are added to equal volumes of HTV-infected MT-4 cells (50,000 per well) in a 96-well microtiter cell culture plate. Incubation is continued for 3 days at 37°C in 5% CO2 atmosphere.
  • the concentration of HTV p24 antigen is measured by an enzyme immunoassay, described as follows. Aliquots of p24 antigen to be measured are added to microwells coated with a monoclonal antibody specific for HTV core antigen. The microwells are washed at this point, and at other appropriate Steps that follow. Biotinylated HTV-specific antibody is then added, followed by conjugated streptavidin-horseradish peroxidase. A color reaction occurs from the added hydrogen peroxide and tetramethylbenzidine substrate. Color intensity is proportional to the concentration of HTV p24 antigen.
  • pairwise combinations of inhibitors are found to exhibit markedly enhanced inhibition of virus spread, in comparison to each inhibitor alone, or in comparison to merely additive inhibition of each inhibitor.
  • fractional inhibitory concentration ratios are calculated according to Elion, et al., J. Biol. Chem. 1954, 208: 477.
  • the minimum sum of FICs which is the maximum synergy, is determined for various pairwise combinations. The smaller the number, the greater the synergy.
  • reaction temperature was kept below 35 °C.
  • the reaction mixture (yellow/white slurry) was diluted with 10% K 2 CO 3 in water (24 L, 20 mL/g acid) and the reaction slurry was kept below 35 °C.
  • the wet cake was washed with deionized water (12 L, 10 mL/g acid) and dried under vacuum (22" Hg) at 40 °C with a nitrogen purge. Theoretical yield of 1816 g .
  • the pip amide ethanol filtrate (116.37 g containing 10.3 g of racemic pip amide by LC assay) was concentrated in vacuo to a final volume of 40.2 mL (3.9 mL per gram of pip amide) and the slurry is diluted with 82.4 mL (8 mL per gram pip amide) of acetonitrile (ACN) and stined until homogenous.
  • ACN acetonitrile
  • the water content of the two solutions was then determined by Karl Fisher titration.
  • the CSA solution was added to the pip amide solution giving a small exotherm to approx. 31-32 °C.
  • Water 11.02 mL, 1.118 mL per gram of pip amide minus the total water content of the two solutions
  • the acetonitrile:ethanol:water ratio was 26:2.9:1.1 (v/v/v).
  • Solids began to form after 15-30 min.
  • the solution/slurry was heated to 72 °C to completely dissolve all solids.
  • the yellow solution was recooled to 62 °C and seeded with a slurry of 10.3 mg of pip amide salt in 1 mL of acetonitrile. After a two hour age at 62 °C the slurry was allowed to cool to room temperature overnight

Abstract

Piperazine pentanamide compounds of formula (1): wherein A, R?1, R2, R3, and R4¿ are defined herein, are inhibitors of HIV protease and inhibitors of HIV replication. These compounds are useful in the prevention or treatment of infection by HIV and the treatment of AIDS, either as compounds, pharmaceutically acceptable salts, pharmaceutical composition ingredients, whether or not in combination with other antivirals, immunomodulators, antibiotics or vaccines. Methods of treating AIDS and methods of preventing or treating infection by HIV are also described. These compounds are effective against HIV viral mutants which are resistant to HIV protease inhibitors currently used for treating AIDS and HIV infection.

Description

TITLE OF THE INVENTION
PIPERAZENE PENTANA VHDE H PROTEASE INHIBITORS
This application claims the benefit of U.S. Provisonal Application No. 60/294,370, filed May 30, 2001 , the disclosure of which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
The present invention is directed to a class of N-[(3,4-dihydro-3- hydroxy-2H- 1 -benzopyran-4-yl] -γ-hydroxy-4- [ [5-(halophenyl)-2-(f uranyl or oxazolyl)] alkyl] - -(arylmethyl or heteroarylmethyl)-2-[[(2,2,2- trifluoroethyl)amino]carbonyl]-l-piperazinepentanamides which are Η protease inhibitors. The present invention is also directed to pharmaceutical compositions containing these protease inhibitors and methods of using the protease inhibitors.
BACKGROUND OF THE INVENTION
The HTV retrovirus is the causative agent for AIDS. The HIV-1 retrovirus primarily uses the CD4 receptor (a 58 kDa transmembrane protein) to gain entry into cells, through high- affinity interactions between the viral envelope glycoprotein (gp 120) and a specific region of the CD4 molecule found in
T-lymphocytes and CD4 (+) T-helper cells (Lasky L.A. et al., Cell 1987, 50: 975- 985). HTV infection is characterized by an asymptomatic period immediately following infection that is devoid of clinical manifestations in the patient. Progressive HTV-induced destruction of the immune system then leads to increased susceptibility to opportunistic infections, which eventually produces a syndrome called AJDS- related complex (ARC) characterized by symptoms such as persistent generalized lymphadenopathy, fever, and weight loss, followed itself by full blown AIDS.
After entry of the retrovirus into a cell, viral RNA is converted into DNA, which is then integrated into the host cell DNA. The reverse transcriptase encoded by the virus genome catalyzes the first of these reactions (Haseltine W. A. FASEB J. 1991, 5: 2349-2360). At least three functions have been attributed to the reverse transcriptase: RNA-dependent DNA polymerase activity which catalyzes the synthesis of the minus strand DNA from viral RNA, ribonuclease H (RNase H) activity which cleaves the RNA template from RNA-DNA hybrids and DNA- dependent DNA polymerase activity which catalyzes the synthesis of a second DNA strand from the minus strand DNA template (Goff S. P., J. Acq. Imm. Defic. Syndr. 1990, 3: 817-831). The double stranded DNA produced by reverse transcriptase, now called provirus, is then able to be inserted into host genomic DNA.
At the end of reverse transcription, the viral genome in the form of DNA is integrated into host genomic DNA and serves as a template for viral gene expression by the host transcription system, which leads eventually to virus replication (Sakai, H al., J. Virol. 1993, 67: 1169-1174). The preintegration complex consists of integrase, reverse transcriptase, pl7 and proviral DNA (Bukrinsky et al., Proc. Nat. Acad. Sci. USA 1992, 89: 6580-6584). The phosphorylated pl7 protein plays a key role in targeting the preintegration complex into the nucleus of host cell (Gallay et al., Cell 1995, 80:, 379-388).
As in the case of several other retroviruses, HTV encodes the production of a protease which carries out post-translational cleavage of precursor polypeptides in a process necessary for the formation of infectious virions (S. Crawford et al., J. Virol. 1985, 53: 899). These gene products include pol — which encodes the virion RNA- dependent DNA polymerase (reverse transcriptase), an endonuclease, and HTV protease — and gag — which encodes the core-proteins of the virion. (H. Toh et al., EMBO J. 1985, 4: 1267; L.H. Pearl et al., Nature 1987, 329- 351; M.D. Power et al, Science 1986, 231: 1567). A number of synthetic anti-viral agents targeted to various stages in the replication cycle of HTV have been disclosed. These agents include inhibitors of HTV cellular fusion (Turpin et al., Expert Opinion on Therapeutic Patents 2000, 10: 1899- 1909), reverse transcriptase inhibitors (e.g., didanosine, zidovudine (AZT), and efavirenz), integrase inhibitors (Neamati, Expert Opinion on Investigational Drugs 2000, 10: 281-296), and protease inhibitors. Protease inhibitors inhibit the formation of infectious virions by interfering with the processing of viral polyprotein precursors. Processing of these precursor proteins requires the action of virus-encoded proteases which are essential for replication (Kohl, N.E. et al., Proc. Natl. Acad. Sci. USA 1988, 85: 4686). Several HTV protease inhibitors are presently in clinical use for the treatment of AIDS and HTV infection, including indinavir (see US 5413999), nelfinavir (US 5484926), saquinavir (US 5196438), and ritonavir (US 5484801). Each of these protease inhibitors is a peptidomimetic, competitive inhibitor of the viral protease which prevents cleavage of the HTV gag-pol polyprotein precursor. Indinavir, for example, has been found to be highly effective in reducing HTV viral loads and increasing CD4 cell counts in HTV-infected patients, when used in combination with nucleoside reverse transcriptase inhibitors. See, for example, Hammer et al, New England J. Med. 1997, 337: 725-733 and Gulick et al., New England J. Med. 1997, 337: 734-739. A substantial and persistent problem in the treatment of AIDS has been the ability of the HTV virus to develop resistance to the therapeutic agents employed to treat the disease. Resistance to HTV-1 protease inhibitors has been associated with 25 or more amino acid substitutions in both the protease and the cleavage sites. Many of these viral variants are resistant to all of the HTV protease inhibitors currently in clinical use. See Condra et al., Drug Resistance Updates 1998, 1: 1-7; Condra et al, Nature 1995, 374: 569-571; Condra et al., J. Virol. 1996, 70: 8270-8276; Patrick et al., Antiviral Ther. 1996, Suppl. 1: 17-18; and Tisdale et al., Antimicrob. Agents Chemother. 1995, 39: 1704-1710.
Attempts to address the resistance issue with "salvage therapy" consisting of high doses of multiple protease inhibitors have only been moderately successful due to the high level of cross resistance and toxicities associated with these protease inhibitors. Accordingly, there remains a need for new protease inhibitors having improved effectiveness against the viral variants.
SUMMARY OF THE INVENTION
The present invention provides a novel class of N-[(3,4-dihydro-3- hydroxy-2H-l-benzopyran-4-yl]-γ-hydroxy-4-[[5-(halophenyl)-2-(furanyl or oxazolyl)] alkyl] -α-(arylmethyl or heteroarylmethyl)-2-[[(2,2,2- trifluoroethyl)amino]carbonyl]-l-piperazinepentanamides which are potent inhibitors of ΗTV protease including mutant forms thereof that are resistant to known protease inhibitors. A feature of the piperazinepentanamides in this class is that they exhibit little or no inhibition of either or both the CYP 2D6 and 2C9 enzymes relative to their inhibition of CYP 3A4. These compounds are useful in the inhibition of ΗTV protease, the prevention of infection by ΗTV, the treatment of infection by ΗTV and in the treatment of AIDS and/or ARC, when employed as compounds or pharmaceutically acceptable salts or hydrates (when appropriate) thereof, optionally as pharmaceutical composition ingredients, and optionally in combination with other antivirals, anti-infectives, immunomodulators, antibiotics or vaccines. More particularly, the present invention includes a compound of Formula (I):
Figure imgf000006_0001
wherein
A is CH or N;
Rl is _p or -Cl;
R2 and R3 are each independently -H or methyl; and
R4 IS
Figure imgf000006_0002
X is O or S;
Y is pyridyl which is optionally substituted with from 1 to 3 substituents each of which is independently halogen, cyano, Cχ-C6 alkyl, or C1-C6 alkoxy;
each Z is independently hydrogen, halogen, cyano, C1-C6 alkyl, or C1-C6 alkoxy; and
q is an integer from zero to 2; and with the proviso that when A is N and R4 is:
Figure imgf000007_0001
, then R2 and R3 are both -H;
or a pharmaceutically acceptable salt thereof.
The present invention also includes pharmaceutical compositions containing a compound of the present invention and methods of preparing such pharmaceutical compositions. The present invention further includes processes for preparing the compounds of the invention, and also includes methods of inhibiting HIV protease, delaying the onset of AIDS, treating AIDS, preventing infection by HTV, and treating infection by HTV.
The foregoing embodiments as well as other embodiments, aspects and features of the present invention are either further described in or will be apparent from the ensuing description, examples, and appended claims.
DETAILED DESCRIPTION OF THE INVENTION
The present invention includes the compounds of Formula (I) above. These compounds and their pharmaceutically acceptable salts are HTV protease inhibitors.
A first embodiment of the present invention is a compound of Formula
Figure imgf000007_0002
wherein each of the variables is as originally defined above; or a pharmaceutically acceptable salt thereof.
A second embodiment of the invention is a compound of Formula (I), wherein
AisCHorN;
Rl is -F or -Cl;
R2 and R are either both -H or both methyl;
R is
Figure imgf000008_0001
Y is pyridyl which is optionally substituted with from 1 to 3 substituents each of which is independently halogen, cyano, C1-C4 alkyl, or C1-C4 alkoxy;
or a pharmaceutically acceptable salt thereof.
An aspect of the second embodiment is a compound of Formula (I), wherein
AisCHorN;
Rlis-For-Cl;
R2 and R3 are either both -H or both methyl;
R4is
Figure imgf000009_0001
Q is -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy;
and all other variables are as defined in the second embodiment;
or a pharmaceutically acceptable salt thereof.
A third embodiment of the present invention is a compound of Formula (I), wherein
A is CH or N;
Rl is -F or -Cl;
R2 and R are either both -H or both methyl;
Figure imgf000009_0002
X is S or O;
each Z is independently -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy; and
q is an integer from zero to 2;
or a pharmaceutically acceptable salt thereof. An aspect of the third embodiment is a compound of Formula (I), wherein
A is CH or N;
Rl is -F or -Cl;
R2 and R3 are either both -H or both methyl;
R i is
Figure imgf000010_0001
Z is -H, C1-C4 alkyl, or C1-C4 alkoxy;
or a pharmaceutically acceptable salt thereof.
A fourth embodiment of the present invention is a compound of Formula (I), wherein
A is CH or N;
Rl is -F or -Cl;
R2 and R3 are either both -H or both methyl;
R i is
Figure imgf000010_0002
each Z is independently -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy; and
q is an integer from zero to 2; and with the proviso that when A is N and R4 is:
Figure imgf000011_0001
, then R2 and R3 are both -H;
or a pharmaceutically acceptable salt thereof.
An aspect of the fourth embodiment is a compound of Formula (I), wherein
A is CH or N;
Rl is -F or -Cl;
R2 and R3 are either both -H or both methyl; and
R4 is
Figure imgf000011_0002
and with the proviso that when A is N and R4 is:
Figure imgf000011_0003
or , then R2 and R3 are both -H;
or a pharmaceutically acceptable salt thereof.
A fifth embodiment of the present invention is a compound of Formula (I), wherein
A is CH or N;
Rl is -F or -Cl; R2 and R3 are either both -H or both methyl;
R is
Figure imgf000012_0001
each Z is independently -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy; and
q is an integer from zero to 2;
or a pharmaceutically acceptable salt thereof.
An aspect of the fifth embodiment is a compound of Formula (I), wherein
A is CH or N;
Rl is -F or -Cl;
R and R3 are either both -H or both methyl;
R is
Figure imgf000012_0002
Z is -H, C 1-C4 alkyl, or C1-C4 alkoxy;
or a pharmaceutically acceptable salt thereof.
Additional embodiments and aspects of the present invention include a compound of Formula (II) whose variables are as defined in any one of the second, third, fourth, and fifth embodiments set forth above, or in any one of the aspects thereof as set forth above.
Additional embodiments and aspects of the present invention include a compound of Formula (I) or of Formula (II), in which A is CH and all other variables are as originally defined above or as defined in any one of the foregoing embodiments and aspects. Still other embodiments and aspects of the present invention include a compound of Formula (I) or of Formula (II), in which A is N and all other variables are as originally defined above or as defined in any one of the foregoing embodiments and aspects.
Further embodiments and aspects of the present invention include a compound of Formula (I) or of Formula (II), in which Rl is -F and all other variables are as originally defined above or as defined in any one of the foregoing embodiments and aspects. Still further embodiments and aspects of the present invention include a compound of Formula (I) or of Formula (II), in which Rl is -Cl and all other variables are as originally defined above or as defined in any one of the foregoing embodiments and aspects.
Exemplifying the invention are compounds selected from the group consisting of
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
and pharmaceutically acceptable salts thereof.
Other embodiments of the present invention include the following: (a) A pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier. (b) A pharmaceutical composition which comprises the product prepared (i.e., made) by combining (e.g., mixing) a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier.
(c) The pharmaceutical composition of (a) or (b), wherein the composition further comprises a therapeutically effective amount of at least one AIDS treatment agent selected from the group consisting of AIDS antiviral agents, immunomodulators, and anti-infective agents.
(d) The pharmaceutical composition of (a) or (b), wherein the composition further comprises a therapeutically effective amount of at least one antiviral agent selected from the group consisting of non-nucleoside HTV reverse transcriptase inhibitors, nucleoside HTV reverse transcriptase inhibitors, HTV integrase inhibitors, and CCR5 receptor antagonists.
(e) The pharmaceutical composition of (d), further comprising a therapeutically effective amount of an additional HTV protease inhibitor. (g) The pharmaceutical composition of (a) or (b), wherein the composition further comprises a therapeutically effective amount of at least one antiviral agent which is a CCR5 receptor antagonist.
(h) The pharmaceutical composition of (a) or (b), wherein the composition further comprises a therapeutically effective amount of at least one antiviral agent which is an HTV integrase inhibitor.
(i) The pharmaceutical composition of (a) or (b), further comprising a cytochrome P450 monooxygenase inhibitor (e.g., ritonavir or a pharmaceutically acceptable salt thereof) in an amount effective to improve the pharmacokinetics of the compound. (j) A combination which comprises a therapeutically effective amount of a compound of Formula (I) and a therapeutically effective amount of an HTV infection/ AIDS treatment agent selected from the group consisting of HTV/ AIDS antiviral agents, immunomodulators, and anti-infective agents.
(k) The combination of (j), wherein the HTV infection/ AIDS treatment agent is an antiviral selected from the group consisting of non-nucleoside HTV reverse transcriptase inhibitors, nucleoside HTV reverse transcriptase inhibitors, HTV integrase inhibitors, and CCR5 receptor antagonists.
(1) A method of inhibiting HTV protease in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of a compound of Formula (I). (m) A method of preventing or treating infection by HTV in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of a compound of Formula (I).
(n) A method of delaying the onset of or treating AIDS in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of a compound of Formula (I).
(o) The method of (1) or (m) or (n), wherein the compound of Formula (I) is administered in combination with a therapeutically effective amount of at least one AIDS treatment agent selected from the group consisting of AIDS antiviral agents, immunomodulators, and anti-infective agents.
(p) The method of (1) or (m) or (n), wherein the compound of Formula (I) is administered in combination with a therapeutically effective amount of at least one antiviral agent selected from the group consisting of non-nucleoside HTV reverse transcriptase inhibitors and nucleoside HTV reverse transcriptase inhibitors. (q) The method of (1) or (m) or (n), wherein the compound is administered in combination with a cytochrome P450 monooxygenase inhibitor in an amount effective to improve the pharmacokinetics of the compound.
(r) A method of inhibiting HTV protease in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of any one of the compositions set forth in (a) to (i).
(s) A method of preventing or treating infection by HTV in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of any one of the compositions set forth in (a) to (i).
(t) A method of delaying the onset of or treating AIDS in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of any one of the compositions set forth in (a) to (i).
Additional embodiments of the invention include the pharmaceutical compositions, combinations and methods set forth in (a)-(t) above, wherein the compound employed therein is a compound of one of the embodiments or aspects of the compounds described above. It is to be understood that, unless expressly stated to the contrary, the compound of the present invention can be employed in the above- described pharmaceutical compositions, combinations, and methods in its free form or as a pharmaceutically acceptable salt.
As used herein, the term "C1-C6 alkyl" refers to a linear or branched chain alkyl group having from 1 to 6 carbon atoms, and is selected from the hexyl alkyl and pentyl alkyl isomers, n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl. "C1-C4 alkyl" refers to a linear or branched chain alkyl group having from 1 to 4 carbon atoms, and is selected from n-, iso-, sec- and t-butyl, n- and isopropyl, ethyl and methyl. The term "Cχ-C6 alkoxy" means an -O-alkyl group wherein alkyl is Cl to C6 alkyl as defined above. "C1-C4 alkoxy" has an analogous meaning; i.e., it is an alkoxy group selected from methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, and sec-butoxy. Similarly, "C1-C3 alkoxy" is selected from methoxy, ethoxy, n-propoxy, and isopropoxy. The term "aryl" refers to aromatic mono- and poly-carbocyclic ring systems, wherein the carbocyclic rings in the polyring systems may be fused or attached to each other via a single ring carbon. Exemplary aryl groups are phenyl and naphthyl.
The term "heteroaryl" refers to (i) a 5- or 6-membered aromatic ring consisting of carbon atoms and from 1 to 3 heteroatoms selected from N, S, and O or (ii) an 8- to 10-membered bicyclic ring system consisting of carbon atoms and from 1 to 3 heteroatoms selected from N, S, and O, wherein at least one of the rings in the bicyclic system is an aromatic ring.
The term "substituted" refers to mono- and poly-substitution by a named substituent to the extent such single and multiple substitution is chemically allowed and results in a chemically stable compound.
The symbol " fU ' " in front of an open bond in the structural formula of a group marks the point of attachment of the group to the rest of the molecule. When any variable or term occurs more than one time in any constituent or formulas set forth herein (e.g., Formula (I)), its definition on each occurrence is independent of its definition at every other occurrence.
Combinations of substituents and/or variables are permitted only to the extent such combinations result in stable compounds.
The compounds of the present invention are potent inhibitors of HTV protease including mutant forms thereof that are resistant to known protease inhibitors, as may be seen by reference to Examples 12 - 14 below. At the same, the compounds of the instant invention are significantly less potent inhibitors of CYP 2D6 and CYP 2C9 than structurally related piperazinepentanamide protease inhibitors. Accordingly, the compounds of the invention can be expected to avoid and/or have fewer drug-drug interactions compared to their structurally related counterparts. Inhibition of CYP 2D6 and 2C9 can be determined by many methods such as those detailed in Chiba et al., Drug metabolism and Disposition 1997, 25, No. 9: 1022-1031.
The present invention includes pharmaceutical compositions useful for inhibiting HTV protease, comprising an effective amount of a compound of this invention, and a pharmaceutically acceptable carrier. Pharmaceutical compositions useful for preventing or treating infection by HTV, for delaying the onset of AIDS, or for treating AIDS, are also encompassed by the present invention, as well as a method of inhibiting HTV protease, and a method of preventing or treating infection by HTV, of delaying the onset of AIDS, or of treating AIDS. An aspect of the present invention is a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention in combination with a therapeutically effective amount of an agent useful for treating HTV infection and/or AIDS (alternatively referred to as an HTV/ AIDS treatment agent) selected from: (1) an HTV/ATDS antiviral agent,
(2) an anti-infective agent, and
(3) an immunomodulator.
The present invention also includes the use of a compound of the present invention as described above as a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, or (c) treating AIDS. The present invention further includes the use of a compound of the present invention as described above in the preparation of a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, (c) delaying the onset of AIDS, or (d) treating AIDS. The present invention also includes the use of any of the HTV protease inhibiting compounds of the present invention as described above in combination with one or more HTV/ATDS treatment agents selected from an HTV/ATDS antiviral agent, an anti-infective agent, and an immunomodulator for use as a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, (c) delaying the onset of AIDS, or (d) treating AIDS, said medicament comprising an effective amount of the HTV protease inhibitor compound and an effective amount of the one or more treatment agents.
The present invention further includes the use of any of the HTV protease inhibiting compounds of the present invention as described above in combination with one or more HTV/ AIDS treatment agents selected from an HTV/ATDS antiviral agent, an anti-infective agent, and an immunomodulator for the manufacture of a medicament for (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, (c) delaying the onset of ADDS, or (d) treating ADDS, said medicament comprising an effective amount of the HTV protease inhibitor compound and an effective amount of the one or more treatment agents.
The present invention also includes a compound of the present invention for use in (a) inhibiting HTV protease, (b) preventing or treating infection by HTV, or (c) preventing, treating or delaying the onset of ADDS. In this use, the compound of the present invention can optionally be employed in combination with one or more HTV/ATDS treatment agents selected from HTV/ATDS antiviral agents, anti-infective agents, and immunomodulators.
The compounds of the present invention have asymmetric centers and can have additional centers (e.g., where one of R and R3 is -H and the other is methyl), in which event the compound of the present invention may occur as a mixture of stereoisomers or as individual diastereomers, with all isomeric forms being included in the present invention.
A therapeutically effective amount of the compounds of the present invention are useful in the inhibition of HTV protease, the prevention or treatment of infection by HTV and the delay in the onset of or treatment of consequent pathological conditions such as ADDS. Treating or delaying the onset of ADDS or preventing or treating infection by HTV is defined as including, but not limited to, treating a wide range of states of HTV infection: ADDS, ARC, both symptomatic and asymptomatic, and actual or potential exposure to HTV. For example, the compounds of this invention are useful in treating infection by HTV after suspected past exposure to HTV by e.g., blood transfusion, exchange of body fluids, bites, accidental needle stick, or exposure to patient blood during surgery. The compounds of the invention can also be used in "salvage" therapy; i.e., the compounds can be used to treat HTV infection, ADDS, or ARC in HIV-positive subjects whose viral load achieved undetectable levels via conventional therapies employing known protease inhibitors, and then rebounded due to the emergence of HTV mutants resistant to the known inhibitors.
The compounds of this invention are useful in the preparation and execution of screening assays for antiviral compounds. For example, the compounds of this invention are useful for isolating enzyme mutants, which are excellent screening tools for more powerful antiviral compounds. Furthermore, the compounds of this invention are useful in establishing or determining the binding site of other antivirals to HTV protease, e.g., by competitive inhibition. Thus the compounds of this invention are commercial products to be sold for these purposes.
The present invention also provides for the use of a compound of structural formula (I) to make a pharmaceutical composition useful for inhibiting HTV protease, treating or preventing HTV infection, delaying the onset of ADDS, and for treating ADDS.
The compounds of the present invention may be administered in the form of pharmaceutically acceptable salts. The term "pharmaceutically acceptable salt" refers to all acceptable salts of the compounds of Formula (I) (in the form of water- or oil-soluble or dispersible products) and includes the conventional non-toxic salts or the quaternary ammonium salts which are formed, e.g., from inorganic or organic acids or bases. The salt can be used as a dosage form for modifying the solubility or hydrolysis characteristics of the compound or can be used in sustained release or pro-drug formulations.
The compounds of the present invention may be administered orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, in dosage unit formulations containing conventional non-toxic pharmaceutically- acceptable carriers, adjuvants and vehicles.
The term "administration" and variants thereof (e.g., "administering" a compound) in reference to a compound of the invention each mean providing the compound or a prodrug of the compound to the individual in need of treatment. When a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., ADDS antivirals), "administration" and its variants are each understood to include concunent and sequential provision of the compound or prodrug thereof and other agents.
The present invention further provides for preventing or treating HTV infection and for delaying the onset of or treating ADDS by administering to a subject in need of such treatment a pharmaceutical composition comprising a pharmaceutical carrier and a therapeutically effective amount of a compound of the present invention.
As used herein, the term "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 expression "pharmaceutically acceptable" means that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
The term "subject," (alternatively referred to herein as "patient") as used herein refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
The term "therapeutically effective amount" as used herein means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease being treated.
These pharmaceutical compositions may be in the form of orally- administrable suspensions or tablets or capsules, nasal sprays, sterile injectible preparations, for example, as sterile injectible aqueous or oleagenous suspensions or suppositories.
When administered orally as a suspension, these compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may contain microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and 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.
When administered by nasal aerosol or inhalation, these compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
The injectible 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. When rectally administered in the form of suppositories, these compositions may be prepared by mixing the drug with a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters of polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
The compounds of this invention can be administered orally to humans in a dosage range of 0.01 to 1000 mg/kg body weight per day in a single dose or in divided doses. One preferred dosage range is 0.1 to 200 mg/kg body weight per day orally in a single dose or in divided doses. Another preferred dosage range is 0.5 to 100 mg/kg body weight per day orally in single or divided doses. For oral administration, the compositions are preferably provided in the form of tablets containing 1 to 1000 milligrams of the active ingredient, particularly 1, 5, 10, 15. 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy. The present invention is also directed to combinations of the HTV protease inhibitor compounds with one or more agents useful in the treatment of HTV infection and/or ADDS. For example, the compounds of this invention may be effectively administered, whether at periods of pre-exposure and/or post-exposure, in combination with effective amounts of the HIV/ ADDS antivirals, imunomodulators, antiinfectives, or vaccines, such as those in Table 1 as follows:
TABLE 1 - HTV/ATDS ANTTVIRALS, TMUNOMODULATORS, ANTDNFECTTVES, AND OTHER TREATMENTS
ANTΓVΓRALS
Drug Name Manufacturer Indication (Tradename and/or Location)
Amprenavir Glaxo Wellcome HTV infection, ADDS, 141 W94 (AGENERASE®) ARC GW 141 (protease inhibitor)
Abacavir Glaxo Welcome HTV infection, ADDS, ARC GW 1592 (ZIAGEN®) (reverse transcriptase 1592U89 inhibitor)
Acemannan Carrington Labs ARC (Irving, TX)
Acyclovir Burroughs Wellcome HTV infection, ADDS, ARC in combination with AZT
AD-439 Tanox Biosystems HTV infection, ADDS, ARC
AD-519 Tanox Biosystems HTV infection, ADDS, ARC
Adefovir dipivoxil Gilead Sciences HTV infection
AL-721 Ethigen ARC, PGL, HTV positive,
(Los Angeles, CA) ADDS
Alpha Interferon Glaxo Wellcome Kaposi's sarcoma, HTV, in combination w/Retrovir
Ansamycin Adria Laboratories ARC LM 427 (Dublin, OH) Erbamont (Stamford, CT)
Antibody which Advanced Biotherapy ADDS, ARC neutralizes pH labile alpha Concepts (Rockville, aberrant Interferon MD)
AR177 Aronex Pharm HTV infection, ADDS, ARC beta-fluoro-ddA Nat'l Cancer Institute ADDS -associated diseases
BMS-232623 Bristol-Myers Squibb/ HTV infection, ADDS, (CGP-73547) Novartis ARC
(protease inhibitor) BMS-234475 Bristol-Myers Squibb/ HTV infection, ADDS, (CGP-61755) Novartis ARC (protease inhibitor)
CI-1012 Warner-Lambert HΓV-1 infection
Cidofovir Gilead Science CMV retinitis, herpes, papillomavirus
Curdlan sulfate AJI Pharma USA HTV infection
Cytomegalovirus immune Medlmmune CMV retinitis globin
Cytovene Syntex sight threatening CMV Ganciclovir peripheral CMV retinitis
Delavirdine Pharmacia-Upjohn HTV infection, ADDS, (RESCRIPTOR®) ARC (nonnucleoside reverse transcriptase inhibitor)
Dextran Sulfate Ueno Fine Chem. Did. ADDS, ARC, HTV Ltd. (Osaka, Japan) positive asymptomatic ddC Hoffman-La Roche HTV infection, ADDS, ARC
Dideoxycytidine (HJVDD®) ddl Bristol-Myers Squibb HTV infection, ADDS, ARC; Dideoxyinosine (VDDEX®) combination with AZT/d4T
mozenavir AVDD (Camden, NJ) HTV infection, ADDS, ARC (DMP-450) (protease inhibitor)
EL10 Elan Corp, PLC HTV infection (Gainesville, GA)
Efavirenz DuPont HTV infection, ADDS, (DMP 266) (SUSTΓVA®) ARC (non-nucleoside RT
Merck (STOCRTN®) inhibitor)
Famciclovir Smith Kline herpes zoster, herpes simplex
FTC Emory University HIV infection, ADDS, ARC (reverse transcriptase inhibitor)
GS 840 Gilead HTV infection, ADDS, ARC (reverse transcriptase inhibitor) HBY097 Hoechst Marion Roussel HTV infection, ADDS, ARC (non-nucleoside reverse transcriptase inhibitor)
Hypericin VTMRx Pharm. HTV infection, ADDS, ARC
Recombinant Human Triton Biosciences ADDS, Kaposi's sarcoma, Interferon Beta (Almeda, CA) ARC
Interferon alfa-n3 Interferon Sciences ARC, ADDS
Indinavir Merck (CRTXTVAN®) HTV infection, ADDS, ARC, asymptomatic HTV positive, also in combination with AZT/ddl/ddC
ISIS 2922 ISIS Pharmaceuticals CMV retinitis KNI-272 Nat'l Cancer Institute HTV-assoc. diseases Lamivudine, 3TC Glaxo Wellcome HTV infection, ADDS,
(EPTVIR®) ARC (reverse transcriptase inhibitor); also with AZT
Lobucavir Bristol-Myers Squibb CMV infection Nelfinavir Agouron HTV infection, ADDS, (VIRACEPT®) ARC (protease inhibitor)
Nevirapine Boeheringer HTV infection, ADDS,
Ingleheim ARC (non-nucleoside
(VIRAMUNE®) reverse transcriptase inhibitor)
Novapren Novaferon Labs, Inc. HTV inhibitor (Akron, OH)
Peptide T Peninsula Labs ADDS
Octapeptide (Belmont, CA)
Sequence
Trisodium Astra Pharm. Products, CMV retinitis, HTV infection, Phosphonoformate Inc other CMV infections
PNU-140690 Pharmacia Upjohn HTV infection, ADDS, ARC (protease inhibitor)
Probucol Vyrex HTV infection, ADDS RBC-CD4 Sheffield Med. Tech HTV infection, ADDS, (Houston TX) ARC Ritonavir Abbott HTV infection, ADDS, (ABT-538) (RΠΌNAVIJR®) ARC (protease inhibitor)
Saquinavir Hoffmann-LaRoche HTV infection, ADDS, (FORTOVASE®) ARC (protease inhibitor)
Stavudine; d4T Bristol-Myers Squibb HTV infection, ADDS, ARC
Didehydrodeoxy- (ZERIT®) thymidine
Valaciclovir Glaxo Wellcome genital HSV & CMV infections
Virazole Viratek/ICN (Costa asymptomatic HTV positive, Ribavirin Mesa, CA) LAS, ARC
VX-478 Vertex HTV infection, ADDS, ARC
Zalcitabine Hoffmann-La Roche HTV infection, ADDS, ARC, with AZT
Zidovudine; AZT Glaxo Wellcome HTV infection, ADDS, ARC, (RETROVTR®) Kaposi's sarcoma in combination with other therapies (reverse transcriptase inhibitor)
Lopinavir (ABT-378) Abbott HTV infection, ADDS, ARC (protease inhibitor)
Lopinavir + ritonavir Abbott (KALETRA®) HTV infection, ADDS, ARC (ABT-378/r); Kaletra (protease inhibitor)
JE2147/AG1776 Agouron HTV infection, ADDS, ARC (protease inhibitor)
T-20 Trimeris HTV infection, ADDS, ARC (fusion inhibitor)
T-1249 Trimeris HTV infection, ADDS, ARC (fusion inhibitor) atazanavir (BMS 232632); Bristol-Myers-Squibb HTV infection, ADDS, ARC Zrivada (ZRIVADA®) (protease inhibitor)
PRO 542 Progenies HIV infection, ADDS, ARC (attachment inhibitor)
PRO 140 Progenies HTV infection, ADDS, ARC (CCR5 co-receptor inhibitor) TAK-779 Takeda HTV infection, ADDS, ARC (injectable CCR5 receptor antagonist)
DPC 681 & DPC 684 DuPont HTV infection, ADDS, ARC (protease inhibitors)
DPC 961 & DPC 083 DuPont HTV infection ADDS, ARC (nonnucleoside reverse transcriptase inhibitors) abacavir + lamivudine + GlaxoSmithKline HTV infection, ADDS, ARC zidovudine (TRIZTVTR®) (reverse transcriptase inhibitors) tipranavir (PNU-140690) Boehringer Ingelheim HTV infection, ADDS, ARC (protease inhibitor) tenofovir Gilead (VTREAD®) HTV infection, ADDS, ARC (nucleotide reverse transcriptase inhibitor)
TMC-120 & TMC-125 Tibotec HTV infections, ADDS, ARC (non-nucleoside reverse transcriptase inhibitors)
TMC-126 Tibotec HTV infection, ADDS, ARC (protease inhibitor)
IMMUNO-MODULATORS
Drug Name Manufacturer Indication AS-101 Wyeth-Ayerst ADDS Bropirimine Pharmacia Upjohn advanced ADDS Acemannan Carrington Labs, Inc. ADDS, ARC (Irving, TX)
CL246/738 American Cyanamid ADDS, Kaposi's sarcoma Lederle Labs
EL10 Elan Corp, PLC HTV infection (Gainesville, GA)
FP-21399 Fuki ImmunoPharm blocks HTV fusion with CD4+ cells
Gamma Interferon Genentech ARC, in combination w/TNF (tumor necrosis factor) Granulocyte Macrophage Genetics Institute ADDS Colony Stimulating Factor Sandoz
Granulocyte Macrophage Hoeschst-Roussel ADDS Colony Stimulating Factor Immunex
Granulocyte Macrophage Schering-Plough ADDS, combination w/AZT Colony Stimulating Factor
HTV Core Particle Rorer seropositive HTV Immunostimulant
TL-2 Cetus ADDS, in combination
Interleukin-2 w/AZT
IL-2 Hoffman-La Roche ADDS, ARC, HTV, in Interleukin-2 Immunex combination w/AZT
D_-2 Chiron ADDS, increase in CD4 cell
Interleukin-2 (aldeslukin) counts
Immune Globulin Cutter Biological pediatric ADDS, in ntravenous (human) (Berkeley, CA) combination w/AZT
ΓMREG-1 Imreg (New Orleans, ADDS, Kaposi's sarcoma, LA) ARC, PGL
TMREG-2 Imreg (New Orleans, ADDS, Kaposi's sarcoma, LA) ARC, PGL
Lmuthiol Diethyl Dithio Merieux Institute ADDS, ARC Carbamate
Alpha-2 Interferon Schering Plough Kaposi's sarcoma w/AZT, ADDS
Methionine- Enkephalin TNI Pharmaceutical ADDS, ARC (Chicago, TL)
MTP-PE Ciba-Geigy Corp. Kaposi's sarcoma Muramyl-Tripeptide
Granulocyte Colony Amgen ADDS, in combination Stimulating Factor w/AZT
Remune Immune Response Corp. immunotherapeutic rCD4 Recombinant Genentech ADDS, ARC Soluble Human CD4 rCD4-IgG hybrids ADDS, ARC
Recombinant Soluble Biogen ADDS, ARC Human CD4 Interferon Alfa 2a Hoffman-La Roche Kaposi's sarcoma, ADDS, ARC, in combination w/AZT
SK&F106528 Smith Kline HTV infection Soluble T4
Thymopentin Immunobiology HTV infection Research Institute
Tumor Necrosis Factor; Genentech ARC, in combination
TNF w/gamma Interferon etanercept Immunex Corp rheumatoid arthritis (ENBREL®) infliximab Centocor rheumatoid arthritis and
(REMICADE®) Crohn's disease
ANTI-ΓNFECTΓVΈS
Drug Name Manufacturer Indication
Clindamycin with Pharmacia Upjohn PCP Primaquine
Fluconazole Pfizer cryptococcal meningitis, candidiasis
Pastille Nystatin Pastille Squibb Corp. prevention of oral candidiasis
Ornidyl Eflornithine Menell Dow PCP
Pentamidine Isethionate LyphoMed PCP treatment (TM & TV) (Rosemont, TL)
Trimethoprim antibacterial
Trimethoprim/sulfa antibacterial
Piritrexim Burroughs Wellcome PCP treatment
Pentamidine isethionate Fisons Corporation PCP prophylaxis for inhalation
Spiramycin Rhone-Poulenc cryptosporidia diarrhea
Intraconazole-R51211 Janssen Pharm. histoplasmosis; cryptococcal meningitis
Trimetrexate Warner-Lambert PCP OTHER
Drug Name Manufacturer Indication
Daunorubicin NeXstar, Sequus Karposi's sarcoma
Recombinant Human Ortho Pharm. Corp. severe anemia assoc. with Erythropoietin AZT therapy
Recombinant Human Serono ADDS-related wasting, Growth Hormone cachexia
Leukotriene B4 Receptor HTV infection Antagonist
Megestrol Acetate Bristol-Myers Squibb treatment of anorexia assoc. w/ATDS
Soluble CD4 Protein and HTV infection Derivatives
Testosterone Alza, Smith Kline ADDS-related wasting
Total Enteral Nutrition Norwich Eaton diarrhea and malabsorption, Pharmaceuticals related to ADDS
It will be understood that the scope of combinations of the compounds of this invention with HTV/ATDS antivirals, immunomodulators, anti-infectives or vaccines is not limited to the list in Table 1 above, but includes in principle any combination with any pharmaceutical composition useful for the treatment of HTV infection and/or ADDS. When employed in combination with the compounds of the invention, the HTV/ATDS antivirals and other agents are typically employed in their conventional dosage ranges and regimens as reported in the art, including the dosages described in the Physicians' Desk Reference, 54th edition, Medical Economics Company, 2000. The dosage ranges for a compound of the invention in these combinations are the same as those set forth above just before Table 1.
One suitable combination is a compound of the present mvention and a nucleoside inhibitor of HTV reverse transcriptase such as AZT, 3TC, ddC, or ddl. Another suitable combination is a compound of the present invention and a non- nucleoside inhibitor of HTV reverse transcriptase, such as efavirenz, and optionally a nucleoside inhibitor of HTV reverse transcriptase, such as AZT, 3TC, ddC or ddl. Still another suitable combination is any one of the combinations in the preceding paragraph, further comprising an additional HTV protease inhibitor such as indinavir, Compound A, nelfinavir, ritonavir, saquinavir, amprenavir, or abacavir. An aspect of this combination is the combination wherein the additional inhibitor of HTV protease is the sulfate salt of indinavir. Another aspect of this combination is the combination in which the additional protease inhibitor is selected from nelfinavir and ritonavir. Still another aspect of this combination is the combination in which the additional inhibitor of HTV protease is saquinavir, which is typically administered in a dosage of 600 or 1200 mg tid. Other suitable combinations include a compound of the present invention with the following (1) efavirenz, optionally with AZT and/or 3TC and/or ddl and/or ddC, and optionally with indinavir; (2) any of AZT and or ddl and/or ddC and/or 3TC, and optionally with indinavir; (3) d4T and 3TC and/or AZT; (4) AZT and 3TC; and (5) AZT and d4T. Another aspect of the present invention is co-administration of a compound of the present invention with an inhibitor of cytochrome P450 monooxygenase in an amount effective to improve the pharmacokinetics of the compound. Compounds of the invention can be metabolized, at least in part, by cytochrome P450 (CYP3A4). Co-administration of compounds of the invention with a cytcochrome P450 inhibitor can improve the pharmacokinetic profile of the compound in subjects (e.g., humans); i.e., co-administration can increase C aχ (the maximum plasma concentration of the compound), AUC (area under the curve of plasma concentration of the compound versus time), and/or the half -life of the compound. A suitable P450 inhibitor is ritonavir. It is to be understood that the primary role of ritonavir in this circumstance is as a pharmacokinetic modulator and not as a protease inhibitor; i.e., an amount of ritonavir which is effective for improving the pharmacokinetics of the compound can provide a secondary or even negligible contribution to the antiviral effect.
A compound of the present invention can also be administered in combination with an HTV integrase inhibitor such as a compound described in WO 99/62520, WO 99/62513, or WO 99/62897. A compound of the present invention can also be administered in combination with a CCR5 receptor antagonist, such as a compound described in WO 00/59502, WO 00/59503, WO 00/59497, WO 00/59498, WO 00/76511, WO 00/76512, WO 00/76513, WO 00/76514, WO 00/76972, or WO 00/76793. In the above-described combinations, the compound of the present invention and other active agents may be administered together or separately. In addition, the administration of one agent may be prior to, concunent with, or subsequent to the administration of other agent(s). These combinations may have unexpected or synergistic effects on limiting the spread and degree of infection of HTV.
Efavirenz is (-)-6-chloro-4-cyclopropylethynyl-4-trifluoromethyl-l ,4- dihydro-2H-3,l-benzoxazin-2-one, also known as DMP-266 or SUSTTVA® (DuPont) or STOCRTN® (Merck). Efavirenz and its utility as an HTV reverse transcriptase inhibitor is described in US 5519021 and in the corresponding PCT published application, WO 95/20389. Efavirenz can be synthesized by the protocol of US 5633405. Additionally, the asymmetric synthesis of an enantiomeric benzoxazinone by a highly enantioselective acetylide addition and cyclization sequence is described in Thompson et al., Tetrahedron Letters 1995, 36: 8937-40, as well as in the PCT publication, WO 96/37457.
AZT is 3'-azido-3'-deoxythymidine, is also known as zidovudine, and is available from Burroughs-Wellcome under the tradename RETROVTR®. Stavudine is 2',3'-didehydro-3'-deoxythymidine, is also known as 2',3'-dihydro-3'- deoxythymidine and d4T, and is available from Bristol-Myers Squibb under the tradename ZERIT®. 3TC is (2R-cis)-4-amino-l-[2-(hydroxymethyl)-l,3-oxathiolan- 5-yl]-2(lH)-pyrimidinone, is also known as (-)-l-[(2R,5S)-2-(hydroxymethyl)-l,3- oxathiolan-5-yl]cytosine and lamivudine, and is available from Glaxo Wellcome under the tradename EPTVTR®. ddC is 2',3'-dideoxycytidine, is also known as zalcitabine, and is available from Hoffman LaRoche under the tradename HΓVDD®. ddl is 2',3 '-dideoxyinosine, is also known as didanosine, and is available from Bristol- Myers-Squibb under the tradename VDDEX®. The preparation of ddC, ddl and AZT are also described in EPO 0,484,071.
Indinavir is 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, and can be prepared as described in US 5413999. Indinavir is generally administered as the sulfate salt at a dosage of 800 mg three times a day. Indinavir sulfate is available from Merck under the tradename CRTXTVAN®.
Compound A is N-(2(R)-hydroxy-l(S)-indanyl)-2(R)-phenylmethyl- 4(S)-hydroxy-5-(l-(4-(2-benzo[b]furanylmethyl)-2(S)-N'-(t-butylcarboxamido)- piperazinyl))pentaneamide, preferably administered as the sulfate salt. Compound A can be prepared as described in US 5646148.
Ritonavir is [5S-(5R*,8R*,10R*, HR*)]-10-hydroxy-2-methyl-5-(l- methylethyl)-l -[2-( 1 -methylethyl)-4-thiazolyl]-3 ,6-dioxo-8 , 11 -bis(phenylmethyl)-2, 4, 7, 12-tetraazatridecan-13-oic acid 5-thiazolylmethyl ester, also known as 5- thiazolylmethyl [(aS)-a-[(lS,3S)-l-hydroxy-3-[(2S)-2-[3-[(2-isopropyl-4- thiazolyl)methyl]-3-methylureido]-3-methylbutyramido]-4- phenylbutyl]phenethyl]carbamate. It is available from Abbott under the tradename NORVTR®. Ritonavir can be prepared as described in US 5484801. Nelfinavir is [3S-[2(2S*,3S*),3a,4ab,8ab]]-N-(l,l- dimethylethyl)decahydro-2- [2-hydroxy-3 - [(3 -hydroxy-2-methylbenzoyl)amino] -4- (phenylthio)butyl]-3-isoquinolinecarboxamide, also known as (3S,4aS,8aS)-N-tert- Butyl-2-[(2R,3R)-3-(3,2-crestoamido)-2-hydroxy-4-(phenylthio)butyl]decahydro-3- isoquinolinecarboxamide. VERACEPT®, the monomethanesulfonate salt of nelfinavir (nelfinavir mesylate) is commerically available from Agouron. Nelfinavir can be prepared as described in US 5484926.
Saquinavir is N-tert-butyl-decahydro-2-[2(R)-hydroxy-4-phenyl- 3(S)-[[N-(2-quinolylcarbonyl)-L-asparaginyl]amino]butyl]-(4aS,8aS)- isoquinoline-3(S)-carboxamide. Saquinavir can be prepared in accordance with procedures disclosed in US 5196438. TNVTRASE® (saquinavir mesylate) is available from Roche Laboratories.
Amprenavir is 4-amino-N-((2 syn,3S)-2-hydroxy-4-phenyl-3-((S)- tetrahydrofuran-3-yloxycarbonylamino)-butyl)-N-isobutyl-benzenesulfonamide, also known as Compound 168 and 141 W94. Amprenavir is an aspartyl protease inhibitor that can be prepared by following the procedures described in
US 5585397. Amprenavir is available under the tradename AGENERASE® from Glaxo Wellcome. Amprenavir can be prepared as described in US 5783701.
Abacavir is (lS,4R)-cis-4-[2-amino-6-(cyclopropylamino)-9H- purin-9-yl]-2-cyclopentene-l-methanol, also known as 1592U89. Abacavir can be prepared by following the protocol of EP 0434450.
Abbreviations used in the instant specification include the following:
ACN = acetonitrile AcOH = acetic acid ADDS = acquired immune deficiency syndrome Alloc = allyloxycarbonyl
ARC = ADDS related complex
BOC or Boc = t-butyloxycarbonyl
BOC-ON = 2-(tert-butoxycarbonylamino)-2-phenyl acetonitrile 5 Bu = butyl
CBZ = carbobenzoxy (alternatively, benzyloxycarbonyl)
CSA = camphorsulfonic acid
DCE = dichloroethane
DCM = dichloromethane 10 DMF = dimethylformamide
DMSO = dimethylsulfoxide
DTEA = diisopropylethylamine
EDC = l-ethyl-3-(3-dimethylaminopropyl) carbodiimide
ES = electron spray (ionization) 15 Et = ethyl
Et2θ = diethyl ether
EtOAc = ethyl acetate
EtOH = ethanol
HBTU = 1-hydroxybenzotriazole 20 HTV = human immunodeficiency virus
HO AT = l-hydroxy-7-azabensotriazole
HOBT = 1-hydroxy benzotriazole hydrate
HPLC = high performance liquid chromatography
LC = liquid chromatography 25 LDA = lithium diisopropylamide
Me = methyl
MeOH = methanol
MS = mass spectrometry
NMP = N-methyl pynolidinone 30 NMR = nuclear magnetic resonance
Ph = phenyl
TB AF = tetrabutylammonium fluoride
TBDC = di t-butyl dicarbonate
TBSC1 = t-butyldimethylsilyl chloride 35 TBSOTf = t-butyldimethylsilyl triflate TEA = triethylamine TFA = trifluoroacetic acid TFEA = trifluoroethylamine Tf2θ = triflic anhydride THF = tetrahydrofuran
TLC = thin layer chromatgraphy TMSCN = trimethylsilyl cyanide TsOH = p-toluenesulfonic acid
The compounds of the present invention can be readily prepared according to the following reaction schemes and examples, or modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants which are themselves known to those of ordinary skill in this art, but are not mentioned in greater detail. Furthermore, other methods for preparing compounds of the invention will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples. Unless otherwise indicated, all variables are as defined above.
The preparation of the compounds of the present invention can be carried out in sequential or convergent synthetic routes, as shown in Schemes 1-7 below. A compound of Formula (I) can be prepared in accordance with Scheme 1, wherein Compound I is readily prepared via literature procedures described in Dorsey et al., J. Med. Chem. 1994, 37: 3443-3451, and also in US 5413999. Treatment of the hydroxyl compound 1 with triflic anhydride and lutidine in an inert solvent such as dichloromethane provides triflate 2. Displacement of the triflate with piperazine 3 occurs on heating in an inert solvent such as isopropanol to give lactone 4. Hydrolysis of lactone 4 with an aqueous lithium hydroxide provides the hydroxy acid which is conveniently protected with a standard silyl protecting group such as t-butyldimethylsilyl by reaction with either t-butyldimethylsilyl chloride in the presence of imidazole in an inert solvent or the reaction with the silyl triflate and diisopropyl ethylamine in an inert solvent such as dichloromethane. Mild aqueous hydrolysis of the silyl ester provides the protected hydroxy-acid 5. Amide coupling of compound 5 with aminochromanol to obtain 6 is typically performed by the carbodiimide method with reagents such as l-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) and HOBT in an inert solvent such as dichloromethane. Other methods of forming the amide or peptide bond include, but are not limited to, the synthetic routes via an acid chloride, azide, mixed anhydride or activated ester. The silyl protecting group is removed with fluoride to arrive at compound 7. The BOC protecting group on the amine is then removed with a strong acid such as trifluoroacetic acid or hydrochloric acid in an alcoholic solvent such as methanol to give the penultimate intermediate 8. Penultimate 8 is then reacted with the desired aldehyde 9 and a reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride in an inert solvent such as dichloromethane to give compound 10.
SCHEME 1
Figure imgf000038_0001
SCHEME 1 (continued)
Figure imgf000039_0001
SCHEME 1 (continued)
Figure imgf000040_0001
A more convergent route to compounds of the present invention is presented in Scheme 2, below. The orthogonally protected piperazine 11 can be selectively deprotected. The BOC protecting group can be removed by treatment with strong acids such as trifluoroacetic acid in dichloromethane or HCI in methanol. The resulting amine 12 can then be reacted with an aldehyde in the presence of a reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride to give piperazine 13. Removal of the Alloc protecting group is readily accomplished with a palladium catalyst in the presence of a nucleophilic trapping agent such as 1,3- dimethylbarbituric acid or as in J. Org. Chem. 1993, 58, 6109-6113. Displacement of the triflate of 2 with piperazine 14, as in Scheme 1 gives lactone 15 which is then converted into compounds of the present invention following the route depicted in Scheme 1. SCHEME 2
Figure imgf000041_0001
An alternative route to the instant compounds is presented in Scheme 3. Compound 16 can be easily prepared according to the procedures described in the literature including, but not limited to, those described in Tetrahedron Letters 1995, 36: 2195-2198, US 5646148, Tetrahedron Letters 1991, 32: 711-714, Tetrahedron Letters 1995, 36: 3993-3996 and Synthesis 1998, 938-961. A procedure for preparing czs-aminochromanols by the stereoselective hydrogen bromide-promoted hydrogenation of an α-hydroxyoxime is described in Davies et al., Tetrahedron Letters 2000, 41: 8021-8025.
As shown in Part A of Scheme 3, the epoxide opening can be carried out by heating piperazine 3 and the epoxide in an inert solvent. Acidic removal of the protecting groups can be accomplished by treatment with hydrochloric acid in an alcoholic solvent such as methanol, ethanol or isopropanol. The resulting intermediate 18 is then reductively aminated as in Scheme 1 to provide the compounds of the present invention. Alternatively, as shown in Part B of Scheme 3, the epoxide opening can be preformed with fully elaborated piperazine 14 to give 20. Once again the protecting group is removed with strong acid to give 19.
SCHEME 3
Figure imgf000042_0001
Figure imgf000043_0001
Piperazine intermediates are readily prepared from the known piperazine carboxylic acid 21, which can be prepared as described in He/. Chem. Acta. 1960, 43: 888-896. Selective monoprotection of the piperazine is carried out using BOC anhydride as described in Tetrahedron Letters 1989, 30: 5193-5196. The remaining unprotected amine can then be protected with any number of chloroformates including allyl chloroformate or benzyl chloroformate to give 23. Amide couplings of 23 with trifluoroethylamine to give 24 are performed using standard amide coupling reactions as described above. Acidic removal of the BOC protecting group as before gives 25. The Alloc group can be removed as before. The CBZ group is readily removed by hydrogenolysis with a palladium catalyst under a hydrogen atmosphere in an alcoholic solvent such as methanol or ethanol. Removal of the protecting groups can also be accomplished by a number of methods known in the art, such as those described in Greene, Protective Groups in Organic Synthesis, John Wiley and Sons, New York, 1991. These deprotected intermediates are then carried onto compounds of the instant invention via the synthetic routes shown in Schemes 1, 2 and 3.
SCHEME 4
Figure imgf000045_0001
Alloc]
NH£ \
EDC, HOBT CF3
Figure imgf000045_0002
The desired aldehyde intermediates are, in many cases, commercially available (e.g., Aldrich Chemical). Other aldehydes of interest can be prepared by literature methods including classical methods familiar to those skilled in the art. Stille and Suzuki coupling of commercially and readily available aryl and heteroaryl halides, aryl trialkylstannanes, and arylboronic acids also provides the desired aldehydes as exemplified for bromofuran in Scheme 5 below. Aldehyde 27 can be reacted with trialkylarylstannane 26 in the presence of a palladium catalyst by the method of Gronowitz et al. , J. Heterocyclic Chem. 1995, 35: 771, to give 28. Alternatively, trialkylstannane 30 can be coupled with arylhalides such as 29 to give 31 which can be deprotected under mild conditions with dilute hydrochloric acid to give aldehyde 28. Other aldehydes are available via metal halogen exchange followed by anion quenching with DMF as described by Vogel et al., J. Chem. Soc. Perkin Trans I, 1974, 37. Metalation of a biaryl or heterobiaryl compound such as 32 with a strong base such as n-butyllithium at low temperature in an inert solvent such as THF followed by anion trapping with DMF also provides aldehydes such as 28.
SCHEME 5
ArSnMθ3
Figure imgf000046_0001
26 27 28
[Ar = aryl]
HCI
Figure imgf000046_0002
29 30 31
Figure imgf000046_0003
32 28
When R and R3 are alkyl, the necessary intermediates can be formed as shown in Scheme 6 below. Piperazine 12 can be treated with TMSCN and a ketone in acetic acid to give intermediate 34 according to the method described in J. Org. Chem. 1990, 55, 4207-4209. The Alloc protecting group is removed as described in Scheme 4 and the resulting intermediate, 35, is then treated with an excess of a Grignard to give the gem-dialkyl compound 14 A. This intermediate is then converted to the compounds of the present invention via chemistry described in Schemes 2 and 3 above. SCHEME 6
Figure imgf000047_0001
Oxazolyl piperazine intermediates such as 40 are available via the route shown in Scheme 7 below. Alkylation of piperazine 25 with bromo acid 36 in the presence of silver triflate in an inert solvent such as THF, according to methods detailed in J. Org. Chem. 1995, 60: 4013-4016, provides 37. Amide coupling of amine 38 to acid 37 to provide 39 can be carried out by any of the methods described above including the EDC /HOBT method. Amines such as 38 are prepared via chemistry described in Org. Synth. 1986, 64: 19-26 and Tetrahedron Letters 1999, 40: 6739-6743. Oxazole formation is accomplished by the action of a strong acid such as sulfuric acid on 39 in an inert solvent at elevated temperature, or as described in J. Med. Chem. 1996, 39: 2753-2763, to give intermediate 40. Again, intermediates such as these can be transformed into compounds of the instant invention via synthetic routes shown in Schemes 1,2, and 3. SCHEME 7
Figure imgf000048_0001
Oxazolyl piperazine intermediate 40 can also be obtained via the route shown in Scheme 8 below. Treatment of piperazine amide 41 with acetone at an elevated temperature will afford the acetonide which can be treated with acid HL (wherein L" is a non-nucleophilic anion such as OTf-) to afford the acid salt 42. Reaction of 42 with a dimethoxyalkane (e.g., 2,2-dimethoxypropane) will provide iminium salt 43, which can be coupled with a suitably metallated oxazole to provide coupled product 44. Suitable metallated oxazoles include those obtained by metallating the oxaole with an alkyllithium (e.g., n-butyllithium) or an alkylmagnesium halide (e.g., isopropylmagnesium bromide) in an inert solvent (e.g., an ether such as THF). Oxazolyl piperazine intermediate 40 can be obtained from 44 by treating 44 with acid (e.g., HCI, TFA, or an arylsulfonic acid). SCHEME 8
Figure imgf000049_0001
The following examples serve only to illustrate the invention and its practice. The examples are not to be construed as limitations on the scope or spirit of the invention.
EXAMPLE 1 (αR,γlS,25)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-N-[(31S,4S)-2,3-dihydro-3- hydroxy-3H- 1 -benzopyran-4-yl] -γ-hydroxy-α- [ [4-(3 -pyridinyl)phenyl]methyl] -2- [ [(2,2,2-trifluoroethyl)amino]carbonyl]- 1 -piperazinepentanamide
Figure imgf000050_0001
Step A
Figure imgf000050_0002
To a solution of 4-chromanone (10 g, 67.49 mmol) in 400 mL dichloromethane at 0 °C was added bromine (4.45 mL, 86.39 mmol) dropwise slowly. The reaction was monitored by TLC. After half an hour the reaction mixture was diluted with methylene chloride (100 mL) and was washed with water (300 mL). The organic layer was dried over anhydrous sodium sulfate and concentrated. The resulting product was dissolved in HOAc (100 mL) and sodium sulfite (8 g) was added. The reaction mixture was st red at room temperature and reaction progress was monitored by TLC. After 48 hours the reaction mixture was poured into water and the product was extracted with methylene chloride. The organic layer was dried over anhydrous sodium sulfate and concentrated in vacuo to give the titled compound as a white solid. lH NMR (CDCI3, 400 MHz): 7.93 (d, /= 8.8 Hz, IH), 7.54 (t, IH),
7.08 (t, IH), 7.02 (d, J = 8.0 Hz, IH), 4.63 (m, 4H)
Step B
Figure imgf000050_0003
To a solution of 3-bromo-4-chromanone (2 g, 8.81 mmol) in methanol (20 mL) was added sodium borohydride (0.4 g, 10.57 mmol). The reaction was stined at room temperature and monitored by TLC. After 2 hours the solvent was removed in vacuo and then diluted with ethyl acetate (50 mL). The resulting solution was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the titled compound as a white solid. lH NMR (CDCI3, 300 MHz): 7.32 (d, /= 7.2 Hz, IH), 7.23 (t, IH), 6.96 (t, IH), 6.84 (d, J =
9.0 Hz, IH), 4.82 (m, IH), 4.54 (m, IH), 4.38 (m, 2H).
Figure imgf000051_0001
To a solution of 3-bromo-4-chromanol (2 g, 8.72 mmol) in acetonitrile (20 mL) was added concentrated sulfuric acid (1 mL, 17.47 mmol). The reaction mixture was stirred at 45 °C - 50 °C for 18 hours. The solvent was removed in vacuo. Then water ( 10 mL) was added. The reaction mixture was heated to reflux. After 5 hours the reaction mixture was cooled to room temperature. The pH of the reaction mixture was adjusted to 12-13 by dropwise addition of aqueous 50% sodium hydroxide. The product was extracted with tetrahydrofuran three times. The organic layer were combined and dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the title compound as a white solid. lH NMR (CDCI3,
300 MHz): 7.29 (d, J = 7.8 Hz, IH), 7.16 (t, IH), 6.93, (t, IH), 6.83 (d, J = 8.4 Hz, IH), 4.12 (m, IH), 3.99 (m, 2H), 3.84 (m, IH).
Step D
To a suspension of the racemic 4-amino-3-chromanol in ethanol (35 mL per gram of 4-amino-3-chromanol) was added 1.0 equivalent of (S)-(+) mandelic acid. The suspension was heated to 70 °C until forming a homogeneous solution. The solution was cooled to room temperature and white crystal was formed. After filtering the white crystal was dissolved in 3 N aqueous sodium hydroxide solution and the resolved product was extracted with ethyl acetate three times. The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the titled compound as a white solid. The purity of the compound was verified by chiral HPLC with Crownpak CR+ column eluted with pH 1.0 perchloric acid solution. lH NMR (CDCI3, 300 MHz): 7.29 (d, J = 7.8 Hz, IH), 7.16 (t, IH),
6.93, (t, IH), 6.83 (d, J = 8.4 Hz, IH), 4.12 (m, IH), 3.99 (m, 2H), 3.84 (m, IH).
Figure imgf000052_0001
To a solution of 4-bromo-3-phenyl propionic acid (7.0g, 30.55 mmol) and amino chromanol from Step D (5.04 g, 30.33 mmol) in 300 mL of 1:1 NN- Dimethyl formamide and dichloromethane was added HOBt (5.37g, 39.72 mmol), NN-Diisopropyl ethyl amine (6.38 mL, 36.66 mmol) and EDC (7.6 g, 39.72 mmol). The resulting solution was stirred at room temperature for 4 hours. The reaction mixture was diluted with dichloromethane (50 mL) and washed with IN HCI, sat. ΝaHCO3 (2X). A white precipitate crashed out. This material was extracted with 10% methanol-dichloromethane (2X100 mL). The combined organic extracts were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the title compound as a white solid.
Figure imgf000053_0001
To a solution of the intermediate from Step E (11.49g, 30.55 mmol) in NN-Dimethyl formamide and dichloromethane (500 mL) was added 2-methoxy propene (14.7 mL, 153 mmol) followed by camphor sulfonic acid (1.42 g, 6.11 mmol). The reaction was heated at 40° C for 6 hours. The reaction mixture was cooled to room temperature and washed with IN ΝaOH (2X), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography using 20% ethyl acetate-hexanes then 40% ethyl acetate-hexanes to give the desired compound as an off white solid.
Figure imgf000053_0002
To a solution of 3-bromopyridine in anhydrous THF (15 mL) cooled to 0° C under a nitrogen atmosphere was added drop wise a solution of isopropyl magnesium chloride. The reaction mixture was stirred at room temperature for 45 minutes. A solution of trimethyl tin chloride in THF (15 mL) was added. The reaction mixture was stirred for 45 minutes at room temperature. The reaction was quenched with saturated ammonium chloride solution and the resulting bi-phasic mixture extracted with ethyl acetate (3X). The organic layers were washed with brine, dried over anhydrous sodium sulfate filtered and concentrated in vacuo to give a light brown oil. This material was purified by flash chromatography using 20% ethyl acetate-hexanes the 40% ethyl acetate-hexanes to give the desired compound as a colorless oil. Step H
Figure imgf000054_0001
To a solution of the intermediate obtained from Step F (3.94 g, 9.46 mmol) and the intermediate obtained from Step G (2.3 g, 9.46 mmol) in NN- Dimethyl formamide (50 mL) was added Pd(PPh3)4 (110 mg, 0.09 mmol) and the reaction heated to 100° C for two hours. The reaction mixture was cooled to room temperature and quenched with saturated potassium fluoride solution (100 mL). After stirring vigorously for one hour the reaction mixture was filtered through celite and washed with ethyl acetate (100 mL). The aqueous layer was extracted with ethyl acetate (3X). The organic layer was washed with brine, dried over anhydrous sodium sulfate filtered and concentrated in vacuo. The residue was purified by flash chromatography using 40% ethyl acetate then 100% ethyl acetate and finally with 5% 2.0M ΝH3-MeOH-ethyl acetate to give the desired compound as a white solid.
Figure imgf000054_0002
To a solution of the intermediate from Step H (2.38g, 5.74 mmol) and allyl bromide (547 μL, 6.32 mmol) in THF (100 mL) cooled to -20° C was added drop-wise a solution of LHMDS (6.32 mL, 1.0 M in THF). The reaction mixture was stirred at —20° C for 45 minutes and quenched with saturated NaHCO solution. The resulting bi-phasic mixture was extracted with ethyl acetate (2X), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography with 60% ethyl acetate-hexanes to give the desired compound as a white solid.
Figure imgf000055_0001
To a solution of the intermediate obtained from Step I (1.9g, 4.18 mmol) in ethyl acetate (80 mL) was added NaHCO3 solution (80 mL, 0.5 M) and the reaction cooled to 0°C. N-iodo-succinimide (1.975g, 8.77 mmol) was added and the reaction stined at room temperature for 18 hours. The reaction mixture was diluted with ethyl acetate. The organic layer was washed with a saturated solution of sodium thiosulfate (2X), water (IX), brine (IX) and dried over anhydrous sodium sulfate. The reaction mixture was filtered and concentrated in vacuo to give the desired compound as a light brown foam.
Figure imgf000055_0002
To a solution of the intermediate from Step J (2.5g, 4.18 mmol) in ethyl acetate (80 mL) was added a solution of sodium methoxide (1.43 mL, 6.27 mmol). After 15 minutes the reaction mixture was quenched with saturated with saturated NaHCO3 solution and extracted with ethyl acetate(3X). The organic layer was washed with brine, dried over anhydrous sodium sulfate filtered and concentrated in vacuo. The residue was purified by flash chromatography with 60% ethyl acetate- hexanes to give the desired compound as an off-white solid.
Figure imgf000056_0001
To a solution of l,4-piperazine-2-(5)-carboxylic acid [bis(+)-CSA salt (30.0 g, 50.0 mmol) in 600 mL THF was added IN aqueous NaOH until the resulting solution was pH 9 (150 mL). The solution was cooled to 0 oc, and BOC-ON (12.3 g, 50.0) was added. The resulting solution was warmed to ambient temperature over 5 hours, then cooled again to 0 °C. Allyl chloroformate (5.31 mL, 50.0 mmol) was added via syringe, followed by an additional 60 mL of IN aqueous NaOH. The solution was warmed to ambient temperature overnight, then concentrated to minimum volume by rotary evaporator. The resulting mixture was acidified to pH 1 with IN aqueous HCI, and extracted with ethyl acetate (400 mL x 2). The organic layers were washed with brine (200 mL) dried (MgSO4) and concentrated in vacuo, affording 23.7 g of a yellow oil. This material was dissolved in 750 mL of dichloromethane, followed by the addition of triethylamine (35.0 mL, 250 mmol), trifluoroethylamine (9.95 mL, 125 mmol), HO AT (10.2 g, 75.0 mmol), and EDC
(14.4 g, 75.0 mmol). After 22 hours at ambient temperature the reaction mixture was quenched by the addition of saturated aqueous NaHCO3 (500 mL). The organic layer was washed with an additional 500 mL of saturated aqueous NaHCO3, then IN aqueous NaHSO4 (500 mL), and additional saturated aqueous NaHCO3 (500 mL). The organic layer was dried (Na2SO4) and concentrated in vacuo. Purification by flash chromatography (40% ethyl acetate in hexane) afforded the title compound as a white solid. lH NMR (CDCI3, 400 MHz) 5.95 (m, IH), 5.35 (d, IH), 5.28 (d, IH),
4.75 (s, IH), 4.68 (d, IH), 4.53 (d, IH), 3.90 (m, 3H), 3.20 (dd, IH), 3.00 (m, IH), 1.45 (s, 9H).
Figure imgf000057_0001
To a solution of the intermediate obtained from Step L (following the removal of the Boc protecting group with trifluoro acetic acid) (890 mg, 3 mmol) in 1,2-dichloroethane (13 mL) was added 5-(4-chlorophenyl)-furan-aldehyde (623 mg, 3 mmol) and sodium triacetoxy borohydride (893 mg, 4.21 mmol). The reaction was quenched after one hour with saturated NaHCO3 solution. The aqueous layer was extracted with dichloromethane(3X). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography with 50% ethyl acetate-hexanes to give the desired compound as a solid.
Figure imgf000057_0002
To a stined solution of the intermediate obtained from Step M (1.05 g,
2.16 mmol) in anhydrous THF (15 mL) was added 1,3-Dimethyl barbituric acid (405 mg, 2.59 mmol) and Pd(PPh3) (250 mg, 0.22 mmol). After 45 minutes the reaction mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with IN HCI (6X15 mL). To the combined aqueous layers was added solid sodium carbonate until the pH=10. The aqueous layer was extracted with 5% methanol- dichloromethane (4X50 mL). The organic layer was dried over anhydrous sodium sulfate filtered and concentrated in vacuo to give the desired compound as a white solid.
Figure imgf000058_0001
To a solution of the intermediate obtained from Step K (65 mg, 0.13mmol) in 2-propanol (4 mL) was added the intermediate from Step N (50 mg, 0.12 mmol) and the resulting mixture heated to 85° C for 18 hours. The reaction mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by flash chromatography using ethyl acetate then 5% 2.0M ammonia- methanol-ethyl acetate to give the title compound as an yellow oil.
Step P
(αR,γ5,2S)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-N-[(3S,4S)-2,3-dihydro-3- hydroxy-3H-l-benzopyran-4-yl]-γ-hydroxy-α-[[4-(3-pyridinyl)phenyl]methyl]-2-
[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
To a solution of the intermediate from Step O (55 mg, 0.063 mmol) in anhydrous methanol (3 mL) was added a solution of ΗC1 in ether (1 mL, l.OM solution). After 1 hour the reaction mixture was concentrated in vacuo. The residue was partitioned between IN ΝaOΗ (5 mL) and ethyl acetate. The aqueous layer was extracted with ethyl acetate (3X). The organic layer was dried over anhydrous sodium sulfate filtered and concentrated in vacuo. The residue was purified by flash chromatography using 5% methanol-ethyl acetate to give the title compound as a white solid.
1H ΝMR(CD3OD, 500 MHz): 8.77 (bs, IH), 8.5 (d, 7=4.6 Hz, IH), 8.08 (dd, 7=1.8, 8.0 Hz, IH), 7.65 (d, 7=8.7 Hz, 2H), 7.58 (d, 7=8.2 Hz, 2H), 7.52 (dd, 7=5.0, 7.8 Hz, IH), 7.4 (m, 5H), 7.15 (d, 7=7.6 Hz, IH), 7.08 (t, 7=8.2 Hz, IH), 6.84 (t, 7=7.6 Hz, IH), 6.74 (m, 2H), 6.4 (d, 7=3.2 Hz, IH), 5.19 (d, 7=4.1 Hz, IH), 3.9-4.25 (m, 4H), 3.8 (m, 3H), 3.6 (s, IH), 3.2 (m, 2H), 3.0 (m, 3H), 2.83 (dd, 7=6.6, 13.5 Hz, IH), 2.75 (bd, 7=11.9 Hz, IH), 2.6 (t, 7=8.4 Hz, IH), 2.54 (t, 7=8.7 Hz, IH), 2.45 (m, 2H), 2.4 (m, 2H), 2.12 (t, 7=11.2 Hz, IH), 1.47 (m, IH). LC-MS (M++l) (El) 832.2.
EXAMPLE 2 (α5,γ5,2S)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-N-[(3S,4,S)-2,3-dihydro- 3-hydroxy-3H-l-benzopyran-4-yl]-γ-hydroxy-α-[5-(3-pyridinyl)-2-furanyl]lmethyl]- 2-[[(2,2,2-trifluoroethyl)arnino]carbonyl]-l-piperazinepentanamide
Figure imgf000059_0001
To a solution of 5-bromo-furaldehyde (2.23 g, 12.74 mmol) and the intermediate from Example 1 Step G (3.7g, 15.3 mmol) in toluene (50 mL) was added Pd(PPh3)4 and the resulting mixture heated to reflux for 18 hours. The reaction mixture was cooled to room temperature and quenched with saturated potassium fluoride solution (100 mL). After stirring vigorously for one hour the reaction mixture was filtered through celite and washed with ethyl acetate (100 mL). The aqueous layer was extracted with ethyl acetate (3X). The organic layer was washed with brine, dried over anhydrous sodium sulfate filtered and concentrated in vacuo. The residue was purified by flash chromatography using 60% ethyl acetate then 80% ethyl acetate-hexanes to give the desired compound as an off-white solid.
Figure imgf000060_0001
To a solution of the intermediate from Step A (2.14 g, 12.35 mmol) and malonic acid (2.83 g, 27.18 mmol) in pyridine (12 mL) was added piperidine (95 μL, 0.95 mmol) and the reaction mixture heated to reflux for 4 hours. The reaction mixture was cooled to room temperature whereupon a yellow solid precipitated out. This material was suspended in dichloromethane (100 mL) and filtered. The residue was dried under vacuum to give the title compound as a yellow powder.
Figure imgf000060_0002
To a solution of the intermediate from Step B (2.3 g) in ethanol (60 mL) was added N,N-Diisopropyl ethyl amine (4.0 mL) followed by Pd/C. The reaction mixture was stined under a hydrogen balloon for 18 hours. The reaction mixture was filtered through celite and concentrated in vacuo to give the title compound as a yellow solid.
Ste D
Figure imgf000060_0003
To a solution of the intermediate from Step C (2.2g, 10.12 mmol) and amino chromanol (1.67 g, 10.12 mmol) in 100 mL of 1:1 N,N-Dimethyl formamide and dichloromethane was added HOBt (1.77 g, 13.15 mmol), NN-Diisopropyl ethyl amine (2.3 mL, 13.15 mmol) and EDC (2.51 g, 13.15 mmol). The resulting solution was stined at room temperature for 4 hours. The reaction mixture was diluted with dichloromethane (50 mL) and washed with IN HCI, sat. NaHCO3 (2X). A white precipitate crashed out. The organic layer was concentrated in vacuo and azeotropically dried with toluene (2X) to give the desired compound as a white solid.
Figure imgf000061_0001
To a solution of the intermediate from Step D (3.7 g, 10.12 mmol) in NN-Dimethyl formamide and dichloromethane (400 mL) was added 2-methoxy propene (4.86 mL, 50 mmol) followed by camphor sulfonic acid (2.82 g, 12.14 mmol). The reaction was stined at room temperature for 18 hours. The reaction mixture was quenched with IN ΝaOH (200 mL) and extracted with dichloromethane (2X), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography using 100% ethyl acetate to give the desired compound.
Figure imgf000061_0002
To a solution of the intermediate from Step E (3.39 g, 8.39 mmol) and allyl bromide (799 μL, 9.23 mmol) in THF (100 mL) cooled to -20° C was added drop wise a solution of LHMDS (9.23 mL, 1.0 M in THF). The reaction mixture was stined at -20° C for 45 minutes and quenched with saturated ΝaHCO3 solution. The resulting bi-phasic mixture was extracted with ethyl acetate (2X), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography with 50% ethyl acetate-hexanes to give the desired compound as an off-white foam.
Step G
Figure imgf000062_0001
To a solution of the intermediate obtained from Step F(3.1 g, 6.97 mmol) in ethyl acetate (50 mL) was added NaHCO3 solution (50 mL, 0.5 M) and the reaction cooled to 0 oC. N-iodo-succinimide (3.3 g, 14.64 mmol) was added and the reaction stined at room temperature for 18 hours. The reaction mixture was diluted with ethyl acetate. The organic layer was washed with a a saturated solution of sodium thiosulfate (2X), water (IX), brine (IX) and dried over anhydrous sodium sulfate. The reaction mixture was filtered and concentrated in vacuo to give the desired compound as a brown foam.
Figure imgf000062_0002
To a solution of the intermediate from Step G (4.1g, 6.97 mmol) in ethyl acetate (75 mL) was added a solution of sodium methoxide (2.39 mL). After 30 minutes the reaction mixture was quenched with saturated with saturated ΝaHCO3 solution and extracted with ethyl acetate(3X). The organic layer was washed with brine, dried over anhydrous sodium sulfate filtered and concentrated in vacuo. The residue was purified by flash chromatography with 60% ethyl acetate-hexanes then 80% ethyl acetate-hexanes to give the title compound as a yellow solid.
Figure imgf000063_0001
To a solution of the intermediate obtained from Step H (63 mg, 0.13mmol) in 2-propanol (4 mL) was added the intermediate from Example 1 Step N (50 mg, 0.12 mmol) and the resulting mixture heated to 85° C for 18 hours. The reaction mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by flash chromatography using ethyl acetate then 5% methanol- ethyl acetate to give the desired compound as an yellow solid.
Step J
1S,γ5,,25)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-N-[(3,S,45)-2,3-dihydro-
3 -hydroxy-3H- 1 -benzopyran-4-yl] -γ-hydroxy-α- [5-(3 -pyridinyl)-2-f uranyl] lmethyl] -
2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazmepentanamide
To a solution of the intermediate from Step I (37mg, 0.043 mmol) in anhydrous methanol (2 mL) was added a solution of ΗC1 in ether (1 mL, l.OM solution). After 1 hour the reaction mixture was concentrated in vacuo. The residue was partitioned between IN ΝaOΗ (5 mL) and ethyl acetate. The aqueous layer was extracted with ethyl acetate (3X). The organic layer was dried over anhydrous sodium sulfate filtered and concentrated in vacuo. The residue was purified by flash chromatography using 5% methanol-ethyl acetate and then 5% 2.0 M ammonia in methanol-ethyl acetate to give the title compound as a white solid. 1H NMR (CD3OD, 500 MHz): 8.83 (bs, IH), 8.37 (bd, 7=4.3 Hz, IH), 8.07 (d, 7=7.8 Hz, IH), 7.65 (d, 7=8.5 Hz, 2H), 7.43 (dd, 7=4.8, 7.7 Hz, IH), 7.37 (d, 7=8.5 Hz, 2H), 7.18 (d, 7=7.8 Hz, IH), 7.11 (t, 7=7.5 Hz, IH), 6.84 (m, 2H), 6.74 (m, 2H), 6.39 (d, 7=3.3 Hz, IH), 6.31 (d, 7=3.2 Hz, IH), 5.25 (d, 7=4.1 Hz, IH), 4.1 (bs, 2H), 3.9 (m, 3H), 3.65-3.85 (m, 3H), 3.6 (bs, 2H), 3.25 (m, IH), 3.0-3.2 (m, 3H), 2.9 (dd, 7=6.7, 14.9 Hz, IH), 2.82 (d, 7=9.9 Hz, IH), 2.73 (m, IH), 2.58 (t, 7=11.0 Hz, IH), 2.4 (m, 3H), 2.15 (t, 7=11.5 Hz, IH), 1.5 (m, IH). LC-MS (M^+l) (El) 822.2.
EXAMPLE 3 (α5,γS,2S)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(3.S,4S)-2,3- dihydro-3 -hydroxy-3H- 1 -benzopyran-4-yl] -γ-hydroxy-α- [5-(3 -pyridinyl)-2- thiazolyl]methyl]-2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
Figure imgf000064_0001
Step A
Figure imgf000064_0002
To a solution of 2-formyl thiazole (6.0g, 53 mmol) in benzene placed in a round bottom flask (100 mL) was added ethylene glycol (7.4 mL, 132.5 mmol) and -toluene sulfonic acid (131 mg, 0.69 mmol). The flask was fitted with a Dean- Stark trap and the solution heated to 120° C for one hour. The reaction was cooled to room temperature and concentrated in vacuo. The residue was diluted with ethyl acetate washed with sat. NaHCO3 and dried over anhydrous sodium sulfate. The reaction mixture was filtered and concentrated in vacuo. The residue was purified by flash chromatography using 15% ethyl acetate-hexanes then 40% ethyl acetate- hexanes to give the desired compound as a light brown oil.
Figure imgf000065_0001
To a solution of the intermediate obtained from Step A (5.0g, 31.8 mmol) in THF (100 mL) cooled to -78° C was added a solution of n-Butyllithium (24 mL, 38.17 mmol). After 10 min a solution of trimethyl tin chloride (7.6 g, 38.17 mmol) in 10 mL of THF was added. The reaction was warmed to 0° C and quenched with saturated NH4CI solution. The resulting bi-phasic layer was extracted with ethyl acetate (3x), washed with sat. NaCl and dried over anhydrous sodium sulfate. The reaction mixture was filtered and concentrated in vacuo. The residue was purified by flash chromatography using 20% ethyl acetate-hexanes to give the desired compound as a light brown oil.
Figure imgf000065_0002
To a solution of the intermediate obtained from Step B (6.0g, 22.5 mmol) in toluene (100 mL) was added 3-bromopyridine (2.16 mL, 22.5 mmol) and Pd(PPh3)4 (1.08 g, 0.937 mmol) and the reaction mixture heated to 120° C for 18 hours. The reaction mixture was cooled to room-temperature and quenched with saturated potassium fluoride solution. The resulting mixture was stined vigorously for 1 hour. The reaction mixture was filtered through celite. The aqueous layer was extracted with ethyl acetate (2X). The combined organic layers were washed with water (2X) and brine (IX) and dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography using 60% ethyl acetate-hexanes then 80% ethyl acetate-hexanes to give the desired compound as a light yellow solid.
Figure imgf000066_0001
To a solution of the intermediate obtained from Step C (3.2 g, 13.65 mmol) in THF (100 mL) was added HC1(47.8 mL, l.OM) and the resulting mixture refluxed for 4 hours. The reaction mixture was cooled to room temperature and washed with saturated solution of sodium carbonate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the title compound.
Figure imgf000066_0002
To a solution of the intermediate from Step D(1.84g, 9.68 mmol) and malonic acid(2.21 g, 21.3 mmol) in pyridine (12 mL) was added piperidine (74 μL, 0.74 mmol) and the resulting mixture refluxed for 18 hours. The reaction mixture was cooled to room temperature and concentrated in vacuo. The residue was azeotropically dried with toluene (3X) to give a dark brown solid. Thia material was suspended in dichloromethane and filtered to give the desired compound as a dark yellow solid.
Figure imgf000067_0001
To a solution of the intermediate from Step E (2.0 g) in ethanol (30 mL) was added NN-Diisopropyl ethyl amine (6.0 mL) followed by Pd C. The reaction mixture was stined under a hydrogen balloon for 18 hours. The reaction mixture was filtered through celite washed with methanol. The filtrate was concentrated in vacuo and azeotropically dried with toluene (3X) to give the desired compound as a light yellow solid.
Figure imgf000067_0002
To a solution of the intermediate from Step F (2.0g, 8.53 mmol) and amino chromanol from example 1 Step E (1.41 g, 8.53 mmol) in 100 mL of 1:1 NN- Dimethyl formamide and dichloromethane was added HOBt(1.5 g, 11.1 mmol), NN- Diisopropyl ethyl amine (1.93 mL, 11.1 mmol) and EDC (2.12 g, 11.1 mmol). The resulting solution was stined at room temperature for 4 hours. The reaction mixture was diluted with dichloromethane (50 mL) and washed with IN HCI, saturated ΝaHCO3 (2X). A white precipitate crashed out of the organic layer. The organic layer was concentrated in vacuo and azeotropically dried with toluene (2X) to give the desired compound as a white solid.
Step H
Figure imgf000067_0003
To a solution of the intermediate from Step G (2.8 g, 7.34 mmol) in N,N-Dimethyl formamide and dichloromethane (300 mL) was added 2-methoxy propene (3.52 mL, 37 mmol) followed by camphor sulfonic acid (2.1 g, 8.8mmol). The reaction was stined at room temperature for 18 hours. The reaction mixture was quenched with IN ΝaOH (200 mL) and extracted with dichloromethane (2X), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography using 100% ethyl acetate then 3% methanol-ethyl acetate to give the desired compound as a light brown solid.
Figure imgf000068_0001
To a solution of the intermediate from Step H (2.74 g, 6.5 mmol) and allyl bromide (675 μL, 7.8 mmol) in THF (100 mL) cooled to -60° C was added drop- wise a solution of LHMDS (7.8 mL, 1.0 M in THF). The reaction mixture was warmed slowly to -20° C and stined for 45 minutes. The reaction was quenched with saturated ΝaHCO3 solution and the resulting bi-phasic mixture was extracted with ethyl acetate (2X), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography with 80% ethyl acetate- hexanes to give the desired compound as an off-white foam.
To a solution of the intermediate obtained from Step 1(2.29 g, 4.96 mmol) in ethyl acetate (50 mL) was added NaHCO3 solution (50 mL, 0.5 M) and the reaction cooled to 0o C. N-iodo-succinimide (2.34 g, 10.42 mmol) was added and the reaction stined at room temperature for 18 hours. The reaction mixture was diluted with ethyl acetate. The organic layer was washed with a saturated solution of sodium thiosulfate (2X), water (IX), brine (IX) and dried over anhydrous sodium sulfate. The reaction mixture was filtered and concentrated in vacuo to give the title compound as a brown foam.
Step K
Figure imgf000069_0002
To a solution of the intermediate from Step J (2.38 g, 3.93 mmol) in ethyl acetate (50 mL) was added a solution of sodium methoxide (1.34 mL, 5.9 mmol, 25% wt). After 30 minutes the reaction mixture was quenched with saturated with saturated ΝaHCO3 solution and extracted with ethyl acetate(3X). The organic layer was washed with brine, dried over anhydrous sodium sulfate filtered and concentrated in vacuo. The residue was purified by flash chromatography with 60% ethyl acetate- hexanes to give the title compound as a brown solid. Step L
Figure imgf000070_0001
To a solution of the piperazine intermediate from Example 1, Step M
(1 g, 3.39 mmol following removal of the BOC group with trifluoroacetic acid) and silver trifluoromethane sulfonate (725 mg, 2.82 mmol) in THF (5 mL) was added triethylamine. While stirring, a solution of 2-bromo-2-methylpropionic acid (472 mg, 2.82 mmol) in 5 mL of THF was introduced slowly (within 1.5 hours) through a syringe pump. The reaction mixture was stined at room temperature for 2 hours after the additions. Then, it was filtered. The filtrate was concentrated and purified by flash column chromatography on silica gel with 10/1 ethyl acetate / methanol as eluant to give the title compound as a white solid. lH NMR (CDC13, 400 MHz): δ 9.02 (broad s, 1 H), 6.98 (broad s, 1 H), 5.90-6.00 (m, 1 H), 5.24-5.28 (m, 2 H), 5.65 ( broad s, 1 H), 4.16 ( broad s, 1 H), 2.90-2.93 (m, 1 H), 2.55-2.57 (m, 1 H), 2.40-2.41 (m, 1 H), 1.59-1.61 (m,lH).
Step M:
Figure imgf000070_0002
To a stirring solution of hexamethylenetetramine (12.0g; 85.7mmol) in
CH2C12 (400mL) was added portionwise α-bromo-p-chloroacetophenone (20.0g;
85.7mmol). After 30 minutes the precipitate was filtered and then suspended in EtOH
(680mL). Cone. HCI was added (45mL) and the suspension stined at 90 °C 1.5hrs. Dissolution occuned followed by solution turning pale yellow and a new precipitate formed. This solid was filtered, washed with EtOH, and dried under vacuum to provide the desired compound. lH NMR (500 MHz, CD3OD): δ 4.60 (s, 2H), 7.61 (d, J=8.7 Hz, 2H), 8.04 (d, J=8.4 Hz, 2H).
Figure imgf000071_0001
To a stined solution of carboxylic acid from Step L (4.96g; 13mmol) in dry NMP (75mL) under nitrogen was added DIEA (9.06 mL; 52 mmol). After cooling the solution to 0 °C, the following solids were added, allowing one to dissolve before adding the next: HOBt (3.95g; 29.3mmol), intermediate from Step M above (3.22g; 15.6mmol), and HBTU (7.4g; 19.5mmol). The reaction was allowed to reach ambient temperature, and the next morning the mixture was poured into EtOAc and washed with saturated NaHCO3, water, brine, 3x dilute NaHCO3, and brine. After drying (MgSO4), filtration, and removal of solvent in vacuo, the residue was purified by flash column chromatography (50% EtOAc/hexane). Residual NMP which remained was removed by dissolution in EtOAc followed by washing with water (2x), brine, drying (MgSO ), filtration, and solvent removal in vacuo. The residue resulting after workup was purified by Biotage column chromatography (40M; 45% EtOAc/hexane) to provide the desired product. lH NMR (500 MHz, CDC13): δ 1.28 (s, 3H), 1.30 (s, 3H), 2.35 (apparent td, J=3.0, 11.6 Hz, IH), 2.45 (apparent dd, J=3.8, 11.8 Hz, IH), 2.86 (d, J=10.6 Hz, IH), 3.22-3.33 (broad, IH), 3.67 (d, J=11.7 Hz, IH), 3.74-4.30 (broad, 3H), 4.51 (d, J=18.6 Hz, IH), 4.70 (d, J=4.5 Hz, 2H), 4.83 (s, IH), 4.89 (1/2ABX, J=6.8, 18.7 Hz, IH), 5.30 (d, J=10.3 Hz, IH), 5.36 (d, J=17.1 Hz, IH), 5.92-6.02 (br s, IH), 6.60-6.72 (br s, IH), 7.49 (d, J=8.5 Hz, 2H), 7.93 (d, J=8.5 Hz, 2H), 8.20-8.30 (br s, IH) ; electrospray ionization mass spectrum: m/e 533.3 (MH+ calcd for C23H29CIF3N4O5, 533.2). Step O
Figure imgf000072_0001
To the amide obtained in Step N (139mg; 0.26mmol) was added
H2SO4 (2.75mL). Partial dissolution occuned. The reaction vessel was purged with nitrogen, P2O5 (HOmg; 0.39mmol) was added, and the mixture was stined at 65 °C for 20 minutes. After cooling to 0 °C, ice chips were added and the reaction adjusted to pH 9 with the addition of 50% NELOH, then cone. NH OH. Extraction with CHC13 (3x) was followed by drying (Na2SO ), filtration and removal of the solvent in vacuo. Flash column chromatography (93:5:2 EtOAc: MeOH: TEA) provided the desired product. lH NMR (500 MHz, CDC13): δ 1.60 (s, 3H), 1.61 (s, 3H), 1.75-1.88 (broad s, IH), 2.56-2.60 (complex m, 2H), 2.77-2.81 (m, IH), 2.86-2.98 (complex m, 3H), 3.50-3.52 (m, IH), 3.93-4.00 (m, 2H), 7.26 (s, IH), 7.42 (d, J=8.5 Hz, 2H), 7.58 (d, J=8.4 Hz, 2H), 8.10-8.18 (broad s, IH) ; electrospray ionization mass spectrum: m/e 431.3 (MH+ calcd for C19H23CIF3N4O2, 431.1).
Step P
Figure imgf000072_0002
To a solution of the intermediate from Step K (60 mg, 0.125 mmol) in 2-propanol (5 mL) was added the intermediate from Step O (250 mg, 0.54 mmol) and the resulting mixture heated at 85° C for 6 hours. The reaction mixture was concentrated in vacuo and purified by flash chromatography using 5% methanol-ethyl acetate. The product obtained was re-purified by reverse-phase HPLC using a
MetaChem 21X150mm Polaris C18-A5 micron column. The gradient used was 35% acetonitrile-water with 0.1% TFA to 60% acetonitrile-water. The product containing fractions were combined and concentrated in vacuo. The residue was dissolved in ethyl acetate and washed with saturated sodium carbonate solution to give the desired compound as a white solid.
Step Q
(αS,γ5,2S)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(35,45)-2,3- dihydro-2-hydroxy-3H- 1 -benzopyran-4-yl] -γ-hydroxy-α- [5-(3 -pyridinyl)-2- thiazolyl]methyl]-2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
To a solution of the intermediate from Step P (20 mg, 0.022 mmol) in anhydrous methanol (2 mL) was added a solution of ΗC1 in ether (1 mL, l.OM solution). After 1 hour the reaction mixture was concentrated in vacuo. The residue was partitioned between IN ΝaOΗ (5 mL) and ethyl acetate. The aqueous layer was extracted with ethyl acetate (3X). The organic layer was dried over anhydrous sodium sulfate filtered and concentrated in vacuo. The residue was purified by flash chromatography using 5% methanol-ethyl acetate and then 5% 2.0 M ammonia in methanol-ethyl acetate to give the title compound as a white solid. 1ΗΝMR (CD3OD, 500 MHz): 8.77 (bs, 7=1.4 Hz, IH), 8.5 (d, 7=4.3 Hz, IH), 8.05 (m, IH), 8.0 (s, IH), 7.68 (d, 7=8.7 Hz, 2H), 7.48 (dd, 7=4.8 Hz, IH), 7.43 (m, 3H), 7.18 (d, 7=7.6 Hz, IH), 7.12 (t, 7=8.2 Hz, IH), 6.84 (t, 7=7.3 Hz, IH), 6.76 (d, 7=7.6 Hz, IH), 5.23 (d, 7=4.1 Hz, IH), 4.15 (m, 2H), 3.9-4.0 (m, 2H), 3.8 (m, 2H), 3.41 (dd, 7=8.3 Hz, IH), 3.25 (m, IH), 3.18 (dd, 7=5.8 Hz, IH), 3.09 (m, IH), 3.0 (m, IH), 2.89 (bd, 7=9.4 Hz, IH), 2.79 (m, IH), 2.64 (t, 7=7.8 Hz, IH), 2.45 (m, 4H), 2.13 (t, 7=10.9 Hz, IH), 1.59 (s, 6H), 1.46 (m, IH). ). LC-MS (M++l) (El) 868.2. EXAMPLE 4 (α5,γ5,25)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-N-[(3S,4S)-2,3-dihydro-3- hydroxy-3H- 1 -benzopyran-4-yl] -α-(furo [3 ,2-c]pyridin-2-ylmethyl)-γ-hydroxy-2- [[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
Figure imgf000074_0001
A suspension of 4-pentynoic acid (9.52 g, 99.89 mmol) and the intermediate from Example 1 Step D (15 g, 90.81 mmol) in TΗF (900 mL) was added EDC (26.1 g, 136.2 mmol), ΗOBt (18.2 g, 136.2) and DTEA (31.5 mL, 181.6 mmol). The reaction mixture was stined at room temperature for 3 days. The solvent was removed in vacuo and the residue was dissolved in ethyl acetate. This organic solution was washed with water, saturated ΝaΗCO3, brine, then it was dried over Na^O^ filtered and concentrated. The resulting residue was dissolved in minimum amount of methylene chloride and hexane was added until white precipitate was formed. After filtration the title compound was obtained as a white solid.
Figure imgf000075_0001
To a solution of intermediate from Step A (3.5 g, 14.3 mmol) in methylene chloride (65 mL) at room temperature was added 2-methoxypropene (6.8 mL, 71.3 mmol) and camphorsulfonic acid (catalytic amt.). After 15 min the reaction was added to IN NaOH and extracted with methylene chloride three times. The combined organic layer was washed with brine, dried over Na2SO4, filtered and concentrated. The crude product was purified by flash column chromatography on silica gel with 3:2 ethyl acetate/hexane as eluant to give the title compound.
Figure imgf000075_0002
A suspension of intermediate from Step B (2.62 g, 9.2 mmol) and 4- hydroxy-3,5-diiodo-pyridine (3.51 g, 10.1 mmol) in pyridine (48 mL) was purged with nitrogen for 5 min, then Cu2O (1.7 g, 11.94 mmol) was added. The resulting suspension was purged with nitrogen for another 5 min. The reaction mixture was heated at 120 °C for 1.5 h. The resulting suspension was filtered through celite and concentrated. The crude product was purified by flash column chromatography on silica gel with ethyl acetate as eluant to give the title compound.
Figure imgf000076_0001
To a solution of intermediate from Step C (2.22 g, 4.4 mmol) in ethanol (40 mL) was added Pd(OH)2 (0.63 g, cat.) and DIEA (3.07 mL, 17.6 mmol). The suspension was stined under a hydrogen balloon at room temperature overnight. This reaction mixture was filtered through celite and concentrated. The crude product was purified by flash column chromatography on silica gel with ethyl acetate as eluant to afford the title compound.
Figure imgf000076_0002
A solution of intermediate from Step D (1.6 g, 4.2 mmol) in anhydrous THF was cooled to - 25 °C. To this solution, ally bromide (0.44 mL, 5.0 mmol) was added followed by addition of LHMDS in THF (1 M in THF, 5.0 mL, 5.0 mmol). After 1 h, the reaction mixture was poured into water and extracted with ethyl acetate three times. The combined organic layer was dried over NajSO,,, filtered and concentrated. The crude product was purified by flash column chromatography on silica gel with 3:1 ethyl acetate/hexane as eluant to afford the title compound. Step F
Figure imgf000077_0001
A solution of intermediate from Step E (1.1 g, 2.7 mmol) in ethyl acetate (25 mL) and 0.5 M NaHCO3 (25 mL) was cooled to 0 °C and NTS (1.53 g, 6.81 mmol) was added. After stirring from 0 °C to room temperature, the reaction mixture was poured into NajSOj solution and extracted with ethyl acetate three times. The combined organic layer was dried over Na,SO4, filtered and concentrated. The resulting residue was dissolved in ethyl acetate (47 mL) and NaOMe (2.52 mL, 25 % by wt in methanol) was added. The reaction mixture was stined at room temperature for 1.5 h, then it was poured into saturated NaHCO3 solution and extracted with ethyl acetate three times. The combined organic layer was dried over Na^O,,, filtered and concentrated. The crude product was purified by flash column chromatography on silica gel with 4: 1 ethyl acetate/ hexane as eluant to give the title compound.
Figure imgf000077_0002
A solution of the intermediate from Step F (80 mg, 0.184 mmol) and the intermediate from Example 1 Step N (111 mg, 0.276 mmol) in 2-propanol (2 ml) was heated to reflux overnight. The solvent was removed in vacuo. The crude reaction mixture was purified by flash column chromatography on silica gel with 70% ethyl acetate/acetone as eluant to give the titled compound as a white solid. LC-MS (M++1) (El) 836.4.
Step H
(αS,γ5,2S)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-N-[(35,45)-2,3-dihydro-3- hydroxy-3H- 1 -benzopyran-4-yl] -α-(furo [3 ,2-c]pyridin-2-ylmethyl)-γ-hydroxy-2- [[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
To a solution of the intermediate from Step G (75.8 mg, 0.091 mmol) in methanol (3.2 ml) was added 1 M ΗC1 in ethyl ether (1.4 ml, 1.4 mmol). The reaction mixture was stined at room temperature overnight. Then the reaction mixture was neutralized by 2 M ammonia in methanol. The solvent was removed in vacuo and the reaction mixture was diluted with methylene chloride and washed with 1 Ν ΝaOΗ. The aqueous layer was extracted with methylene chloride three times. The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by reverse phase ΗPLC. 1H ΝMR (CDC13, 500 MHz): 9.15 (bs, ΝH, IH), 8.81 (m, IH), 8.43 (m, IH), 7.58-7.56 (m, 2H), 7.38-7.37 (m, 3H), 7.16-7.13 (m, 2H), 6.84-6.80 (m, 2H), 6.61-6.58 (m, 3H), 6.34 (d, 7 = 3.4 Hz, IH), 5.29-5.27 (m, IH), 4.17-4.10 (m, 2H), 4.01-3.97 (m, 2H), 3.82-3.78 (m, IH), 3.72-3.48 (m, 3H), 3.34-3.25 (m, 2H), 3.19-3.14 (m, IH), 3.04- 2.94 (m, 3H), 2.72-2.63 (m, 3H), 2.55-2.37 (m, 3H), 1.92-1.86 (m, IH), 1.68-1.58 (m, IH). LC-MS (M++1) (ET) 796.3.
EXAMPLE 5 (2S)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-l-[(2,S,45)-4-(furo[2,3-c]pyridin-2- ylmethyl)-2-hydroxy-5-[[(35,45)-3-hydroxy-3,4-dihydro-2H-chromen-4-yl]amino]-5- oxopentyl]-N-(2,2,2-trifluoroethyl)-2-piperazinecarboxamide
Figure imgf000079_0001
To a stined solution of DTPA (1.28mL; 9.74mmol) in dry THF (26mL) at 0 °C was added dropwise n-BuLi (3.54mL; 8.85mmol). After 15 minutes, the solution was cooled to -78 °C and a solution of (5')-(+)-dihydro-5-(t- butyldimethylsilylhydroxymethyl)-2(3H)-furanone (2.04g; 8.85mmol) in dry TΗF (8mL) was added dropwise. After an additional 30 minutes, propargyl bromide (1.56mL; lO.όmmol) was added dropwise. After 45 minutes the reaction was quenched with 10% citric acid, poured into Et2O, and washed with Η2O (2x) and brine, dried (MgSO4), filtered, and solvents removed in vacuo. Purification by Biotage column chromatography (40M; 8% EtOAc/hexane) provided the desired compound.
Figure imgf000079_0002
To a stined solution of the intermediate from Step A above (903mg;
3.46mmol) in dry THF (21mL) was added HF-pyridine complex (1.2mL). The next morning, the reaction mixture was cooled to 0 °C and adjusted to pH 10 with NH4.OH/Η2O (2:1). Much solvent was removed in vacuo and the residue was poured into EtOAc and washed with saturated NaHCO3, 2x H2O and brine. Drying (MgSO ), filtration, removal of solvent in vacuo, and purification by Biotage column chromatography (40M; 50% EtOAc/hexane) provided the desired product.
Figure imgf000080_0001
To a stined solution of intermediate from Step B above (720mg; 4.67mmol) in dry CH2C12 (19mL) at 0 °C was added 2,6-lutidine (816uL; 7.0mmol) followed dropwise by trifluoromethanesulfonic anhydride (1.02mL; 6.07mmol).
After 1.75 hours, the reaction mixture was poured into 20mL ice/brine and stined 30 minutes. This mixture was poured into CH2C12 and washed with 2x H O and 2x brine. After drying (MgSO4), filtration, and removal of solvent in vacuo, the residue was purified by Biotage column chromatography (40M; 20% EtOAc/hexane) to provide the desired intermediate. lH NMR (500 MHz, CDC13): δ 2.08 (m, IH), 2.38- 2.44 (complex m, IH), 2.52-2.59 (complex m, IH), 2.67 (m, 2H), 2.94-2.98 (complex m, IH), 4.57 (1/2ABX, J=4.1, 11.2 Hz, IH), 4.71 (1/2ABX, J=3.0, 11.2 Hz, IH), 4.89 (m, IH).
Figure imgf000080_0002
To a stined solution of piperazine (lOOmg; 0.25mmol) from Step C above and triflate (72mg; 0.25mmol) from Step C above in dry iPrOH (1.25mL) was added DTEA (52uL; 0.30mmol). The next morning, the reaction mixture was poured into EtOAc and washed with saturated NaHCO3, water, brine, water, and brine. Drying (MgSO4), filtration, and removal of solvent in vacuo was followed by flash column chromatography (70% EtOAc/hexane) to provide the desired intermediate.
Figure imgf000081_0001
To a solution of acetylene (845mg; 1.9mmol) from Step D above and 4-iodo-3-hydroxypyridine (501mg; 2.23mmol) in dry pyridine (lOmL) was added Cu2O (323mg; 2.3mmol). After 35 minutes at 120 °C, the mixture was cooled to ambient temperature and filtered through celite, washing with EtOAc. The organics were washed with aqueous Na2SO , 2x water, brine, dried (Na2SO ), filtered, and the solvents removed in vacuo. Purification by flash column chromatography (gradient elution 3% to 5% MeOH/CH2Cl2) provided the desired intermediate
Figure imgf000081_0002
To a solution of the lactone (565mg; 1.04mmol) obtained from Step E above in dry dimethoxyethane (lOmL) cooled to 0 °C under nitrogen was added aqueous LiOH (1.04mL;1.0M). The flask was allowed to warm to ambient temperature and stined for 3.5hr. The reaction was azeotroped from MeCN, MeCN/benzene, and benzene, taking care to keep the water bath below 35 °C. This salt was further dried under vacuum, then dissolved in dry DMF (lOmL) and imidazole (1.41g; 21.0mmol) was added. This solution was cooled to 0 °C and TBSC1 was added (1.56g; 10.4mmol). The flask was allowed to warm to ambient temperature and stined overnight. The next morning the reaction was quenched with pH 7 phosphate buffer and extracted 3x EtOAc. After drying (Na2SO4), filtration, and removal of solvent in vacuo, the residue was dissolved in 3mL THF/water (2: 1). After lhr the mixture was azeotroped from MeCN, benzene, and MeCN to provide the carboxylic acid, which was used without further purification.
Figure imgf000082_0001
To a stined solution of acid (672 mg; .296mmol) from Step F in dry NMP (3mL) at 0 °C was added DIEA (155uL; 0.89mmol) followed by the following solids, allowing one to dissolve before adding the next: HOBt (90mg; 0.66mmol), amino chromanol obtained from Example 1 Step D (68mg; 0.41mmol), and HBTU (168mg; 0.44mmol). The reaction was allowed to reach ambient temperature and the next morning the mixture was poured into EtOAc and washed with saturated NaHCO3, water, brine, 3x dilute NaHCO3, and brine. After drying (Na2SO ), filtration, and removal of the solvent in vacuo, the residue was purified by flash chromatography (95% EtOAc/hexane) to provide the desired product.
Figure imgf000083_0001
The intermediate from the previous Step ( 48 mg) was stined in a mixture of 30% TFA in dichloromethane (1 mL) for 1.5 hours. The solution was then poured into EtOAc ( 40 mL) containing sat NaHCO3 solution. The aqueous layer was washed with water, brine, and then dried and filtered to give the desired product.
Figure imgf000083_0002
To a solution of the intermediate obtained from Step H (27mg, 0.037 mmol) in DMF (0.5 mL) was added 5-(4-chlorophenyl)-furan-aldehyde (10 mg, 0.048 mmol) and sodium triacetoxy borohydride (10.3 mg, 0.048 mmol). The reaction was quenched after one hour with saturated NaHCO3 solution. The aqueous layer was extracted with dichloromethane (3X). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography with 5% methanol-dichloromethane to give the title compound (17mg) . Step J
(25)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-l-[(2lS,45)-4-(furo[2,3-c]pyridin-2- ylmethyl)-2-hydroxy-5-[[(35,45)-3-hydiOxy-3,4-dihydiO-2H-chromen-4-yl]amino]-5- oxopentyl]-N-(2,2,2-trifluoroethyl)-2-piperazinecarboxamide
To a solution of the intermediate (17mg; 0.0187mmol) from Step I in dry TΗF (0.5mL) was added TBAF (56uL; 0.056mmol). The solution was stined at 55 °C for 2 hours. The reaction mixture was poured into EtOAc and washed with saturated ΝaΗCO3, water, and brine. Drying (Na2SO4), filtration, and removal of the solvent in vacuo followed by purification by MPLC (Lobar column; linear gradient 10% to 90% MeCN/H2O) provided the titled compound after lyophilization from MeCN/water (1:1). . 1H NMR (CD3OD, 500 MHz): 8.70 (s, IH), 8.26 (d, 7= 5.2 Hz, IH), 7.65-7.63 (m, 2H), 7.59 (d, 7= 5.5 Hz, IH), 7.37-7.36 (m, 2H), 7.17 (d, 7 = 7.6 Hz, IH), 7.12-7.09 (m, IH), 6.84-6.81 (m, IH), 6.76-6.73 (m, 3H), 6.39 (d, 7= 3.2 Hz, IH), 5.23 (d, 7 = 4.1 Hz, IH), 4.84-3.70 (m, 6H), 3.64 (s, 2H), 3.28-3.23 (m, 2H), 3.11-3.00 (m, 3H), 2.82-2.71(m, 2H), 2.57-2.35 (m, 5H), 2.16-2.11 (m, IH), 1.51- 1.46(m, IH). LC-MS (M++1) (El) 796.3.
EXAMPLE 6
(αS,γ5,2S)-4-[[5-(3-chlorophenyl)-2-furanyl]methyl]-N-[(35,45)-2,3-dihydro-3- hydroxy-3H- 1 -benzopyran-4-yl] -α-(f uro [3 ,2-c]pyridin-2-ylmethyl)-γ-hydroxy-2- [[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
Figure imgf000084_0001
Figure imgf000085_0001
A solution of the intermediate from Example 4 Step F (0.3 g, 0.7 mmol) and the Boc piperidine intermediate from Example 16 Step 5 (0.3 g, 1.0 mmol) in 2-propanol (8 mL) was refluxed overnight. Then the solvent was removed and the residue was purified by flash column chromatography on silica gel with ethyl acetate as eluant. The resulting product was dissolved in methanol (2 mL) and 4 N HCI in dioxane (1 mL) was added. After stirring at room temperature for 4 h, the solvent was removed and 1 N NaOH and methylene chloride was added to the resulting residue. The organic layer was separated, dried over Na2SO4, filtered and concentrated to give the title compound.
Figure imgf000085_0002
To a solution of 5-bromo-2-furaldehyde (5.6 g, 32.0 mmol) in DME (130 mL) was added tetrakis(triphenylphosphine)palladium (1.85 g, 1.6 mmol). The mixture was stined at room temperature for 10 min. Then 3-chloro-phenylboronic acid (5 g, 32 mmol) was added followed by 2 M Na2CO3 (32 mL). The reaction mixture was refluxed overnight. Then it was poured into 1 N NaOH and extracted with ethyl acetate three times. The combined organic layer was washed with brine, dried over Na2SO4, filtered and concentrated. The crude product was purified by flash column chromatography on silica gel with 1:9 ethyl acetate/hexane as eluant to give the title compound. Step C
(α5,γ5,2S)-4-[[5-(3-chlorophenyl)-2-furanyl]methyl]-N-[(3S,41S)-2,3-dihydro-3- hydroxy-3H- 1 -benzopyran-4-yl] -α-(furo [3 ,2-c]pyridin-2-ylmethyl)-γ-hydroxy-2- [[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
To a solution of intermediate from Step B (18.8 mg, 0.09 mmol) and intermediate from Step A (50 mg, 0.08 mmol) in 1,2-dichloroethane (0.4 mL) was added ΝaB(OAc)3Η (26.3 mg, 0.12 mmol). The reaction mixture was stined at room temperature overnight. Then solvent was removed and the crude product was purified by flash column chromatography on silica gel with 7 % 2 M NH3 in MeOH/ethyl acetate as eluant to give the title compound. 1H NMR (CD3OD, 500 MHz): 8.75 (s, IH), 8.33 (d, 7 = 5.7 Hz, IH), 7.67 (m, IH), 7.58 (d, 7= 7.8 Hz, IH), 7.51 (d, 7 = 5.7 Hz, IH), 7.35 (t, 7= 8.0 Hz, 7= 7.7 Hz, IH), 7.24 (m, IH), 7.17 (d, 7= 4.8 Hz, IH), 7.10 (t, 7 = 8.0 Hz, 7 = 7.7 Hz, IH), 6.84-6.74 (m, 4H), 6.40 (d, 7= 3.4 Hz, IH), 5.24 (d, 7 = 4.1 Hz, 1 H), 4.84-3.71 (m, 6H), 3.65 (s, 2H), 3.28-3.20 (m, IH), 3.12-3.10 (M, IH), 3.03-2.98 (m, 2H), 2.82-2.70 (m, 2h), 2.58-2.36 (m, 6H), 2.15-2.09 (m, IH), 1.51-1.46 (m, IH). LC-MS (M++l) (El) 796.3.
EXAMPLE 7 (αS,γS,2S)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(35,45)-2,3- dihydro-3-hydroxy-3H- 1 -benzopyran-4-yl] -γ-hydroxy-α-(5-thiazolylmethyl)-2- [ [(2,2,2-trifluoroethyl)amino]carbonyl]- 1 -piperazinepentanamide
Figure imgf000086_0001
Step A
To a solution of n-butyllithium (2.5 M in hexanes, 131 mL, 327.4
Figure imgf000087_0001
mmol) in dry ether (700 mL) was added, at -78 °C, a solution of 2- (trimethylsilyl)thiazole (50.0 g, 317.8 mmol) in dry ether (300 mL) slowly through a dropping funnel. After it was stined at -78 °C for 30 minutes, N-formyl morpholine (35.2 mL, 349.6 mmol) was dropped in slowly. The reaction solution was stined at - 78 °C for one hour. After it was washed with saturated aqueous sodium bicarbonate solution ( 500 ml), the organic layer was dried over anhydrous sodium sulfate, concentrated in vacuo, and purified by flash column chromatography on silica gel with hexanes / ethyl acetate = 2 / 1 as eluant to get the title compound as a slightly yellow solid. 1H ΝMR (CDC13, 500 MHz): δ 10.14 (s, 1 H), 9.13 (s, 1 H), 8.55 (s, I H).
Figure imgf000087_0002
A suspension of the aldehyde from the previous Step (21.4 g, 189.2 mmol) and malonic acid (43.3 g, 416.2 mmol) in pyridine (100 mL) and piperidine (1.9 mL) was refluxed for 2 hours. It was concentrated in vacuo. The residue was distributed between ether (1 L) and 1 Ν hydrochloric acid ( 200 mL). The organic layer was concentrated in vacuo to constant weight to get the title compound as a pale powder. 1H ΝMR (DMSO, 500 MHz): δ 9.15 (s, 1 H), 8.24 (s, 1 H), 7.80 (d, J=15.5 Hz, 1 H), 6.25 (d, J=15.5 Hz, 1 H).
Figure imgf000087_0003
To a suspension of the carboxylic acid obtained from the previous Step (5.0 g, 32.2 mmol) in a mixture of acetic acid and methanol ( 1 / 1, 330 mL) was added palladium on activated carbon ( 10 %, 2 g). After it was subjected to hydrogen gas (50 pci) for 24 hours, one more gram of palladium on activated carbon was added. It was subjected to hydrogen gas under the same pressure for 12 hours. Then, another gram of palladium on activated carbon was added. The reaction mixture was subjected to hydrogen gas under the same pressure for 10 more hours. It was filtered. The solid was washed with hot methanol for several times. The combined filtrate and washes was concentrated to get the title compound as pale solid. 1H NMR (DMSO, 500 MHz): δ 8.89 (s, 1 H), 7.65 (s, 1 H), 3.05 (d, J=7.5 Hz, 2 H), 2.57 (d, J=7.5 Hz, 2 H).
Figure imgf000088_0001
To a solution of the carboxylic acid from the previous Step (4.5 g, 28.6 mmol) and aminochromanol from Example 1 Step D (4.73 g, 28.6 mmol) in DMF (20 mL) was added l-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (7.2 g, 37.2 mmol), HOBt (5.1 g, 37.2 mmol) and diisopropylethylamine (15 mL, 85.9 mmol). After it was stined at room temperature for 3 hour, the solvent was removed in vacuo. The residue was washed with hot water (60 °C) to get the title compound as a pale solid. LC-MS (M++1) (El) 305.0.
Figure imgf000088_0002
To a suspension of the amide from the previous Step (7.5 g, 24.6 mmol) and para-toluenesulfonic acid (1.17 g, 6.16 mmol) in THF (50 mL) was added 2-methoxypropene (11.8 mL, 123.2 mmol). After it was stined at 40 °C for 3 hours, ethyl acetate (500mL) was added. It was washed with IN NaOH solution. Organic layer was dried over anhydrous sodium sulfate, concentrated in vacuo, and purified by flash column chromatography on silica gel with ethyl acetate as eluant to get the title compound as a white oily solid LC-MS (M++l) (El) 345.0.
Figure imgf000089_0001
To a solution of the protected amide from the previous Step (6.6 g, 19.2 mmol) and allyl bromide (1.83 ml, 21.1 mmol) in anhydrous THF (50 mL) was added dropwise of a solution of lithium bis(trimethylsilyl)amide in THF (1.0 M, 21.1 mL, 21.1 mmol) at -25 °C. The reaction solution was stined at -20 °C to -15 °C for 3 hours. Then, 500mL of ethyl acetate was added. It was washed with water. The organic layer was dried over anhydrous sodium sulfate, concentrated in vacuo, and purified by flash column chromatography on silica gel with hexanes / ethyl acetate =1 / 1 as eluant to get the title compound as a white solid. LC-MS (M++1) (El) 385.1.
Figure imgf000089_0002
To a solution of the olefin from the previous Step (6.13 g, 15.9 mmol) in isopropyl acetate (30 mL) was added sodium bicarbonate (1.0 g, 12 mmol) and water (25 mL). After it was cooled to 5 °C, N-chlorosuccinimide (3.86 g, 28.9 mmol) and sodium iodide (4.33 g, 18.9 mmol) were added. It was stined at 6 °C ~ 11 °C for 15minutes and warmed to 25 °C, at which it was stined for 3 hours. The organic layer was washed with aqueous sodium bicarbonate solution and with a solution of sodium sulfite (2.2 g) in water (12 mL). It was dried over anhydrous sodium sulfate, concentrated in vacuo, and purified by flash column chromatography on silica gel with hexanes / ethyl acetate =1 / 1 as eluant to get the title compound as a slightly yellow solid LC-MS (M++l) (El) 529.1.
Figure imgf000090_0001
To a solution of the intermediate from the previous Step (6.7 g, 12.7 mmol) in isopropyl acetate (20 mL) was added sodium methoxide in methanol ( 25 Wt %, 5.8 mL, 25.4 mmol) slowly. The reaction solution was stined at room temperature for 3 hours. After it was quenched with water, the organic layer was dried over anhydrous sodium sulfate, concentrated in vacuo, and purified by flash column chromatography on silica gel with ethyl acetate as eluant to get the title compound as a white solid. LC-MS Qs/ +1) (El) 401.5.
Figure imgf000090_0002
A solution of the epoxide from the previous Step (186 mg, 0.464 mmol) and the piperazine from Example 3, Step O (200 mg, 0.464 mmol) in ethanol (5 mL) was refluxed for 14 hours. After the solvent was removed, the residue was purified on a Biotage cartridge using ethyl acetate / methanol = 100 / 3 as eluent to get the title compound as a white solid. LC-MS (M++l) (El) 831.1. Step J
(a5 5,2S)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(3.S,4S)-2,3- dihydro-3-hydroxy-3H-l-benzopyran-4-yl]-γ-hydroxy-α-(5-thiazolylmethyl)-2- [ [(2,2,2-trifluoroethyl)amino]carbonyl]- 1 -piperazinepentanamide
To a solution of the protected final product from the previous Step (348 mg, 0.42 mmol) was in methanol (10 mL) was added ΗC1 in ether (1 M, 2.1 mL). It was stined at room temperature for 10 hours. After the solvent was removed, the residue was purified on preparative reverse phase ΗPLC. After neutralized with ammonia in methanol the fraction collected was concentrated in vacuo and run through a short silica gel column to get the title compound as a white solid. 1H ΝMR (CD3OD, 500 MHz): δ 8.80 (s, 1 H), 7.69 (d, J=8.5 Hz, 2 H), 7.65 ( s, 1 H), 7.47 ( d, J = 8.5 Hz, 2 H), 7.44 (s, 1 H), 7.08 - 7.13 ( m, 2 H), 6.82 (dt, J=7.5 Hz, 1.0 Hz, 1 H), 6.75 (dd, J=8.5 Hz, 1.0 Hz, 1 H), 5.20 (d, J=4.5 Hz, 1 H), 4.04-4.06 (m, 2 H), 3.92-3.98 ( m, 1 H), 3.78-3.82 (m, 1 H), 3.72-3.77 (m, 2 H), 3.06-3.10 (m, 1 H), 2.96- 3.03 (m, 2 H), 2.88-2.94 (m, IH), 2.85 (d, J=11.2 Hz, 1 H), 2.70-2.77 (m, 2 H), 2.63- 2.67 (m, 1 H), 2.44-2.50 (m, 1 H), 2.34-2.44 (m, 4 H), 2.00-2.04 (m, 1 H), 1.60 (s, 3 H), 1.59 (s, 3 H), 1.35-1.38 (m, 1 H). LC-MS (MM) (El) 791.4.
EXAMPLE 8 (α1S,γS,2S)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-N-[(3S,45)-2,3-dihydro- 3-hydroxy-3H-l-benzopyran-4-yl]-γ-hydroxy-α-(5-thiazolylmethyl)-2-[[(2,2,2- trifluoroethyl)amino]carbonyl] - 1 -piperazinepentanamide
Figure imgf000091_0001
Figure imgf000092_0001
A solution of the epoxide from Example 7, Step I (50 mg, 0.125 mmol) and the piperazine from Example 1, Step N ( 51mg, 0.125 mmol) in ethanol (5 mL) was refluxed for 12 hours. After the solvent was removed, the residue was purified on a Biotage cartridge using ethyl acetate / methanol = 100 / 3 as eluent to get the title compound as a white solid. LC-MS (MM) (El) 802.3.
Step B
(αS,γ5,25)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-N-[(3S,45)-2,3-dihydro-
3-hydroxy-3H-l-benzopyran-4-yl]-γ-hydroxy-α-(5-thiazolylmethyl)-2-[[(2,2,2- trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
To a solution of the protected final product from the previous Step (74 mg, 0.09 mmol) was in methanol (10 mL) was added ΗC1 in ether (1 M, 0.5 mL). It was stined at room temperature for 36 hours. After the solvent was removed, the residue was purified on preparative reverse phase ΗPLC. After neutralized with ammonia in methanol the fraction collected was concentrated in vacuo and run through a short silica gel column to get the title compound as a white solid. 1H ΝMR (DMSO, 500 MHz): δ 8.86 (s, 1 H), 8.41-8.84 (m, 1 H), 7.91 (d, J=8.5 Hz, 2 H), 7.63- 7.66 ( m, 3 H), 7.44 ( d, J = 8.5 Hz, 2 H), 7.03-7.08 (m, 2 H), 6.91 - 6.92 ( m, 1 H), 6.76 (t, J=7.5 Hz, 1 H), 6.69 (d, J=8.5 Hz, 1 H), 6.40-6.41 (m, 1 H), 5.23 (s, 1 H), 5.12-5.14 (m, 1 H), 4.67 (d, J= 3 Hz, 1 H), 4.13-4.15 (m, 1 H), 4.05-4.08 (m, 1 H), 3.85-3.93 ( m, 1 H), 3.71-3.82 (m, 2 H), 3.62-3.68 (m, 1 H), 3.09-3.13 (m, 1 H), 2.91-2.97 (m, 4 H), 2.65-2.67 (m, 2 H), 2.37-2.49 (m, 1 H), 2.18-2.32 (m, 3 H), 1.97 (t, J=12 Hz, 3 H), 1.20 (t, J=12 Hz, 3 H), 1.35-1.38 (m, 1 H). LC-MS (MM) (El) 762.3. EXAMPLE 9 (aS^5,2S)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(3.S,4S)-2,3- dihydro-3 -hydroxy-3H- 1 -benzopyran-4-yl] -γ-hydroxy-α-(thieno [3 ,2-c]pyridin-2- ylmethyl)-2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
TsCl (52.56 g, 0.275 mol) and triethylamine (41.89 mL, 0.298 mol) were added slowly to a solution of aminoacetaldehyde dimethyl acetal (24.12 g, 0.229 mol) in methylene chloride (100 mL) at 0°C. The resulting slurry was stined at 0°C and room temperature for 4 h. The mixture was diluted with methylene chloride (300 mL) and washed with water (100 mL), brine (100 mL), and dried over sodium sulfate. Biotage purification using EtOAc/Hexane (3:7) as the elute afforded the titled compound as a colorless oil.
Figure imgf000094_0001
To a solution of the titled compound from Step A (18.1 g, 69.8 mmol), triphenylphosphine (20.1 g, 76.9 mmol), and 3-thiophenemethanol (8.78 g, 76.9 mmol) in 150 mL of THF under N2 at 0°C, diisopropyl azodicarboxylate was added dropwise. The solution was stined at 0°C for 30 min. The solution was then placed in a cold room (5°C) overnight. The reaction was quenched with saturated NaHCO3 solution. THF was removed via vacuum. The mixture was extracted with EtOAc (3 x 150 mL). The combined EtOAc layers were washed with brine (100 mL), and dried over sodium sulfate. Biotage purification using EtOAc/Hexane (1:4) as the elute gave the titled compound as an oil.
Step C
NCQ
The mixture of the titled compound from Step B (25.14 g, 74.9 mmol) and concentrated HCI (25 mL) in dioxane (100 mL) was heated to reflux overnight. The solvent was removed via a vacuum. The pH of the aqueous solution was adjusted to 9 with concentrated NH OH. The aqueous was extracted with methylene chloride (3x100 mL). The combined methylene chloride layers were washed with brine and dried over sodium sulfate. Biotage purification using EtOAc/Hexane (1:1) as the elute afforded the titled compound as an oil.
Figure imgf000094_0002
To a solution of the titled compound from Step C (5.41 g, 40.0 mmol) in THF (50 mL) at -78°C, LDA (22.0 mL, 44.0 mmol, 2M solution) was added dropwise. The resulting yellowish solution was stined at -78°C for 30 min. Ethyl formate (3.86 mL, 48 mmol) was added slowly. The solution turned into slurry and it was allowed to stir at -78°C for 3 h. Saturated ammonium chloride solution (50 mL) was added. The mixture was warmed up to room temperature and extracted with EtOAc (3x150 mL). The EtOAc layers were washed with brine and dried over sodium sulfate. Biotage purification using EtOAc/Hexane as the elute gave the titled compound a yellow solid.
Figure imgf000095_0001
To a solution of triethyl phosphonoacetate (4.68 mL, 23.6 mmol) in THF (50 mL) at 0°C, NaH (1.13 g, 28.36 mmol, 60%) was added. The mixture was stined at 0°C for 30 min until no hydrogen formed. A solution of the titled compound from Step D (3.21 g, 19.69 mmol) in THF (50 mL) was added. The solution was stined at 0°C for 2 h. Saturated ammonium chloride (50 mL) was added and the mixture was extracted with EtOAc (3x100 mL). The combined EtOAc layers were washed with brine and dried over sodium sulfate. The titled compound was obtained as a white solid after flash chromatography using EtOAc/Hexane (6:4) as the elute.
Figure imgf000095_0002
A solution of the titled compound from Step E (0.21 g, 0.90 mmol) in
MeOH was hydrogenated at 1 atm using Pd/C as the catalyst. The mixture was stined for 2 h and filtered through Celite. The filtrate was concentrated via vacuum. The residue was dissolved in MeOH (5 mL). An aqueous of NaOH (0.036 g, 0.91 mmol) was added. The solution was stined at room temperature for 2 h. The solvents were removed. Toluene was added to the solid and removed (2 x 5 mL). 1 mL of IN HCI solution was added and removed. The above process was repeated with toluene (2 x). To a mixture of the residue in methylene chloride (10 mL), aminochromanol from Example 1, Step D (0.19 g, 1.15 mmol), EDC (0.27 g, 1.43 mmol), HOBT (0.19 g, 1.43 mmol), and DTEA (0.83 mL, 4.75 mmol) were added. The mixture was stined at room temperature overnight. The resulting slurry was filtered and washed with methylene chloride (2 x 10 mL). The titled compound was obtained as a white solid.
Figure imgf000096_0001
To a solution of the titled compound from Step F (0.55 g, 1.55 mmol) and 2-methoxypropene (1.48 mL, 15.5 mmol) in methylene chloride (20 mL) at room temperature, 10-camphorsulfonic acid (0.27 g, 1.16 mmol) was added portionwise in a period of 1 h. The progress of the reaction was monitored by TLC (EtOAc). Upon completion of addition of the acid, color of the reaction turned to light brown. It was allowed to stir for another 30 min and TLC indicated that there was no starting material left. The solution was diluted with methylene chloride (100 mL). The solution was washed with saturated sodium bicarbonate, brine, and dried over sodium sulfate. Biotage purification using EtOAc Hexane (8:2) as the elute gave the titled compound as a white solid.
Figure imgf000096_0002
To a solution of the titled compound from Step G (0.55 g, 1.396 mmol) and allyl bromide (0.18 mL, 2.09 mmol) and THF at -25°C, LiN(TMS)2 (1.67 mL, 1.67 mmol, IM THF solution) was added dropwise. The resulting yellow solution was stined at -25 °C for lh. TLC (EtOAc) showed no starting material left. The reaction was quenched with saturated ammonium chloride (5 mL) and extracted with EtOAc (3x50 mL). The combined EtOAc layers were washed brine and dried over sodium sulfate. Biotage purification using EtOAc/Hexane (8:2) as the elute afforded the titled compound as a white solid.
Figure imgf000097_0001
To a mixture of the titled compound from Step H (0.50 g, 1.15 mmol) and aqueous NaHCO3 (0.5 M, 50 mL) in EtOAc (50 mL) at 0°C, N-iodosuccinimide (NTS) (0.57 g, 2.53 mmol) was added slowly. The mixture was stined at room temperature for 24 h. LC/MS indicated that there was significant starting material left. Another 2 eq of NTS was added and the reaction was allowed to stined for one more day. After 48 h, LC/MS showed no more starting material left. The mixture was extracted with EtOAc (3x50 mL), washed with brine (50 mL), and dried over sodium sulfate. The solvent was removed via vacuum and the residue was dissolved in EtOAc (20 mL). NaOMe (3.37 mL, 1.68 mmol, 0.5 M in MeOH) was added dropwise to the solution. The brown solution was stined at room temperature for lh. TLC (EtOAc) showed no more starting material. The reaction was quenched with saturated NaHCO3 solution and extracted with EtOAc (3x50 mL). The organic layers were washed with brine (50 mL), and dried over sodium sulfate. Biotage purification using EtOAc as the elute gave the titled compound as a yellow solid.
Step J
Figure imgf000097_0002
A mixture of the titled compound from Step I (0.060 g, 0.133 mmol) and the titled compound from Example 3, Step O (0.069 g, 0.16 mmol) in 2-propanol (1 mL) was heated to reflux overnight. The solvent was removed. The titled compound was obtained as a white solid after flash chromatography using EtO Ac/acetone (7:3) as the elute.
Step K
(αS,γlS,2S)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(3»S,4S)-2,3- dihydro-3-hydroxy-3H-l-benzopyran-4-yl]-γ-hydroxy-α-(thieno[3,2-c]pyridin-2- ylmethyl)-2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentananιide
The titled compound from Step J (0.04 g, 0.045 mmol) was mixed with 1 mL of MeOΗ and 2 mL of IM ΗC1 in ether. The solution was stined at room temperature for 2 h. The solvent was removed. The residue was mixed with saturated ΝaΗCO3 (2 mL) and extracted with EtOAc (3x20 mL). The combined EtOAc layers were washed with brine, and dried over sodium sulfate. The titled compound was obtained as a white solid after flash chromatography using EtO Ac/acetone (1:1) as the elute. 1H NMR (δ, CDC13, 500 MHz): 9.32 (m, IH), 9.08 (s, IH), 8.40 (m, IH), 7.90 (m, IH), 7.59 (d, 7= 8.5 Hz, 2H), 7.42 (d, 7= 8.5 Hz, 2H), 7.40 (m, IH), 7.18 (m IH), 7.10 (m, IH), 6.80 (m, 2H), 5.28 (m, IH), 4.10 (m, 4H), 3.95 (m, IH), 3.90 (m, IH), 3.77 (m, IH), 3.43 (m, ZH), 3.15 (m, 3H), 3.08 (m, IH), 2.98 (IH), 2.65-2.90 (m, 3H), 2.58 (m, 3H), 1.85 (m, IH), 1.60 (s, 6H).LC-MS (M+l) (El): 842.
EXAMPLE 10
(α/?,γ5,25)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(35,4S)-3,4- dihydro-3-hydroxy-2H-l-benzopyran-4-yl]-α-(furo[2,3-c]pyrimidin-2-ylmethyl)-γ- hydroxy-2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
Figure imgf000099_0001
Step A:
Figure imgf000099_0002
To the title lactone from Example 5 Step C ( 0.67 mmol, 192 mg) in isopropanol (2.2 mL) containing DTEA (1 mmol, 175 μL) was added the title piperidine from Example 3 Step O (0.74 mmol, 318 mg). After 16 h the reaction was concentrated and the product was purified by flash chromatography eluting with 3% MeOH DCM, affording a light yellow foamy solid. MS (ESI): m/z 567 (M + H).
Step B:
Figure imgf000099_0003
A solution of 5-hydroxypyrimidine (7.3 mmol, 700 mg) and N- iodosuccinimide (23.4 mmol, 5.28 g) in saturated NaHCO3 (5 mL) and EtOAc (5 mL) was stined for 72 h. LC-MS analysis revealed the presence of the mono- and bis- iodinated products in approximately a 4:6 ratio. The aqueous layer containing the product was washed with hexanes and concentrated. The residue was triturated in EtOH and filtered. The filtrate was reduced and purified by flash chromatography eluting with a gradient of 2 - 4% MeOH/DCM (0.5% NH4OH), affording a mixture of mono- and bis-iodinated products. The monoiodide was isolated by reverse-phase chromatography eluting with a gradient of 10% - 90% MeCN/H2O (0.1 % TFA), affording the desired product as a reddish oil. MS (ESI): m/z 223 (M + H).
Figure imgf000100_0001
The iodopyrimidine from previous Step B (0.25 mmol, 113 mg), the acetylene from previous Step A (0.25 mmol, 142 mg), Pd(Ph3P)2Cl2 (0.0075 mol, 5.3 mg), Cul (0.013 mmol, 2.3 mg) and triethylamine (1.5 mmol, 150 mg) were stined in dry DMF (3 mL) at 60° under N2. After 1.5 h the acetylene was still present, and the reaction was replenished with 0.25 mmol of the iodopyrimidine, 0.0075 mmol of Pd(Ph3P)2Cl and 0.013 mol of Cul. After 2 h the reaction was concentrated. The product was purified by flash chromatography on silica eluting with 3% MeOH/DCM, followed by reverse-phase chromatography eluting with a gradient of 10% - 90%
MeCN/H2O (0.1%o TFA). The product was partitioned between NaHCO3 and EtOAc, and the organic phase was concentrated affording the desired product as a yellow solid. MS (ESI): m/z 661 (M + H).
Step D:
Figure imgf000101_0001
To the title compound from the previous Step C (0.07 mmol, 46 mg) in dioxane (1 mL) was added a solution of LiOH (0.2 mmol, 9 mg) in H O (200 μL). After 1.5 h the solvent was removed and the residue was concentrated twice from toluene. The compound was dissolved in dry DMF(1 mL) and brought to 0°. tert- Butyldimethylsilyl chloride (0.7 mmol, 105 mg) and imidazole (1.4 mmol, 95 mg) were added and the reaction was stined at 0° for 3 h. The reaction mixture was partitioned between EtOAc and pH 7 buffer. The organic phase was concentrated and the product was lyophilized from benzene and used without further purification. MS (ESI): m/z 793 (M + H).
Step E:
Figure imgf000101_0002
To a solution of the title compound from previous Step D (0.07 mmol), the title compound in Example 1 Step D (0.1 mmol, 13 mg), DIEA ( 0.21 mmol, 40 μL) and HOBt (0.14 mmol, 21 mg) in DMF (1 mL) was added EDC (0.1 mmol, 13 mg). After 3 h the reaction mixture was partitioned between EtOAc and H2O. The organic layer was concentrated and the product was lyophilized from benzene affording a solid which was used without further purification.
Step F:
(αR,γ5,2S)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(35,4,S)-3,4- dihydro-3-hydroxy-2H-l-benzopyran-4-yl]-α-(furo[2,3-c]pyrimidin-2-ylmethyl)-γ- hydroxy-2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide
The title compound from the previous Step E (0.07 mmol) was stined in a 2 mL solution of TΗF containing ΗF-pyridine (0.08 mmol) for 24 h. The reaction mixture was taken up in EtOAc (10 mL) and washed twice with saturated ΝaΗCO3. The product was purified by reverse-phase chromatography eluting with a linear gradient of 30% - 50% MeCN/ H2O (0.1 % TFA). The combined product fractions were partitioned between saturated NaHCO3 and EtOAc. The organic layer was concentrated, and the residue was lyophilized from MeCN/H2O affording the title product as an off-white powder. MS (ESI): m/z 826 (M + H).
EXAMPLE 11 (αS,γ>S,2S)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(35J4>S)-2,3- dihydro-3 -hydroxy-3H- 1 -benzopyran-4-yl] -γ-hydroxy-α- [(5 -pyridin-3-yl- 1 ,4-oxazol- 2-yl)methyl]-2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide, tris- trifluoroacetic acid salt
Figure imgf000103_0001
Step A
Figure imgf000103_0002
To a solution of 3-bromopyridine (57 g, 160 mmol) in 150 mL anhydrous THF at 0 °C was added 178 mL 2.13 M i-propylmagnesium bromide in THF dropwise slowly. The cooling bath was removed after finishing addition. The reaction mixture was stined for 1.5 hours. A solution of 40 g BOC-Gly-N(OMe)Me in 150 mL THF was added over 10 minutes. The reaction mixture was stined for 5 hours and quenched by adding saturated ammonium chloride solution. It was extracted with ethyl acetate. The combined organic layer was dried over anhydrous sodium sulfate and concentrated. The resulting product was purified by flash chromatography using 2:1 hexanes and ethyl acetate to give the titled compound. lH NMR (CDCI3, 500 MHz): 9.23 (d, 7= 1.9 Hz, IH), 8.88 (dd, 7= 1.4 and 4.8 Hz, IH),
8.34-8.37 (m, IH), 7.57 (dd, 7 = 5.1 and 8.1 Hz, IH), 5.48 (br s, IH), 4.69 (d, 7= 4.4 Hz, IH), 1.49 (s, 9H). LC-MS: 1.04 min. (237.1 m/Z, El).
Step B
Figure imgf000103_0003
To a solution of 29.3 g of the product from Step A above in methanol (25 mL) at 0 °C was added 200 mL 4.36 M HCI in methanol. The reaction was stined overnight and allowed to warm to room temperature. The solid was collected by filtration, washed with cold methanol and dried to give the title product. lH NMR (CD3OD, 500 MHz): 9.46 (d, 7 = 1.8 Hz, IH), 9.10 (dd, 7= 1.2 and 5.6 Hz, IH),
9.05-9.07 (m, IH), 8.205 (dd, 7 = 5.6 and 8.1 Hz, IH), 4.77 (s, 2H).
Figure imgf000104_0001
To a solution of 8.0 g of the product from Step B above in methylene chloride (200 mL) was added methyl 4-chloro-4-oxobutyrate (7.1 mL, 57.5 mmol). The reaction mixture was cooled at 0 °C. Diisopropylethylamine (20 mL, 114.9 mmol) was added to the reaction mixture dropwise. After 4 hours, the reaction mixture was concentrated under vacuum without heating, mixed with 25 g silica gel, and dry-loaded onto a silica gel column. It was eluted with 3-6% methanol in ethyl acetate to give the title product as a yellowish solid. lH NMR (CD3OD, 500 MHz):
9.12 (d, 7= 1.6 Hz, IH), 8.76 (dd, 7= 1.6 and 4.8 Hz, IH), 8.38-8.40 (m, IH), 7.59 (dd, 7 = 5.1 and 8.0 Hz, IH), 4.70 (s, 2H), 3.67 (s, 3H), 2.63 (s, 4H). LC-MS: 0.43 min. (251.2 m/Z, El).
Figure imgf000104_0002
A solution of 3.08 g of the product from Step C above in 100 mL phosphorus oxychloride was refluxed under nitrogen for 2 hours. The reaction mixture was cooled, concentrated under vacuum to dryness, mixed with some silica gel, dry-loaded onto a silica gel column, and purified by flash chromatography to give pure title compound as a solid. lH NMR (CDCI3, 500 MHz): 8.86 (d, 7= 1.4 Hz, IH), 8.49 (dd, 7= 1.2 and 4.9 Hz, IH), 8.08-8.11 (m, IH), 7.54 (s, IH), 7.50 (dd, 7 = 4.8 and 8.0 Hz, IH), 3.69 (s, 3H), 3.17 (t, 7 = 7.1 Hz, 2H) 2.89 (t, 7 = 7.1 Hz, 2H). LC-MS: 0.77 min. (233 m/Z, El).
Figure imgf000105_0001
To a solution of 0.9 g product from Step D above in 10 mL methanol was added a solution of 186 mg sodium hydroxide in 6 mL water. The reaction mixture was stined at room temperature for 3 hours. The solvents were removed under vacuum. The residue was re-dissolve in 10 mL water and the pH was adjusted to 6.5 with 1 N HCI. The resulting precipitated was collected by filtration and dried to give title compound as a solid. lH NMR (CD3OD, 500 MHz): 8.88 (d, 7= 1.8 Hz,
IH), 8.49 (dd, 7= 1.5 and 4.9 Hz, IH), 8.10-8.13 (m, IH), 7.56 (s, IH), 7.51 (dd, 7 = 5.1 and 8.1 Hz, IH), 3.15 (t, 7 = 7.2 Hz, 2H) 2.86 (t, 7= 7.2 Hz, 2H). LC-MS: 0.43 min. (219.2 m/Z, El).
Figure imgf000105_0002
To a suspension of the intermediate from Step E (0.92 g, 4.22 mmol) in NN-Dimethyl formamide (50 mL) was added diisopropylethylamine (1.83 mL, 10.55 mmol). To this clear solution was added (35,45)-4-amino-3,4-dihydro-2H- chromen-3-ol from Step D of Example 1 (0.73 g, 4.43 mmol), 1-hydroxybenzotriazole hydrate (0.86 g, 6.33 mmol), and l-ethyl-3-(3-dimethylaminopyropyl)carbodiimide hydrochloride (1.21 g 6.33 mmol) in that order. Additional portion of (3S,4S)-4- amino-3,4-dihydro-2H-chromen-3-ol (0.17 g) was added after 5 hours. The reaction mixture was stined for 60 hours and purified by flash chromatography using 5-10% methanol in ethyl acetate with 1% triethylamine to give title compound as an off- white solid.lΗ ΝMR (CD3OD, 500 MHz): 8.895 (d, 7 = 2.0 Hz, IH), 8.50 (dd, 7 = 1.5 and 4.9 Hz, IH), 8.12-8.14 (m, IH), 7.59 (s, lh), 5.505 (dd, 7 = 4.9 and 8.1 Hz, IH), 7.08-7.11 (m, 2H), 6.75-6.79 (m, 2H), 5.26 (d, 7 = 4.1 Hz, IH), 4.16 (d, 7= 3.7 Hz, 2H), 4.07-4.10 (m, IH), 3.20-3.32 (m, 2H), 2.82-2.94 (m, 2H). LC-MS (M+l, El) 366.1.
Step G
Figure imgf000106_0001
To a solution of the intermediate from Step F above (0.47 g, 1.29 mmol) in 50 mL 1:1 dimethyl formamide and methylene chloride was added 2- methoxypropene (0.465 g, 6.45 mmol) followed by camphor sulfonic acid (0.36 g, 1.55 mmol). The reaction mixture was stined overnight, diluted with ethyl acetate, washed with water (4x), saturated brine, and dried over anhydrous sodium sulfate. Solvents were removed in vacuo. The resulting crude product was purified by flash chromatography (2.5 and 3% MeOH in ethyl acetate) to give the title compound. lH NMR (CD3OD, 500 MHz) showed about 1 :3 ratio of amide rotomers. LC-MS (El)
406.1 (M+l) and 428.1 (M+Na).
Step H
Figure imgf000106_0002
To a solution of the intermediate obtained from Step G (0.368 g, 0.908 mmol) and allyl bromide (94.3 μL, 1.09 mmol) in THF (13 mL) cooled to -78 °C was added drop- wise a solution of LHMDS (1.1 mL, 1.0 M in THF). The reaction mixture was stined at -78 °C, warmed to -20 °C over 45 minutes, and quenched with saturated NaHCO3 solution. The resulting bi-phasic mixture was extracted with ethyl acetate (2X), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography with 0 and 1% methanol-ethyl acetate to give the desired compound. lH NMR (CDCI3, 500 MHz) showed about 1:3 ratio of amide rotomers. LC-MS (El) 913.3 (2M+Na).
Figure imgf000107_0001
To a solution of the intermediate obtained from Step H (0.84 g, 1.89 mmol) in ethyl acetate (15 mL) was added NaHCO3 solution (15 mL, 0.5 M) and the reaction cooled to 0 °C. N-iodo-succinimide (1.06 g, 4.73 mmol) was added and the reaction stined at room temperature for 4 hours. The reaction mixture was diluted with ethyl acetate. The organic layer was washed with a saturated solution of sodium thiosulfate (2x), water (3x), brine (lx) and dried over anhydrous sodium sulfate. The reaction mixture was filtered and concentrated in vacuo to give the desired compound as light brown foam. . lH ΝMR (CDCI3, 500 MHz) showed about 1 :3 ratio of amide rotomers. LC-MS (El) 590.0 (M+l).
Figure imgf000107_0002
To a solution of the intermediate from Step I (0.899 , 1.52 mmol) in ethyl acetate (13 mL) was added a solution of 0.5 M sodium methoxide in methanol (4.6 mL). After 20 minutes the reaction mixture was quenched with saturated ΝaHCO3 solution and extracted with ethyl acetate(3x). The organic layer was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography with 1% methanol in ethyl acetate to give the desired compound as a yellowish solid. lH NMR (CDCI3, 500 MHz) showed about 1:4 ratio of amide rotomers. LC-MS (ET) 462.1 (M+l).
Step K
Figure imgf000108_0001
To a solution of the intermediate obtained from Step J (67.4 mg, 0.146 mmol) in 2-propanol (1.5 mL) was added the intermediate from Example 3 Step O (52.5 mg, 0.122 mmol) and the resulting mixture refluxed overnight. The reaction mixture was purified on RP HPLC using 25-55% MeCN gradient over 15 minutes at 6.0 mL per minute with 0.1% TFA on a 9.4x250 mm Zorbax SB-C18 column. Pure product fractions were pooled and evaporated in vacuo to give the title compound. lH NMR (CD3OD, 500 MHz) showed about 1:5 ratio of amide rotomers. LC-MS (El)
892.3 (M+l) and 914.3 (M+Na).
Step L
(αS,γ5,2,S)-4-[l-[5-(4-chlorophenyl)-2-oxazolyl]-l-methylethyl]-N-[(31S,45)-2,3- dihydro-3 -hydroxy-3H- 1 -benzopyran-4-yl] -γ-hydroxy-α- [(5 -pyridin-3-yl- 1 ,4-oxazol- 2-yl)methyl]-2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide, tris- trifluoroacetic acid salt
To a solution of the intermediate from Step K (49.7 mg mg, 0.04 mmol) in anhydrous methanol (2 mL) was added a solution of ΗC1 in ether (0.80 mL, 1.0 M solution). After 20 hours, the reaction mixture was concentrated in vacuo. The residue was purified on RP ΗPLC using 30-60% MeCΝ gradient over 15 minutes at 6.0 mL per minute with 0.1%) TFA on a 9.4x250 mm Zorbax SB-C18 column. Pure product fractions were pooled, evaporated in vacuo, and lyophilized to give the title compound as a white solid. lH NMR (CD3OD, 500 MHz): 8.88 (d, 7= 1.8 Hz, IH), 8.50 (d, 7 = 4.8 Hz, IH), 8.255 (d, 8.7 Hz, IH), 8.11-8.13 (m, IH), 7.67-7.70 (m, 2H), 7.45-7.55 (m, 4H), 7.20 (d, 7= 6.9 Hz, IH), 7.10-7.13 (m, IH), 6.84-6.87 (m, IH), 6.77 (d, 8.3 Hz, IH), 5.23-5.25 (m, IH), 4.12-4.15 (m, 2H), 4.03-4.05 (m, IH), 3.96-4.01 (m, IH), 3.76-3.9 (m), 3.22-3.27 (m), 3.04-3.08 (m), 2.45-2.65 (m), 2.05-2.15 (m), 1.61 (s, 6H), 1.46- 1.53 (m), 1.28-1.34 (m). LC-MS (El) 852.3 (M+l).
EXAMPLE 12 (α5,γS,25)-4-[[5-(4-chloroρhenyl)-2-furanyl]methyl]-N-[(35,45)-2,3-dihydro- 3-hydroxy-3H-l-benzopyran-4-yl]-γ-hydroxy-α-[(5-pyridin-3-yl-l,4-oxazol-2- yl)methyl]-2-[[(2,2,2-trifluoroethyl)amino]carbonyl]-l-piperazinepentanamide, tris- trifluoroacetic acid salt
Figure imgf000109_0001
Figure imgf000110_0001
To a solution of the intermediate obtained from Example 11, Step J (51.6 mg, 0.12 mmol) in 2-propanol (1.5 mL) was added the intermediate from
Example 1 Step N (55.5 mg, 0.12 mmol) and the resulting mixture refluxed overnight. The reaction mixture was purified on RP HPLC using 30-60% MeCN gradient over 15 minutes at 6.0 mL per minute with 0.1%) TFA on a 9.4x250 mm Zorbax SB-C18 column. Pure product fractions were pooled and evaporated in vacuo to give the title compound. LC-MS (El) 863.3 (M+l) and 885.3 (M+Na).
Step B
(α5,γS,2S)-4-[[5-(4-chlorophenyl)-2-furanyl]methyl]-N-[(3S,45)-2,3-dihydro- 3 -hydroxy-3H- 1 -benzopyran-4-yl] -γ-hydroxy-α- [(5 -pyridin-3-yl- 1 ,4-oxazol-2- yl)methyl] -2- [ [(2,2,2-trifluoroethyl)amino] carbonyl] - 1 -piperazinepentanamide, tris- trifluoroacetic acid salt
To a solution of the intermediate from Step A above (61.4 mg, 0.051 mmol) in anhydrous methanol (2 mL) was added a solution of ΗC1 in ether (1.02 mL, l.O M solution). After 20 hours, the reaction mixture was concentrated in vacuo. The residue was purified on RP ΗPLC using 30-60% MeCΝ gradient over 15 minutes at 6.0 mL per minute with 0.1% TFA on a 9.4x250 mm Zorbax SB-C18 column. Pure product fractions were pooled, evaporated in vacuo, and lyophilized to give the title compound as a white solid. lΗ ΝMR (CD3OD, 500 MHz): 9.05-9.07 (m, IH), 8.64-8.68 (m, lh), 8.48-8.57
(m, IH), 8.25 (d, 8.7 Hz, 1ΝH), 7.80-7.88 (m, IH), 7.70-7.72 (m, 3H), 7.41 (d, 7 = 8.5 Hz, 2H), 7.20 (d, 7 = 7.8 Hz, IH), 7.10-7.13 (m, IH), 6.88 (d, 7= 3.4 Hz, IH), 6.83-6.86 (m, IH), 6.74-6.78 (m, 2H), 5.24-5.26 (m, IH), 4.28-4.40 (m, 2H), 4.08-4.14 (m, 2H), 4.01-4.03 (m, IH), 3.84-3.97 (m, 3H), 3.79-3.81 (m, IH), 3.37-3.42 (m, IH), 3.25-3.30 (m, IH), 3.09-3.15 (m, IH), 2.97-3.03 (m, IH), 2.70-2.79 (m, 2H), 2.00-2.06 (m, IH), 1.56- 1.52 (m. IH). LC-MS (El) 823.2 (M+l).
EXAMPLE 13 Preparation of Enzymes
Synthetic oligonucleotide cassettes of 444 base pairs were designed according to the wild-type sequence of pET-3b-HTVPR. Point mutations were incorporated into the DNA with a bias toward optimal codon usage in E. Coli to yield amino acid mutations listed in Table 2 below. The oligonucleotides were annealed and ligated into pUC-18 or pUC-19 by Midland Certified Reagent Company. The primary sequence was verified before subcloning into a pET-3b expression vector via Nde I and Bpull021 sites and reconfirmed by automated double-stranded DNA sequencing. Clones carrying the mutant DNA were transformed and expressed as previously described in Schock et al., 7. Biol. Chem. 1996, 271: 31957-31963 and Chen et al., J. Biol. Chem. 1995, 270: 21433-21436. The cells were lysed in 50 mM Tris-HCl pH 8.0, 1 mM EDTA, 0.1% NP40, 10 mM MgCl2 , and lOOμg / mL DNase I using a microfluidizer processor ( Microfluidics International Corp., Newton, MA). The mutant protease was extracted, refolded, and purified over affinity columns as previously described in Schock et al., 7. Biol. Chem. 1991, 271: 31957-31963. Protein concentrations were determined by amino acid analysis and purity was confirmed by SDS gel electrophoresis.
EXAMPLE 14 Assay for Inhibition of Microbial Expressed HTV Protease
Inhibition studies of the reaction of the protease (which was expressed in Eschericia coli) with a peptide substrate [Val-Ser-Gln-Asn-(betanapthyl)Ala-Pro- De-Val, 0.5 mg/mL at the time the reaction is initiated] were in 50 mM Na acetate, pH 5.5, 0.1% bovine serum albumin, 3.75% DMSO at 30°C for 1 hour. Various concentrations of inhibitor in 2 mL DMSO were added to 50 μL of the peptide solution in buffer. The reaction is initiated by the addition of 28 μL of 14.3 picomolar (wild type, K-60, Q-60) and 28.6 pM (V-18) protease in a solution of 50mM Na acetate pH 5.5 and 0.1% bovine serum albumin. The reaction was quenched with 120 μL of 10% phosphoric acid. Products of the reaction were separated by HPLC (VYDAC wide pore 5 cm C-18 reverse phase, acetonitrile gradient, 0.1% phosphoric acid). The extent of inhibition of the reaction was determined from the peak heights of the products. HPLC of the products, independently synthesized, proved quantitation standards and confirmation of the product composition. The compounds of the invention prepared in Examples 1-12 exhibited IC50 values ranging from about 0.05 to about 1 nM against the wild-type enzyme. The indinavir IC50 value against the wild type enzyme is 0.6 nM (average). The compounds of the invention prepared in Examples 1-12 exhibited IC50 values in the range of about 0.02 to about 5 nM against the mutant enzymes Q-60, K-60, and V-18. These IC50 values range from about 4-fold to greater than about 100-fold more potent than the values of of 20 to 50 nM obtained for indinavir against these same mutant enzymes.
EXAMPLE 15
Preparation of Viral Constructs Mutant viruses were constructed using gapped-duplex oligonucleotide mutagenesis of a subclone of plasmid pWT-6 as described in Colonno et al., Proc. Natl Acad. Sci. 1988, 85: 5449-5453. Infectious mutant proviral clones were constructed by subcloning the 833-b.p. Apal-Sse83871 fragment containing the mutagenized protease gene into the conesponding sites of plasmid pNL4-3 (see J. Virol. 1986, 59: 284-291). After transfection of the mutant proviral clone into HeLa cells and growth of viral stocks in cocultivated H9 human T-lymphoid cells, the complete sequence of the viral protease gene from the mutant viral population was verified as described in Nature 1995, 374: 569-571. The amino acid changes from wild type sequence for three of these viral constructs are shown in Table 2. Table 2 - Wild-type and Mutant HTV-1 Protease Sequences
Figure imgf000113_0001
Figure imgf000113_0002
See Condra et al., 7. Virol. 1996, 70: 8270-8276 and Olsen et al., 7. Biol. Chem. 1999, 274: 23699-23701 for further details.
EXAMPLE 16
Cell Spread Assay Inhibition of the spread of HTV in cell culture was measured according to Nunberg et al., J. Virol. 1991, 65: 4887. In this assay, MT-4 T-lymphoid cells were infected with HTV-1 (wild-type, unless otherwise indicated) by using a predetermined inoculum, and cultures were incubated for 24 h. At this time, <1% of the cells were positive by indirect immunofluorescence. Cells were then extensively washed and distributed into 96-well culture dishes. Serial twofold dilutions of inhibitor were added to the wells, and cultures were continued for 3 additional days. At 4 days postinfection, 100% of the cells in control cultures were infected. HTV-1 p24 accumulation was directly conelated with virus spread. The cell culture inhibitory concentration was defined as the inhibitor concentration in nanomoles/liter which reduced the spread of infection by at least 95%, or CIC95. The compounds of the invention prepared in Examples 1-12 exhibited CIC95 values in the range of from less than about 8 to about 50 nM against the wild-type viral construct. The CIC95 of indinavir against the wild-type viral construct is from 50 to 100 nM. The compounds of the invention prepared in Examples 1-12 exhibited CIC95 values in the range of about 8 to about 125 nM against the viral constructs Q60, K-60, and V-18. These CIC95 values range from about 4-fold to more than about 100-fold more potent than the values of greater than 1000 nM obtained for indinavir against these same viral constructs.
EXAMPLE 17 Inhibition of Virus Spread
A. Preparation of HTV-infected MT-4 cell Suspension MT cells are infected at Day 0 at a concentration of 250,000 per ml with a 1:1000 dilution of HTV-1 strain Tub stock (final 125 pg p24/ml; sufficient to yield <1% infected cells on day 1 and 25-100% on day 4). Cells are infected and grown in the following medium: RPMI 1640 (Whittaker BioProducts), 10% inactivated fetal bovine serum, 4 mM glutamine (Gibco Labs) and 1:100 Penicillin- Streptomycin (Gibco Labs). The mixture is incubated overnight at 37°C in 5% CO2 atmosphere.
B. Treatment with Inhibitors
A matrix of nanomolar range concentrations of the pairwise combinations is prepared. At Day 1, aliquots of 125 ml of inhibitors are added to equal volumes of HTV-infected MT-4 cells (50,000 per well) in a 96-well microtiter cell culture plate. Incubation is continued for 3 days at 37°C in 5% CO2 atmosphere.
C. Measurement of Virus Spread Using a multichannel pipettor, the settled cells are resuspended and
125 ml harvested into a separate microtiter plate. The supernatant is assayed for HTV p24 antigen.
The concentration of HTV p24 antigen is measured by an enzyme immunoassay, described as follows. Aliquots of p24 antigen to be measured are added to microwells coated with a monoclonal antibody specific for HTV core antigen. The microwells are washed at this point, and at other appropriate Steps that follow. Biotinylated HTV-specific antibody is then added, followed by conjugated streptavidin-horseradish peroxidase. A color reaction occurs from the added hydrogen peroxide and tetramethylbenzidine substrate. Color intensity is proportional to the concentration of HTV p24 antigen.
Calculation of Degree of Synergy
When there is synergy, pairwise combinations of inhibitors are found to exhibit markedly enhanced inhibition of virus spread, in comparison to each inhibitor alone, or in comparison to merely additive inhibition of each inhibitor.
The data is processed as follows: fractional inhibitory concentration ratios (F C) are calculated according to Elion, et al., J. Biol. Chem. 1954, 208: 477. The minimum sum of FICs, which is the maximum synergy, is determined for various pairwise combinations. The smaller the number, the greater the synergy.
EXAMPLE 18 Preparation of 4-(tert-butyloxycarbonyl)-2(5)-((2,2,2-trifluoroethyl)aminocarbonyl) piperazine Step One: Preparation of the pyrazine amide
Figure imgf000116_0001
Pyrazine 2-carboxylic acid (1204 g) was suspended in DMF (4.8 L, 4 mL/g acid). 2,2,2-trifluoroethylarnine»HCl (TFEA»HC1) (1200 g), 1- hydroxybenzotriazole (HOBT) (60 g) and triethylamine (TEA) (1410 mL) were then added sequentially (exotherm upon addition of TEA, flask cooled with ice bath and temperature kept below 35 °C). The reaction was cooled to 15 °C and l-(3- dimethylaminopropyl)-3-ethylcarbodiimide«HCl (EDC»HC1) (1940 g) was added portionwise over 15-30 min. The reaction temperature was kept below 35 °C. When the reaction appeared complete (approx. two hours, <5% pyrazine 2-carboxylic acid byLC assay), the reaction mixture (yellow/white slurry) was diluted with 10% K2CO3 in water (24 L, 20 mL/g acid) and the reaction slurry was kept below 35 °C. The slurry was cooled to 10 °C, aged for two hours and filtered (mother liquor assay=3- 4mg/mL). The wet cake was washed with deionized water (12 L, 10 mL/g acid) and dried under vacuum (22" Hg) at 40 °C with a nitrogen purge. Theoretical yield of 1816 g . Actual yield 1533 g (84%). 1H NMR: (CD3CN, 400 MHz): δ 9.29(d, J=1.5 Hz, IH), 8.82 (d, J= 2.5 Hz, IH), 8.63 (dd, J= 2.6,1.4 Hz, IH), 8.40 (bs, IH), 4.14 (dq, J=9.4, 6.8 Hz, 2H).
HPLC Assay conditions: Waters Xtena RP8 column, elution with acetonitrile and 5 mM K phosphate adjusted to pH= 8, detection at 220 nm.
Step Two: Preparation of the piperazine amide
Figure imgf000116_0002
Pyrazine amide (60.2 g 0.268 mol, not conected for water content) was suspended in absolute ethanol (550 mL) in a 1.0 L autoclave hydrogenation vessel and cooled to 15 °C. Wet 20% Pd(OH)2/C 11.0 g (20wt%, 50wt%wet) was added and reaction was purged with N2 three times. H2 (5 psig) was introduced with stirring and the temperature maintained at 15 °C for 60 minutes. The temperature was then increased to 60 °C and the hydrogen pressure increased to 40 psig and the reaction mixture stined for 18 additional hours. The reaction was considered complete when conversion is >99% by LC assay. The reaction mixture was filtered through Solka- Floe and the catalyst solids were washed with ethanol 2 X 110 mL. Assay of the combined filtrate and washes gave 53.5 g of racemic piperazine amide (Yield = 86%) IH NMR (CD3CN, 400 MHz): δ 7.58 (bs, IH), 3.90 (dq, J=9.5,6.7 Hz, 2H), 3.24(dd, J=7.9, 5.5 Hz, IH), 2.96 (dd, J= 12.1, 3.6 Hz, IH), 2.84-2.78 (m, IH), 2.77-2.67 (m, 3H), 2.66-2.56 (m, IH), 1.90 (s, 2 H). HPLC Assay conditions: YMC Basic column, elution with acetonitrile and 0.1% aqueous H3PO4, detection at 210 nm.
Step Three: Resolution of the piperazine amide
Figure imgf000117_0001
The pip amide ethanol filtrate (116.37 g containing 10.3 g of racemic pip amide by LC assay) was concentrated in vacuo to a final volume of 40.2 mL (3.9 mL per gram of pip amide) and the slurry is diluted with 82.4 mL (8 mL per gram pip amide) of acetonitrile (ACN) and stined until homogenous. Separately (S)- camphorsulfonic acid ( (S)-CSA) (19.26 g, MW= 232.30, 1.7 eq) was dissolved in 185 mL of ACN (18 mL per gram of pip amide). The water content of the two solutions was then determined by Karl Fisher titration. The CSA solution was added to the pip amide solution giving a small exotherm to approx. 31-32 °C. Water (11.02 mL, 1.118 mL per gram of pip amide minus the total water content of the two solutions) was then added, such that the acetonitrile:ethanol:water ratio was 26:2.9:1.1 (v/v/v). Solids began to form after 15-30 min. The solution/slurry was heated to 72 °C to completely dissolve all solids. The yellow solution was recooled to 62 °C and seeded with a slurry of 10.3 mg of pip amide salt in 1 mL of acetonitrile. After a two hour age at 62 °C the slurry was allowed to cool to room temperature overnight
(crystallization was complete when loss to mother liquors was < 21 mg pip amide/mL by LC assay. The slurry was filtered then washed with 2 x 30 mL of ACN:EtOH:H2O [(26:2.9:1.1), (v:v:v)] solution. The wet cake (-13 g, white solid) was dried at 40 °C in a vacuum oven (24 in Hg, nitrogen sweep) to give 11.16 g of product (yield = 33%). Assay method (Pip Amide) as above. Chiral assay gives an enantiomeric excess (ee) of 98.0%.
IH NMR (CD3OD, 400 MHz): d4.84(bs, 5H), 4.64 (dd, J=12.0, 3.6 Hz, IH), 4.13- 3.94 (m, 3H), 3.77 (m, 2H), 3.66 (m, IH), 3.54-3.43 (m, 2H), 3.28(d, J= 14.7 Hz, 2H), 2.82 (d, 14.7 Hz, 2H), 2.55 (m, 2H), 2.36 (m, 2H), 2.12-1.998 (m, 4H), 1.92 (d, J=18.4 Hz, 2H), 1.72 (m, 2H), 1.45 (m, 2H), 1.09 (s, 6H), 0.87 (s, 6H).
Enantiomeric excess determined by chiral HPLC of the mono BOC piperazine amide. HPLC assay conditions: Chiral AGP column, elution with acetonitrile and 10 mM Kphospate, pH=6.5, detection at 210 nm.
Step Four: Upgrade of ee of (SVpiperazine amide bis (S)-CS A salt
S)-CSA- H20
Figure imgf000118_0001
To a 12 L flask was charged (S)-pip amide salt (412.87 g) having an ee of less than 98%), 7.43 L of ACN and 825 mL of 190 proof EtOH. The slurry was heated to 75 °C, aged for 1 hr at 75 °C (during heating the slurry thickened considerably), then allowed to cool to 25 °C overnight. The slurry was filtered and washed with EtOH (190 proof):ACN (10:90) (2 x 800 mL, 2 mL g). The white solid was dried in a vacuum oven at 24 in Hg, 40 °C with a nitrogen sweep to give 400 g of product with an ee of 99%. Assays (normal and chiral) were performed as described above in the prior steps.
Step Five: Procedure for (S)-Mono BOC piperazine amide: BOC Protection
Figure imgf000119_0001
Bis (S)-CS A piperazine amide salt (20 g) was suspended in a mixture of 113 mL of isopropyl acetate (TPAc) and 57 mL of acetonitrile. Triethylamine (8.26 mL, 2 eq) was added and the mixture stined until homogenous. A solution of di t- butyl dicarbonate (TBDC) (6.46 g, 1.0 eq) in a mixture of 20 mL isopropyl acetate and 10 mL of acetonitrile (ACN) was then added over 10 minutes. After aging for two hours the solution was assayed as necessary by LC (Pip Amide Assay, see above) until the reaction was complete (i.e., less than 5% starting material). When the reaction was complete, 100 mL of water and 135 mL of isopropyl acetate were added, the resulting layers were separated and the organic layer was concentrated to 28 mL. The residue was then diluted with 28 mL of isopropyl alcohol and reconcentrated to 28 mL. This was repeated two additional times. The yield of BOC pip amide was 87% with a mono:bis BOC ratio of 95:5, as determined by HPLC. 1H NMR (CDC13, 400MHz): δ=7.39 (app t, 7=6.3Hz, IH), 3.96 (dd, 7=3.5, 13.4Hz, IH), 3.88 (m, 2H), 3.67 (d, 7=11.5Hz, IH), 3.39 (dd, 7=3.8, 8.6Hz, IH), 3.13 (dd, 7=8.6, 13.3Hz, IH), 3.02 (br, IH), 2.91 (m, IH), 2.77 (m, IH), 1.43 (s, 9H).
13C NMR (CDC13, ) δ= 171.43, 154.41, 123.89 (q, J=78.5Hz), 80.16, 57.65, 43.63, 45.6 (br), 44.0 (br), 40.20 (q, J=34.7Hz), 28.19. HPLC Assay conditions: YMC Basic column, elution with acetonitrile and 0.1% aqueous H3PO , detection at 210 nm. While the foregoing specification teaches the principles of the present invention, with examples provided for the purpose of illustration, the practice of the invention encompasses all of the usual variations, adaptations and/or modifications that come within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A compound of formula:
Figure imgf000121_0001
wherein
A is CH or N;
Rl is -F or -Cl;
R2 and R3 are each independently -H or methyl; and
R4 is
Figure imgf000121_0002
X is O or S;
Y is pyridyl which is optionally substituted with from 1 to 3 substituents each of which is independently halogen, cyano, Cχ-C6 alkyl, or Cχ-C6 alkoxy; each Z is independently hydrogen, halogen, cyano, Cχ-C6 alkyl, or Cχ-C6 alkoxy; and
q is an integer from zero to 2;
and with the proviso that when A is N and R4 is:
Figure imgf000122_0001
o- , then R2 and R3 are both -H;
or a pharmaceutically acceptable salt thereof.
The compound according to claim 1, wherein the compound is of formula:
Figure imgf000122_0002
or a pharmaceutically acceptable salt thereof.
The compound according to claim 1, wherein
R2 and R3 are either both -H or both methyl; R4 IS
Figure imgf000123_0001
Y is pyridyl which is optionally substituted with from 1 to 3 substituents each of which is independently halogen, cyano, C1-C4 alkyl, or C1-C4 alkoxy;
or a pharmaceutically acceptable salt thereof.
4. The compound according to claim 3, wherein
R i is
Figure imgf000123_0002
Q is -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy;
or a pharmaceutically acceptable salt thereof.
5. The compound according to claim 1, wherein
R2 and R3 are either both -H or both methyl;
Figure imgf000123_0003
X is S or O;
each Z is independently -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy; and
q is an integer from zero to 2;
or a pharmaceutically acceptable salt thereof.
The compound according to claim 5, wherein
R4 IS
Figure imgf000124_0001
Z is -H, C1-C4 alkyl, or C1-C4 alkoxy;
or a pharmaceutically acceptable salt thereof.
The compound according to claim 1, wherein
R and R3 are either both -H or both methyl;
R4 i is
Figure imgf000124_0002
each Z is independently -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy; and
q is an integer from zero to 2;
and with the proviso that when A is N and R is:
Figure imgf000125_0001
then R2 and R3 are both -H;
or a pharmaceutically acceptable salt thereof.
The compound according to claim 7, wherein
R2 and R3 are either both -H or both methyl; and
R4 is
Figure imgf000125_0002
and with the proviso that when A is N and R4 is:
Figure imgf000125_0003
or , then R2 and R3 are both -H;
or a pharmaceutically acceptable salt thereof.
9. The compound according to claim 1, wherein
R2 and R3 are either both -H or both methyl;
R4 IS
Figure imgf000125_0004
each Z is independently -H, -F, -Cl, -Br, C1-C4 alkyl, or C1-C4 alkoxy; and
q is an integer from zero to 2; or a pharmaceutically acceptable salt thereof.
10. The compound according to claim 9, wherein
R4 i is
Figure imgf000126_0001
Z is -H, C1-C4 alkyl, or C1-C4 alkoxy;
or a pharmaceutically acceptable salt thereof.
11. A compound selected from the group consisting of
Figure imgf000126_0002
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
and pharmaceutically acceptable salts thereof.
12. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
13. A pharmaceutical composition which comprises the product made by combining a therapeutically effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
14. A combination useful for inhibiting HTV protease, for treating or preventing infection by HTV, or for preventing, treating or delaying the onset of ADDS, which comprises a therapeutically effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt thereof, and a therapeutically effective amount of an HTV infection/ ADDS treatment agent selected from the group consisting of HTV/ATDS antiviral agents, immunomodulators, and anti-infective agents.
15. The combination according to claim 14, wherein the HTV infection/ ADDS treatment agent is an antiviral selected from the group consisting of non-nucleoside HTV reverse transcriptase inhibitors, nucleoside HTV reverse transcriptase inhibitors, HTV integrase inhibitors, and CCR5 receptor antagonists.
16. A method of inhibiting HTV protease in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of the compound according to claim 1 or a pharmaceutically acceptable salt thereof.
17. A method for preventing or treating infection by HTV or for preventing, treating or delaying the onset of ADDS in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of the compound according to claim 1 or a pharmaceutically acceptable salt thereof.
18. The method according to claim 17, wherein the compound is administered in combination with a therapeutically effective amount of at least one
HTV infection/ ADDS treatment agent selected from the group consisting of HTV/ ADDS antiviral agents, immunomodulators, and anti-infective agents.
19. The method according to claim 17, wherein the compound is administered in combination with a therapeutically effective amount of at least one antiviral selected from the group consisting of non-nucleoside HIV reverse transcriptase inhibitors, nucleoside HTV reverse transcriptase inhibitors, HTV integrase inhibitors, and CCR5 receptor antagonists.
20. A method of inhibiting HTV protease in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of the composition according to claim 12.
21. A method for preventing or treating infection by HTV or for preventing, treating, or delaying the onset of ADDS in a subject in need thereof which comprises administering to the subject a therapeutically effective amount of the composition according to claim 12.
PCT/US2002/016739 2001-05-30 2002-05-24 Piperazine pentanamide hiv protease inhibitors WO2002096359A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002305720A AU2002305720A1 (en) 2001-05-30 2002-05-24 Piperazine pentanamide hiv protease inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US29437001P 2001-05-30 2001-05-30
US60/294,370 2001-05-30

Publications (2)

Publication Number Publication Date
WO2002096359A2 true WO2002096359A2 (en) 2002-12-05
WO2002096359A3 WO2002096359A3 (en) 2003-03-13

Family

ID=23133114

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/016739 WO2002096359A2 (en) 2001-05-30 2002-05-24 Piperazine pentanamide hiv protease inhibitors

Country Status (2)

Country Link
AU (1) AU2002305720A1 (en)
WO (1) WO2002096359A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6649761B2 (en) * 2001-05-30 2003-11-18 Merck & Co., Inc. Process for preparing piperazinepentaneamide HIV protease inhibitors
JP2007502817A (en) * 2003-08-20 2007-02-15 イーライ リリー アンド カンパニー Compounds, methods and formulations for oral delivery of glucagon-like peptide (GLP) -1 compounds or melanocortin 4 receptor (MC4) agonist peptides
US8552039B2 (en) 2003-08-20 2013-10-08 Emisphere Technologies, Inc. Compounds, methods and formulations for the oral delivery of a glucagon-like peptide (GLP-1) compound or a melanocortin-4 receptor (MC4) agonist peptide

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001038332A1 (en) * 1999-11-24 2001-05-31 Merck & Co., Inc. Gamma-hydroxy-2-(fluoroalkylaminocarbonyl)-1-piperazinepentanamides as hiv protease inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001038332A1 (en) * 1999-11-24 2001-05-31 Merck & Co., Inc. Gamma-hydroxy-2-(fluoroalkylaminocarbonyl)-1-piperazinepentanamides as hiv protease inhibitors

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6649761B2 (en) * 2001-05-30 2003-11-18 Merck & Co., Inc. Process for preparing piperazinepentaneamide HIV protease inhibitors
JP2007502817A (en) * 2003-08-20 2007-02-15 イーライ リリー アンド カンパニー Compounds, methods and formulations for oral delivery of glucagon-like peptide (GLP) -1 compounds or melanocortin 4 receptor (MC4) agonist peptides
US7947841B2 (en) 2003-08-20 2011-05-24 Emisphere Technologies, Inc. Compounds, methods and formulations for the oral delivery of a glucagon-like peptide (GLP)-1 compound or a melanocortin-4 receptor (MC4) agonist peptide
JP4754487B2 (en) * 2003-08-20 2011-08-24 エミスフィアー テクノロジーズ インコーポレイテッド Compounds, methods and formulations for oral delivery of glucagon-like peptide (GLP) -1 compounds or melanocortin 4 receptor (MC4) agonist peptides
US8546581B2 (en) 2003-08-20 2013-10-01 Emisphere Technologies, Inc. Compounds, methods and formulations for the oral delivery of a glucagon-like peptide (Glp)-1 compound or a melanocortin-4 receptor (Mc4) agonist peptide
US8552039B2 (en) 2003-08-20 2013-10-08 Emisphere Technologies, Inc. Compounds, methods and formulations for the oral delivery of a glucagon-like peptide (GLP-1) compound or a melanocortin-4 receptor (MC4) agonist peptide
US8765796B2 (en) 2003-08-20 2014-07-01 Emisphere Technologies, Inc. Compounds, methods and formulations for the oral delivery of a glucagon-like peptide (GLP-1) compound or a melanocortin-4 receptor (MC4) agonist peptide

Also Published As

Publication number Publication date
AU2002305720A1 (en) 2002-12-09
WO2002096359A3 (en) 2003-03-13

Similar Documents

Publication Publication Date Title
US6642237B1 (en) Gamma-hydroxy-2-(fluoroalkylaminocarbonyl)-1-piperazinepentanamides and uses thereof
CN110612294B (en) Human cerebulin ligands and bifunctional compounds comprising same
US7135467B2 (en) HIV integrase inhibitors
JP5765965B2 (en) 1,3,4,8-Tetrahydro-2H-pyrido [1,2-a] pyrazine derivatives and their use as HIV integrase inhibitors
US20070203147A1 (en) 2-Aminothiazole Compounds Useful As Aspartyl Protease Inhibitors
KR20230140606A (en) Modulators of estrogen receptor proteolysis and associated methods of use
JP2020506922A5 (en)
NZ555376A (en) Anhydrous crystalline monopotassium salt of raltegravir
AU692509B2 (en) HIV protease inhibitors
KR101624675B1 (en) Amide compounds as boosters of antivirals
KR20060114374A (en) Therapeutic combinations
CN1694705A (en) Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
US5650412A (en) HIV protease inhibitors useful for the treatment of AIDS
WO2002096359A2 (en) Piperazine pentanamide hiv protease inhibitors
WO2002067683A1 (en) Hiv protease inhibitors supported on cation exchange resins for oral administration
US20040067216A1 (en) Hiv protease inhibitors supported on cation exchange resins for oral administration
US6589962B1 (en) Alpha-hydroxy-gamma-[[(carbocyclic-or heterocyclic-substituted)amino]carbonyl]alkanamide derivatives and uses thereof
US20030125336A1 (en) Hydrohalide salts of an HIV protease inhibitor
RU2795146C2 (en) Cereblon ligands and bifunctional compounds containing them
WO2001005230A1 (en) Alpha-hydroxy-gamma-[[(carbocyclic-or heterocyclic-substituted)amino]carbonyl]alkanamide derivatives and uses thereof
CN1938288A (en) 2-aminothiazole compounds useful as aspartyl protease inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

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

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP