WO2012158550A2 - Selective heterocyclic sphingosine 1 phosphate receptor modulators - Google Patents

Selective heterocyclic sphingosine 1 phosphate receptor modulators Download PDF

Info

Publication number
WO2012158550A2
WO2012158550A2 PCT/US2012/037609 US2012037609W WO2012158550A2 WO 2012158550 A2 WO2012158550 A2 WO 2012158550A2 US 2012037609 W US2012037609 W US 2012037609W WO 2012158550 A2 WO2012158550 A2 WO 2012158550A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
dihydro
inden
alkyl
Prior art date
Application number
PCT/US2012/037609
Other languages
French (fr)
Other versions
WO2012158550A3 (en
WO2012158550A9 (en
Inventor
Marcus F. Boehm
Esther Martinborough
Enugurthi Brahmachary
Manisha Moorjani
Junko Tamiya
Liming Huang
Adam Richard Yeager
Original Assignee
Receptos, Inc.
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 Receptos, Inc. filed Critical Receptos, Inc.
Priority to JP2014510511A priority Critical patent/JP6129159B2/en
Priority to ES12785231T priority patent/ES2758841T3/en
Priority to US14/117,514 priority patent/US9481659B2/en
Priority to EP12785231.7A priority patent/EP2706999B1/en
Publication of WO2012158550A2 publication Critical patent/WO2012158550A2/en
Publication of WO2012158550A3 publication Critical patent/WO2012158550A3/en
Publication of WO2012158550A9 publication Critical patent/WO2012158550A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • C07D285/01Five-membered rings
    • C07D285/02Thiadiazoles; Hydrogenated thiadiazoles
    • C07D285/04Thiadiazoles; Hydrogenated thiadiazoles not condensed with other rings
    • C07D285/121,3,4-Thiadiazoles; Hydrogenated 1,3,4-thiadiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/22Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • C07D277/30Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/24Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • 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
    • C07D417/02Heterocyclic 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 two hetero rings
    • C07D417/10Heterocyclic 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 two hetero rings linked by a carbon chain containing aromatic 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
    • C07D417/02Heterocyclic 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 two hetero rings
    • C07D417/12Heterocyclic 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 two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the invention relates to compounds which are agonists of the sphingosine 1- phosphate receptor subtype 1, methods of their synthesis and methods of their therapeutic and/or prophylactic use.
  • the SlPi/EDGi receptor is a G-protein coupled receptor (GPCR) and is a member of the endothelial cell differentiation gene (EDG) receptor family.
  • Endogenous ligands for EDG receptors include lysophospholipids, such as sphingosine- 1 -phosphate (SIP).
  • SIP sphingosine- 1 -phosphate
  • the present invention is directed to heterocyclic compounds adapted to act as agonists of SIP receptor subtype 1, SlPj; methods of preparation and methods of use, such as in treatment of a malcondition mediated by SIPi activation, or when activation of SIPj is medically indicated.
  • Certain embodiments of the present invention comprise a compound having the structure of Formula (I) or a pharmaceutically acceptable salt, ester, prodrug, homolog, tautomer, stereoisomer, or hydrate, or solvate thereof:
  • a dashed line signifies that a single bond or a double bond can be present, provided that there are two double bonds and three single bonds in the ring comprising A 1 , A , and A .
  • a , A , and A each independently can be CH or S or N; provided that one of A , A 2 , and A 3 is S.
  • R 1 can be di-substituted phenyl or di-substituted pyridinyl where the phenyl and pyridinyl substituents ca each be independently any of halo, nitro, cyano, perfluromethyl, fluorinated methyl, and Ci-4-alkoxy.
  • R 1 is di-substituted phenyl, such phenyl is para- substituted with C -alkoxy.
  • X can be -NR'R" or -OR'";
  • R' can be H, C 1-4 alkyl, n-hydroxy C alkyl, -S0 2 -R 3 , or -CO-R 3 .
  • R" can be H, -S0 2 -R 5 , C alkyl optionally substituted with 1 or more R 4 , or a ring moiety optionally substituted with R 6 wherein such ring moiety is pyridinyl, oxazolyl, piperidinyl, cyclohexyl, morpholinyl, thiazolyl, pyrazolyl, pyrrolidinyl, imidazolyl, or phenyl.
  • R'" can be H, C alkyl, or -CO-R 3 .
  • R' and R" taken together with the nitrogen atom to which they are bound can form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle is optionally singly or multiply substituted with substituents independently selected from the group consisting of -OH, oxo, -NH 2 , n-hydroxy-CM alkyl, -COOH, -(CH 2 ) m -COOH, - (CH 2 ) m -COOR 3 , -N(R 3 R 3 ), and -(CH 2 ) m -CO-N(R 7 R 7 ).
  • Each R 3 can be independently C M alkyl or H.
  • Each R 5 can be independently R 4 , C M alkyl, C 3-6 cycloalkyl, or C M alkyl optionally substituted with 1 or more R 4 .
  • Each R 6 can be independently halo, OH, -NH 2 , -NHR 3 , -N(R 3 R 3 ), -COOH, - COOR 3 , -NHCO-R 3 .
  • Each R 7 can be independently C 1-4 alkyl or H, or two R 7 taken together with the nitrogen atom to which they are bound can form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle can be optionally substituted with -OH, -NH 2 , -N(R 3 R 3 ), n-hydroxy C 1-4 alkyl, -(CH 2 ) m -COOH, -(CH 2 ) m -COOR 3 .
  • Each m can be independently 0, 1, 2, or 3.
  • a pharmaceutical composition comprising a compound of the invention and a suitable excipient is provided.
  • a pharmaceutical combination comprising a compound of the invention and a second medicament is provided.
  • the second medicament is medically indicated for the treatment of multiple sclerosis, transplant rejection, acute respiratory distress syndrome or adult respiratory distress syndrome.
  • a method of activation or agonism of a sphingosine-1- phosphate receptor subtype 1 comprising contacting the receptor subtype 1 with a compound of claim 1 is provided.
  • the compound of claim 1 activates or agonizes the sphingosine-1 -phosphate receptor subtype 1 to a greater degree than the compound activates or agonizes a sphingosin-1 -phosphate receptor subtype 3.
  • a method of treatment of a malcondition in a patient for which activation or agonism of an SlPj receptor is medically indicated is provided.
  • selective activation or agonism of an SI Pj receptor such as with respect to an S1P 3 receptor, is medically indicated.
  • the malcondition comprises multiple sclerosis, transplant rejection, or acute respiratory distress syndrome.
  • a method for chiral synthesis of certain compounds including compounds of the invention is provided.
  • the invention provides certain intermediate compounds associated with such methods of chiral synthesis.
  • Certain embodiments of the present invention comprise a compound having the structure of Formula (I) or a pharmaceutically acceptable salt, ester, prodrug, homolog, tautomer, stereoisomer, or hydrate, or solvate thereof:
  • a dashed line signifies that a single bond or a double bond can be present, provided that there are two double bonds and three single bonds in the ring comprising A 1 , A , and A .
  • a , A , and A each independently can be CH or S or N; provided that one of A , A 2 , and A 3 is S.
  • R l can be di-substituted phenyl or di-substituted pyridinyl where the phenyl and pyridinyl substituents ca each be independently any of halo, nitro, cyano, perfluromethyl, fluorinated methyl, and C 1-4 -alkoxy.
  • R 1 is di-substituted phenyl, such phenyl is para- substituted with Ci-4-alkoxy.
  • R 2 can be
  • X can be -NR'R" or -OR'"
  • R' can be H, CM alkyl, n-hydroxy C M alkyl, -S0 2 -R 3 , or -CO-R 3 .
  • R" can be H, -S0 2 -R 5 , C alkyl optionally substituted with 1 or more R 4 , or a ring moiety optionally substituted with R 6 wherein such ring moiety is pyridinyl, oxazolyl, piperidinyl, cyclohexyl, mo holinyl, thiazolyl, pyrazolyl, pyrrolidinyl, imidazolyl, or phenyl.
  • R'" can be H, C alkyl, or -CO-R .
  • R' and R" taken together with the nitrogen atom to which they are bound can form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle is optionally singly or multiply substituted with substituents independently selected from the group consisting of -OH, oxo, -NH 2 , n-hydroxy-C ⁇ alkyl, -COOH, -(CH 2 ) M -COOH, - (CH 2 ) M -COOR 3 , -N(R 3 R 3 ), and -(CH 2 ) M -CO-N(R 7 R 7 ).
  • Each R 3 can be independently CM alkyl or H.
  • Each R 5 can be independently R 4 , C alkyl, C 3-6 cycloalkyl, or C alkyl optionally substituted with 1 or more R 4 .
  • Each R 6 can be independently halo, OH, -NH 2 , -NHR 3 , -N(R 3 R 3 ), -COOH, - COOR 3 , -NHCO-R 3 .
  • Each R 7 can be independently CM alkyl or H, or two R 7 taken together with the nitrogen atom to which they are bound can form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle can be optionally substituted with -OH, -NH 2 , -N(R 3 R 3 ), n-hydroxy CM alkyl, -(CH 2 ) M -COOH, -(CH 2 ) M -COOR 3 .
  • Each m can be independently 0, 1, 2, or 3.
  • the compounds of the invention have the structure of a specific of Formula I or a pharmaceutically acceptable salt, ester, prodrug, homolog, hydrate or solvate thereof.
  • the invention provides compounds which are substantially enantiomerically pure.
  • the compounds are enantiomerically pure with respect to a chiral carbon on an indanyl or tetrahydronaphthalenyl moiety.
  • the invention provides compounds which have an EC50 as an agonist of the wild type SIP receptor subtype 1 which is at least ten times smaller than the EC50 of such compound as an agonist of a mutant SIP receptor subtype 1 having a single mutation with respect to wild type SIP receptor subtype 1 such that the 101 st amino acid residue is changed from asparagine to alanine.
  • the invention provides compounds which have an EC 50 as an agonist of the wild type SIP receptor subtype 1 which is at least twenty times smaller than the EC50 of such compound as an agonist of a mutant SIP receptor subtype 1 having a single mutation with respect to wild type SIP receptor subtype 1 such that the 101 st amino acid residue is changed from asparagine to alanine.
  • the invention provides compounds which have a therapeutic index of at least 5 as measured in rats following 5 or 14 days of dosing of rats with the compound where the therapeutic index is calculated as a ratio of (i) the highest dose of such compound which achieves less than or equal to a ten percent increase in the ratio of lung to terminal body weight at the conclusion of such 5 or 14 days of dosing, to (ii) the dose of such compound achieving 50% lymphopenia in rats.
  • such therapeutic index is at least 10 and in certain embodiments the therapeutic index is at least 20.
  • the therapeutic index for a compound is at least five times greater than the therapeutic index for the enantiomer of such compound.
  • the invention provides compounds which have a therapeutic index of at least 5 as measured in rats following 5 or 14 days of dosing of rats with the compound where the therapeutic index is calculated as a ratio of (i) the highest dose of such compound which achieves less than or equal to a ten percent increase in the ratio of lung to terminal body weight at the conclusion of such 5 or 14 days of dosing, to (ii) the dose of such compound achieving 50% lymphopenia in rats.
  • such therapeutic index is at least 10 and in certain embodiments the therapeutic index is at least 20.
  • the therapeutic index for a compound is greater than the therapeutic index for the enantiomer of such compound.
  • the therapeutic index for a compound is at least 150% of the therapeutic index for the enantiomer of such compound.
  • the invention provides compounds where the structure of Formula I is selected from the roup consisting of formulas a-i through a-x:
  • the invention provides compounds where A 1 is S, in other embodiments the invention provides compounds where A is S and in other embodiments the
  • A is S. In certain embodiments A is N and A is CH
  • A is CH and in others A is N.
  • R is C 2 -4 alkyl; and Y is -CN, -CI, -O-R , or -CF 3 . In certain such embodiments R isopropyl or ethyl. In certain embodiments Y is -CN or -0-C 2 H 5 .
  • the invention provides compounds where R 2 is
  • the compound is substantially enantiomerically pure.
  • the invention provides compounds where Y is CI, in other embodiments the invention provides compounds where Y is CF 3 and in other embodiments the invention provides compounds where Y is CN.
  • the invention provides compounds where X is -NR'R", in other embodiments the invention provides compounds where X is -OR'". In certain embodiments the invention provides compounds where X is -OR'". In certain embodiments the invention provides compounds where X is -OH and in other embodiments the invention provides compounds where X is -OCO-R .
  • the invention provides compounds where R 3 is C 1-3 alkyl; in other embodiments the invention provides compounds where R' is H.
  • the invention provides compounds where R' is -COR 3 ; in other embodiments the invention provides compounds where R' is S0 2 -R 3 . In certain embodiments the invention provides compounds where R" is H.
  • the invention provides compounds where R" is -S0 2 -R 5 ; in other embodiments the invention provides compounds where R" is C 1-4 alkyl where the Ci. 4 alkyl is optionally substituted with 1 or more substituents defined by R 4 .
  • the invention provides compounds where R" is -(CR ⁇ n -R 4 and each R a and each R b can be independently any of H, hydroxyl and methyl or where R a and R b are bound to the same carbon they can be taken together to form oxo (i.e. with the carbon to which they are bound forming a carbonyl moiety).
  • n can be 0, 1, 2, or 3 and in certain embodiments n is 2.
  • R can be-OH, -NH 2 , - NHR 3 , -N(R 7 R 7 ), or -COOH.
  • the invention provides compounds where R 5 is C alkyl optionally substituted with 1 or more R 4 .
  • the invention provides compounds where R 4 is OH; in other embodiments the invention provides compounds where R 4 is Ci_ 3 alkoxy.
  • the invention provides compounds where R 5 is (CH 2 ) 2 -OR 3 .
  • the invention provides compounds where Y is CN and X is -NH-S0 2 -R . In certain embodiments the invention provides compounds where R is - C 2 H 5 -N((R 7 R 7 ) or -CH 2 -CO-N(R 7 R 7 ). In certain embodiments the invention provides compounds where Y is CN and X is -NH-CO-N(R 7 R 7 ).
  • X is -NH 2 and in certain of such embodiments Y is CN.
  • the invention provides compounds having the following formula:
  • a 2 is CH or N
  • R m is H, -S0 2 -R 5 , or C H alkyl optionally substituted with 1 or more R 4 ;
  • each R 4 is independently H, OH, oxo, NH 2 , -COOH, S0 2 -R 3 , -N(R 3 )-S0 2 -R 3 , - COOR 3 , -C0-N(R 7 R 7 ), N(R 7 R 7 ), C 1-3 alkyl, C 1-3 alkoxy; or a ring moiety selected from pyridinyl, oxazolyl, pyrrolidinyl, or imidazolyl.
  • the invention provides compounds having the following formula:
  • the invention provides compounds where R M is -S0 2 -R 5 .
  • R 5 is NH 2 , or C alkyl optionally substituted with 1 or more R 4
  • R 4 is OH, C 1-3 alkoxy, -COOH, -COOR 3 , CO-N(R 7 R 7 ) or N(R 7 R 7 ).
  • the invention provides compounds where R M is is C 1-4 alkyl optionally substituted with 1 or more R 4 . In certain embodiments, the invention provides compounds where at least one of the 1 or more R 4 is oxo. In certain embodiments, the invention provides compounds where one of the 1 or more R 4 is OH, or C 1-3 alkoxy.
  • the invention provides one or more of compounds 1-227:
  • the invention provides a compound selected from compounds 43, 46, 47, 56, 58, 166, 172, 186 and 246 or any pharmaceutically acceptable salt, ester, tautomer, stereoisomer, solvate, hydrate, homolog, or prodrug thereof.
  • the invention provides compound 43, 46, 166 or 246 or any pharmaceutically acceptable salt, ester, tautomer, solvate, hydrate, homolog, or prodrug thereof.
  • the invention provides one or more of compounds 307- 314:
  • the invention provides one or more of compounds 228- 306:
  • an invention compound of Formula I wherein the compound has at least one chiral center and is substantially enantiomerically pure.
  • a pharmaceutical composition comprising an invention compound of Formula I and a suitable excipient is provided.
  • a pharmaceutical combination comprising an invention compound and a second medicament is provided.
  • a pharmaceutical combination comprising an invention compound and a second medicament is provided wherein the second medicament is medically indicated for the treatment of multiple sclerosis, transplant rejection, or adult respiratory distress syndrome.
  • a method of use of an invention compound for preparation of a medicament is provided.
  • a method of activation or agonism of a sphingosine-1 - phosphate receptor subtype 1 by contacting the receptor subtype 1 with an effective amount of an invention compound in certain embodiments, a method of activation or agonism of a sphingosine-1 -phosphate receptor subtype 1 by contacting the receptor subtype 1 with an effective amount of an invention compound is provided, wherein the compound activates or agonizes the sphingosine-1 -phosphate receptor subtype 1 to a greater extent than the compound activates or agonizes a sphingosine-1 -phosphate receptor subtype 3.
  • a method of activation or agonism of a sphingosine-1 -phosphate receptor subtype 1 by contacting the receptor subtype 1 with an effective amount of an invention compound is provided, wherein the sphingosine-1 -phosphate receptor subtype 1 is disposed within a living mammal.
  • a method for treatment of a malcondition in a patient for which activation or agonism of an sphingosine-1 -phosphate receptor subtype 1 is medically indicated by administering an effective amount of an invention compound to the patient at a frequency and for a duration of time sufficient to provide a beneficial effect to the patient.
  • a method for treatment of a malcondition in a patient for which activation or agonism of an sphingosine-1 -phosphate receptor subtype 1 is medically indicated, by administering an effective amount of an invention compound to the patient at a frequency and for a duration of time sufficient to provide a beneficial effect to the patient, wherein selective activation or agonism of an SIP subtype 1 receptor with respect to other subtypes of SIP receptor is medically indicated.
  • a method for treatment of a malcondition in a patient for which activation or agonism of an sphingosine-1 -phosphate receptor subtype 1 is medically indicated by administering an effective amount of an invention compound to the patient at a frequency and for a duration of time sufficient to provide a beneficial effect to the patient, wherein the malcondition comprises rejection of transplanted organs or tissue; graft-versus-host diseases brought about by transplantation; autoimmune syndromes including rheumatoid arthritis; acute respiratory distress syndrome; adult respiratory distress syndrome; influenza; cancer; systemic erythematosus; Hashimoto's thyroiditis; lymphocytic thyroiditis; multiple sclerosis; myasthenia gravis; type I and II diabetes; uveitis; posterior uveitis; uveitis associated with Behcet's disease; uveomeningitis syndrome; allergic encephalomyelitis; chronic allograft vasculopathy; post-infectious autoimmune
  • the malcondition is one or more of rejection of transplanted organs or tissue; graft- versus-host diseases brought about by transplantation; autoimmune syndromes including rheumatoid arthritis, multiple sclerosis, myasthenia gravis; pollen allergies; type I diabetes; prevention of psoriasis; Crohn's disease; ulcerative colitis, acute respiratory distress syndrome; adult respiratory distress syndrome; influenza; post-infectious autoimmune diseases including rheumatic fever and post-infectious glomerulonephritis; and metastasis of carcinoma.
  • the malcondition is one of influenza, ulcerative colitis, multiple sclerosis, transplant rejection, acute respiratory distress syndrome or adult respiratory distress syndrome.
  • methods are provided for use of an invention compound for preparation of a medicament adapted for treatment of a disorder or a malcondition wherein activation or inhibition of a sphingosine-1 -phosphate receptor subtype 1 is medically indicated.
  • the invention provides a method for the chiral synthesis of a compound comprising an indane moiety having a chiral carbon in the five-member ed ring of the indane moiety where the compound is enantiomerically enriched with respect to the chiral carbon.
  • the method of the invention provides the steps of
  • step (iii) forming the chiral center at the indane moiety carbon previously bound to the oxo group by either reacting such compound with a suitable reducing agent along with the chiral reagent in step (ii) or reacting the result of the reaction of such compound with a suitable reducing agent.
  • R is t-butyl, sec-butyl, isopropyl, cyclopropyl, adamantyl, C 3- 6 branched alkyl, or optionally bridged C 3-8 cycloalkyl.
  • the chiral reagent is the Corey Bakshita Shibata-oxazaborolidine and the compound comprising an indane moiety is enantiomerically enriched with respect to a carbon-oxygen bond on a ring carbon of the five-membered ring of the indane moiety.
  • a suitable reducing reagent includes a borohydride such as BH 3 -DMS or NaB3 ⁇ 4.
  • the chiral reagent is (i?)-(-)-(2)-methyl-CBS- oxazaborolidine or (5 -(-)-(2)-methyl-CBS-oxazaborolidine.
  • step (i) the compound comprising an indane moiety provided in step (i) is contacted with the chiral reagent to form in step (ii) Formula VI-R or VI-S:
  • Z is CI, Br or I.
  • the method further comprises the step of protecting the hydroxy group of Formula VI-R or VI-S by treating Formula VI-R or VI-S with a protecting agent to form Formula -R or Vla-S:
  • Protecting groups can render chemical functionality inert to specific reaction conditions and can be appended to and removed from such functionality in a molecule without substantially damaging the remainder of the molecule. Practitioners in the art would be familiar with suitable protecting groups for use in the synthetic methods of the invention. See, e.g., Greene and Wuts, Protective Groups in Organic Synthesis, 2 nd ed., John Wiley & Sons, New York, 1991. In certain embodiments such protecting agent is t-butyldimethylsilyl chloride (TBSC1).
  • TBSC1 t-butyldimethylsilyl chloride
  • the method further comprises the step of reacting Formula VTa-R or Vla-S with boronic acid or bis(pinacolato)diboron to form a boronic acid or boronate ester of F
  • step (i) the compound comprising an indane moiety provided in step (i) is contacted with the chiral reagent to form in step (ii) Form -R or VII-S:
  • Z is CI, Br or I.
  • the method further comprises the step of contacting Formula VIII-R or VIII-S with 1 ,4-dioxane in the presence of an acid to form Formula Vlb- R or VIb-S or Formul -R or IX-S:
  • the method further comprises the step of protecting the amino group by treating Formula IX-R or IX-S with a protecting agent to form Formula IXa- R or IXa-S:
  • the protecting agent is di-tert-butyldicarbonate.
  • the method further comprises the step of reacting Formula IXa-R or IXa-S with boronic acid or bis(pinacolato)diboron to form a boronic acid or boronate ester of
  • the method further comprises the step of reacting Formula VIb-R, Formula VIb-S, Formula IXb-R or Formula IXb-S with Formula XI:
  • each A and each A is independently N, or CH;
  • R is di-substituted phenyl or di-substituted pyridinyl where the phenyl and pyridinyl substituents are each independently selected from the group consisting of halo, nitro, cyano, perfluromethyl, fluorinated methyl, and Ci-4-alkoxy; provided that if R 1 is di- substituted phenyl, such phenyl is para-substituted with d-4-alkoxy; and
  • X is NH or O.
  • R 1 is di-substituted phenyl where the phenyl substituents are F and Y, wherein Y is -CN, -CI, or -CF3. In still further embodiment Y is -CN.
  • the method further comprises the step of reacting Formula XII-R -S with /PrOH in the presence of NaO/Pr to from Formula XIII-R or XIH-S:
  • the method further comprises the step of deprotecting the hydroxyl group wherein X is O, or the amino group wherein X is NH, by treating Formula XIII-R or XIII-S with a deprotecting agent.
  • the method further comprises the step of converting the deprotected amino group to a secondary amine.
  • a 1 is N and A 2 is N.
  • Formula XI is prepared following the process comprising the step of a) treating a di-substituted benzaldehyde with potassium phosphate monobasic to form a di-substituted benzoic acid; b) contacting the di-substituted benzoic acid with H 2 NNHCSNH 2 to form an amino- 1,3-4-thiadizole having a di-substituted phenyl group substituted on the thiadiazole moiety; and c) treating the amino- 1,3-4-thiadizole in step b) with a mixture of copper bromide and isoamylnitrite.
  • a 1 is N and A 2 is CH.
  • Formula XI is prepared following the process comprising the step of
  • a 1 is CH and A 2 is N.
  • Formula XI is prepared following the process comprising the step of
  • the method of the invention provides the steps of
  • step (iii) forming a chiral center at the indane moiety carbon previously bound to the oxo group by either reacting such compound with a suitable reducing agent along with the chiral reagent in step (ii) or reacting the result of the reaction of such compound with a suitable reducing agent.
  • the chiral reagent is a Corey Bakshita Shibata- oxazaborolidine and X is -OR'".
  • the chiral reagent is (R)-(-)-(2)- methyl-CBS-oxazaborolidine or (S)-(-)-(2)-methyl-CBS-oxazaborolidine.
  • a suitable reducing reagent includes a borohydride such as BH 3 -DMS or NaB3 ⁇ 4.
  • step (iii) forming a chiral center at the indane moiety carbon previously bound to the oxo group by either reacting such compound with a suitable reducing agent along with the chiral reagent in step (ii) or reacting the result of the reaction of such compound with a suitable reducing agent.
  • step (iii) forming a chiral center at the indane moiety carbon previously bound to the oxo group by either reacting such compound with a suitable reducing agent along with the chiral reagent in step (ii) or reacting the result of the reaction of such compound with a suitable reducing agent.
  • the invention provides a method for chiral synthesis of a chiral compound comprising an indane moiety having a chiral carbon in the five-membered ring of the indane moiety or a chiral compound comprising an oxadiazole- indane moiety having a chiral carbon in the five-membered ring of the indane moiety where the chiral compound has an enantiomeric enrichment of at least 75%, 85%, 90%, 95%, 98%, or 99%.
  • the invention provides a method for synthesis of a chiral compound of the invention having an enantiomeric enrichment of at least 75%, 85%, 90%, 95%, 98%, or 99%.
  • a method for the synthesis of a compound comprising an indane moiety having a chiral carbon in the five-membered ring of the indane moiety where the compound is enantiomerically enriched with respect to the chiral carbon is provided.
  • a method comprising a step of providing a compound of the structures described herein is provided.
  • the invention provides a method for the chiral synthesis of a compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six- membered saturated ring of the tetrahydronaphthalene moiety where the compound is enantiomerically enriched with respect to the chiral carbon, the method comprising the steps of
  • the chiral reagent is RuCl(p-cymene)[(i?, ⁇ )-Ts-DPEN] or RuCl(p-cymene)[(S, ⁇ -Ts-DPEN] .
  • the compound comprising a tetrahydronaphthalene moiety provided in step (i) is contacted with the chiral reagent to form in step (ii)
  • the method further comprises the step of reversing the chiral configuration of the chiral carbon in the six-membered saturated ring of the tetrahydronaphthalene moiety that was previously bound to the oxo group by treating Formula XIV-R or XIV-S with diphenylphosphoryl azide (DPP A) to form an azido tetrah drona hthalene of Formula XV-S r -R: where the azido substituent in the six-membered saturated ring of the tetrahydronaphthalene moiety replacing the hydroxy substituent and the chiral carbon that is bound to the azido substituent has a reverse chiral configuration of the chiral carbon when it was previously bound to the hydroxy substituent .
  • DPP A diphenylphosphoryl azide
  • the method further comprises the step of converting the azido tetrahydronaphthalene of Formula XV-S or XV-R to an amine substituted tetrahydronaphthalene of Formula XVI-S or XVI-R: wherein X m is NH-PG, NH 2 or NRTT; R' is H, C 1-4 alkyl, n-hydroxy C alkyl, -S0 2 -R 3 , or -CO-R 3 ; R" is H, -SO2-R 5 , C alkyl optionally substituted with 1 or more R 4 , or a ring moiety optionally substituted with R 6 wherein such ring moiety is pyridinyl, oxazolyl, piperidinyl, cyclohexyl, morpholinyl, thiazolyl, pyrazolyl, pyrrolidinyl, imidazolyl, or phenyl; and PG
  • the step of converting the azido tetrahydronaphthalene of Formula XV-S or XV-R to an amine substituted tetrahydronaphthalene of Formula XVI-S or XVI-R comprises contacting the azido tetrahydronaphthalene of Formula XV-S or XV-R with a carbonate ester.
  • the carbonate ester is di-tert-butyl dicarbonate.
  • the method further comprises the step of reacting Formula XVI-S or XVI-R with a boronic acid to form a boronic acid of Formula XVII-S or XVII-R:
  • the boronic acid is pinacolborane or bis(pinacolato)diboron.
  • the method further comprises the step of forming a substituted 1,3,4-thiadiazole on the tetrahydronaphthalene moiety by reacting an intermediate of XVII-S or XVII-R with a substituted 2-bromo- 1,3,4-thiadiazole of Formula XVTV and a coupling reagent to form a compound of Formula XW-S or XW-R:
  • the coupling reagent comprises a palladium. In certain embodiments, the coupling reagent comprises tetrakis (triphenylphosphine) palladium.
  • the method provides compound of Formula XW-S -R:
  • the invention provides the method of synthesizing a chiral compound where the compound provided in step (i) is
  • the invention provides the method where the resulting compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six- membered saturated ring of the tetrahydronaphthalene moiety is enantiomerically enriched at least 90%. In certain of such embodiments the resulting compound is enantiomerically enriched at least 95%. In certain of such embodiments the resulting compound is enantiomerically enriched at least 98%. In certain of such embodiments the resulting compound is enantiomerically enriched at least 99%.
  • the invention provides compounds which can be intermediates in the herein described methods for chiral syntheses.
  • the invention provides one or more of the following intermediate compounds:
  • the invention provides a method for the synthesis of a compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six-membered saturated ring of the tetrahydronaphthalene moiety where the compound is enantiomerically enriched with respect to the chiral carbon, the method comprising a step of providing an intermediate compound.
  • the invention provides a method for chiral synthesis of a chiral compound comprising an tetrahydronaphthalene moiety having a chiral carbon in the six-membered ring of the tetrahydronaphthalene moiety or a chiral compound comprising an 1,3,4-thiadiazole-tetrahydronaphthalene moiety having a chiral carbon in the six-membered ring of the tetrahydronaphthalene moiety where the chiral compound has an enantiomeric enrichment of at least 75%, 85%, 90%, 95%, 98%, or 99%.
  • the invention provides a method for synthesis of a chiral compound of the invention having an enantiomeric enrichment of at least 75%, 85%, 90%, 95%, 98%, or 99%.
  • a method for the synthesis of a compound comprising an tetrahydronaphthalene moiety having a chiral carbon in the six-membered ring of the tetrahydronaphthalene moiety where the compound is enantiomerically enriched with respect to the chiral carbon is provided.
  • a method comprising a step of providing a compound of the structures described herein is provided.
  • mammals as in the subject of the treatment
  • Mammals include, for example, humans; non-human primates, e.g. apes and monkeys; cattle; horses; sheep; and goats.
  • Non-mammals include, for example, fish and birds.
  • SlP t refers to subtype 1 of a sphingosine-1 -phosphate receptor, while other sphingosine-1 -phosphate receptor subtypes are referred to in a corresponding manner, for example, sphingosine-1 -phosphate receptor subtype 3 is referred to as " S1P 3 ".
  • a "receptor”, as is well known in the art, is a biomolecular entity usually comprising a protein that specifically binds a structural class of ligands or a single native ligand in a living organism, the binding of which causes the receptor to transduce the binding signal into another kind of biological action, such as signaling a cell that a binding event has occurred, which causes the cell to alter its function in some manner.
  • An example of transduction is receptor binding of a ligand causing alteration of the activity of a "G-protein" in the cytoplasm of a living cell.
  • SIPj compound or "SIPi agonist” or “SlPj activator” or “S inhibitor” or “SI Pi antagonist” as the terms are used herein refer to compounds that interact in some way with the SIP receptor subtype 1. They can be agonist or activators, or they can be antagonists or inhibitors.
  • An "SIPi compound” of the invention can be selective for action on subtype 1 of the SIP receptor family; for example a compound of the invention can act at a lower concentration on subtype 1 of the S IP receptor family than on other subtypes of the SIP receptor family; more specifically, an "SIPi compound” of the invention can selectively act on subtype 1 receptors compared to its action on subtype 3, or "S1P 3 " receptors.
  • compounds of the invention are orthostatic agonists.
  • compounds of the invention are allostenc agonists.
  • Receptor agonists may be classified as either orthosteric or allosteric.
  • An orthosteric agonist binds to a site in the receptor that significantly overlaps with the binding of the natural ligand and replicates the key interactions of the natural ligand with the receptor.
  • An orthosteric agonist will activate the receptor by a molecular mechanism similar to that of the natural ligand, will be competitive for the natural ligand, and will be competitively antagonized by pharmacological agents that are competitive antagonists for the natural ligand.
  • Allosteric agonist binds to a site in the receptor that makes some significant interactions that are partly or wholly non-overlapping with the natural ligand. Allosteric agonists are true agonists and not allosteric potentiators. Consequently, they activate receptor signaling alone and without a requirement for a sub-maximal concentration of the natural ligand. Allosteric agonists may be identified when an antagonist known to be competitive for the orthosteric ligand shows non-competitive antagonism. The allosteric agonist site can also be mapped by receptor mutagenesis.
  • Orthosteric agonists may destabilize GPCR structure and conformation, while allosteric agonists may either stabilize or destabilize GPCR structure and conformation. Allosteric agonists, by virtue of their different interactions with receptor, may be pharmaceutically useful because the allosteric site may confer additional opportunities for agonist potency and selectivity within a related family of receptor subtypes that share a similar orthosteric ligand. In addition, the allosteric site may require very different physical and chemical properties of an agonist compared to the orthosteric ligand. These chemico- physical properties, which include hydrophobicity, aromaticity, charge distribution and solubility may also provide advantages in generating agonists of varying pharmacokinetic, oral bioavailability, distributional and metabolism profiles that facilitate the development of effective pharmaceutical substances.
  • substantially as the term is used herein means completely or almost completely; for example, a composition that is "substantially free” of a component either has none of the component or contains such a trace amount that any relevant functional property of the composition is unaffected by the presence of the trace amount, or a compound is "substantially pure” is there are only negligible traces of impurities present.
  • Substantially enantiomerically pure means a level of enantiomeric enrichment of one enantiomer with respect to the other enantiomer of at least 90%, 95%, 98%, 99%, 99.5% or 99.9%.
  • Treating or “treatment” within the meaning herein refers to an alleviation of symptoms associated with a disorder or disease, or inhibition of further progression or worsening of those symptoms, or prevention or prophylaxis of the disease or disorder.
  • the expression "effective amount”, when used to describe use of a compound of the invention in providing therapy to a patient suffering from a disorder or malcondition mediated by a sphingosine-l-phospate receptor of subtype 1 refers to the amount of a compound of the invention that is effective to bind to as an agonist or as an antagonist a SIPi receptor in the individual's tissues, wherein the SlPj is implicated in the disorder, wherein such binding occurs to an extent sufficient to produce a beneficial therapeutic effect on the patient.
  • an "effective amount” or a “therapeutically effective amount” of a compound of the invention refers to an amount of the compound that alleviates, in whole or in part, symptoms associated with the disorder or condition, or halts or slows further progression or worsening of those symptoms, or prevents or provides prophylaxis for the disorder or condition.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result by acting as an agonist of sphingosine-1 -phosphate receptor subtype 1 (SI Pi) activity.
  • SI Pi sphingosine-1 -phosphate receptor subtype 1
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of compounds of the invention are outweighed by the therapeutically beneficial effects.
  • a therapeutically effective amount of an SI Pi agonist of the invention is an amount sufficient to control the malcondition, to mitigate the progress of the malcondition, or to relieve the symptoms of the malcondition.
  • malconditions that can be so treated include multiple sclerosis, transplant rejection, adult respiratory distress syndrome.
  • Diseases, disorders and conditions which may be treated by compounds of the invention include rejection of transplanted organs or tissue; graft- versus-host diseases brought about by transplantation; autoimmune syndromes including rheumatoid arthritis; acute respiratory distress syndrome; adult respiratory distress syndrome; influenza; cancer; systemic erythematosus; Hashimoto's thyroiditis; lymphocytic thyroiditis; multiple sclerosis; myasthenia gravis; type I and II diabetes; uveitis; posterior uveitis; uveitis associated with Behcet's disease; uveomeningitis syndrome; allergic encephalomyelitis; chronic allograft vasculopathy; post-infectious autoimmune diseases including rheumatic fever and postinfectious glomerulonephritis; inflammatory and hyperproliferative skin diseases; cutaneous manifestations of immunologically-mediated disorders; psoriasis; atopic dermatitis; osteomyelitis; contact dermatitis
  • compounds of Formula I-R or I-S are also useful, in combination with one or several immunosuppressant agents, for the treatment of diseases, disorders and conditions associated with an activated immune system and selected from the list as above- mentioned.
  • said immunosuppressant agent is selected from the group comprising or consisting of cyclosporin, daclizumab, basiliximab, everolimus, tacrolimus (FK506), azathiopirene, leflunomide, 15- deoxyspergualin, or other immunosuppressant drugs
  • the isomers resulting from the presence of a chiral center comprise a pair of non-superimposable isomers that are called "enantiomers.”
  • Single enantiomers of a pure compound are optically active, i.e., they are capable of rotating the plane of plane polarized light.
  • Single enantiomers are designated according to the Cahn-Ingold-Prelog system.
  • isolated optical isomer means a compound which has been substantially purified from the corresponding optical isomer(s) of the same formula.
  • the isolated isomer is at least about 80%, more preferably at least 90% pure, even more preferably at least 98% pure, most preferably at least about 99% pure, by weight.
  • the preferred compounds of the present invention have a particular spatial arrangement of substituents on the aromatic rings, which is related to the structure activity relationship demonstrated by the compound class. Often such substitution arrangement is denoted by a numbering system; however, numbering systems are often not consistent between different ring systems. In six-membered aromatic systems, the spatial arrangements are specified by the common nomenclature "para” for 1,4-substitution, "meta” for 1,3 -substitution and "ortho" for 1,2-substitution as shown below.
  • substituted refers to an organic group as defined herein in which one or more bonds to a hydrogen atom contained therein are replaced by one or more bonds to a non-hydrogen atom such as, but not limited to, a halogen (i.e., F, CI, Br, and I); an oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy groups, aralkyloxy groups, oxo(carbonyl) groups, carboxyl groups including carboxylic acids, carboxylates, and carboyxlate esters; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfoxide groups, sulfone groups, sulfonyl groups, and sulfonamide groups; a nitrogen atom in groups such as amines, hydroxylamines, nitriles, nitro groups, N-oxides, hydrazides, azides,
  • Non-limiting examples of substituents that can be bonded to a substituted carbon (or other) atom include F, CI, Br, I, OR', OC(0)N(R') 2 , CN, CF 3 , OCF 3 , R', O, S, C(O), S(O), methylenedioxy, ethylenedioxy, N(R') 2 , SR', SOR', S0 2 R ⁇ S0 2 N(R') 2 , S0 3 R*, C(0)R', C(0)C(0)R', C(0)CH 2 C(0)R', C(S)R*, C(0)OR', OC(0)R', C(0)N(R') 2 , OCiOWh, C(S)N(R') 2 , (CH 2 ) 0-2 NHC(O)R', (CH 2 ) 0- 2 N(R')N(R') 2 , N(R')N(R')C(0)R', N(R')N(R
  • Substituted alkyl, alkenyl, alkynyl, cycloalkyl, and cycloalkenyl groups as well as other substituted groups also include groups in which one or more bonds to a hydrogen atom are replaced by one or more bonds, including double or triple bonds, to a carbon atom, or to a heteroatom such as, but not limited to, oxygen in carbonyl (oxo), carboxyl, ester, amide, imide, urethane, and urea groups; and nitrogen in imines, hydroxyimines, oximes, hydrazones, amidines, guanidines, and nitriles.
  • the substituents of the substituted groups can further be substituted with alkyl, alkenyl, cycloalkyl, aryl, heteroaryl, and alkynyl groups as defined herein, which can themselves be further substituted.
  • alkyl alkenyl, cycloalkyl, aryl, heteroaryl, and alkynyl groups as defined herein, which can themselves be further substituted.
  • an CM alkyl group can be substituted with an amide, and the amide can further be substituted with another C alkyl, which can further be substituted.
  • Substituted ring groups such as substituted aryl, heterocyclyl and heteroaryl groups also include rings and fused ring systems in which a bond to a hydrogen atom is replaced with a bond to a carbon atom. Therefore, substituted aryl, heterocyclyl and heteroaryl groups can also be substituted with alkyl, alkenyl, cycloalkyl, aryl, heteroaryl, and alkynyl groups as defined herein, which can themselves be further substituted.
  • heteroatoms refers to non-carbon and non-hydrogen atoms, capable of forming covalent bonds with carbon, and is not otherwise limited. Typical heteroatoms are N, O, and S.
  • sulfur (S) When sulfur (S) is referred to, it is understood that the sulfur can be in any of the oxidation states in which it is found, thus including sulfoxides (R-S(O)- R') and sulfones (R-S(0) 2 -R'), unless the oxidation state is specified; thus, the term “sulfone” encompasses only the sulfone form of sulfur; the term “sulfide” encompasses only the sulfide (R-S-R 1 ) form of sulfur.
  • heteroatoms selected from the group consisting of O, NH, NR' and S or "[variable] is O, S . . .” are used, they are understood to encompass all of the sulfide, sulfoxide and sulfone oxidation states of sulfur.
  • Alkyl groups include straight chain and branched alkyl groups and cycloalkyl groups having from 1 to about 20 carbon atoms (Ci -2 o alkyl), and typically from 1 to 12 carbons (C 1-12 alkyl)or, in some embodiments, from 1 to 8 carbon atoms (C 1-8 alkyl) or, in some embodiments, from 1 to 4 carbon atoms (C1-4 alkyl) or, in some embodiments, from 1 to 3 carbon atoms (Ci -3 alkyl).
  • straight chain alkyl groups include, but are not limited to methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, and n-octyl groups.
  • branched alkyl groups include, but are not limited to, isopropyl, iso-butyl, sec- butyl, t-butyl, neopentyl, isopentyl, and 2,2-dimethylpropyl groups.
  • substituted alkyl groups can be substituted one or more times with any of the groups listed above, for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups.
  • the group "n-hydroxy C ⁇ alkyl” represents an C 1- alkyl substituted with a terminal hydroxy group.
  • Cycloalkyl groups are alkyl groups forming a ring structure, which can be substituted or unsubstituted.
  • Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl groups.
  • the cycloalkyl group has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms range from 3 to 5, 3 to 6, or 3 to 7.
  • Cycloalkyl groups further include polycyclic cycloalkyl groups such as, but not limited to, norbomyl, adamantyl, bornyl, camphenyl, isocamphenyl, and carenyl groups, and fused rings such as, but not limited to, decalinyl, and the like. Cycloalkyl groups also include rings that are substituted with straight or branched chain alkyl groups as defined above.
  • Representative substituted cycloalkyl groups can be mono-substituted or substituted more than once, such as, but not limited to, 2,2-, 2,3-, 2,4- 2,5- or 2,6-disubstituted cyclohexyl groups or mono-, di- or tri-substituted norbomyl or cycloheptyl groups, which can be substituted with, for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups.
  • carbocyclic and “carbocycle” denote a ring structure wherein the atoms of the ring are carbon.
  • the carbocycle has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms is 4, 5, 6, or 7.
  • the carbocyclic ring can be substituted with as many as N substituents wherein N is the size of the carbocyclic ring with for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups.
  • (Cycloalkyl)alkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of the alkyl group is replaced with a bond to a cycloalkyl group as defined above.
  • Alkenyl groups include straight and branched chain and cyclic alkyl groups as defined above, except that at least one double bond exists between two carbon atoms. Thus, alkenyl groups have from 2 to about 20 carbon atoms, and typically from 2 to 12 carbons or, in some embodiments, from 2 to 8 carbon atoms.
  • cycloalkenyl alone or in combination denotes a cyclic alkenyl group wherein at least one double bond is present in the ring structure.
  • Cycloalkenyl groups include cycloalkyl groups having at least one double bond between two adjacent carbon atoms.
  • cycloalkenyl groups include but are not limited to cyclohexenyl, cyclopentenyl, and cyclohexadienyl groups.
  • (Cycloalkenyl)alkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of the alkyl group is replaced with a bond to a cycloalkenyl group as defined above.
  • Alkynyl groups include straight and branched chain alkyl groups, except that at least one triple bond exists between two carbon atoms.
  • alkynyl groups have from 2 to about 20 carbon atoms, and typically from 2 to 12 carbons or, in some embodiments, from 2 to 8 carbon atoms. Examples include, but are not limited to -C ⁇ CH, -C ⁇ (CH 3 ), - C ⁇ C(CH 2 CH 3 ), -CH 2 C ⁇ CH, -CH 2 C ⁇ C(CH 3 ), and -CH 2 C ⁇ C(CH 2 C3 ⁇ 4), among others.
  • Aryl groups are cyclic aromatic hydrocarbons that do not contain heteroatoms.
  • aryl groups include, but are not limited to, phenyl, azulenyl, heptalenyl, biphenyl, indacenyl, fluorenyl, phenanthrenyl, triphenylenyl, pyrenyl, naphthacenyl, chrysenyl, biphenylenyl, anthracenyl, and naphthyl groups.
  • aryl groups contain 6-14 carbons in the ring portions of the groups.
  • aryl groups includes groups containing fused rings, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like), and also includes substituted aryl groups that have other groups, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups, bonded to one of the ring atoms.
  • substituted aryl groups can be mono-substituted or substituted more than once, such as, but not limited to, 2-, 3-, 4-, 5-, or 6-substituted phenyl or naphthyl groups, which can be substituted with groups including but not limited to those listed above.
  • Aralkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to an aryl group as defined above.
  • Representative aralkyl groups include benzyl and phenylethyl groups and fused (cycloalkylaryl)alkyl groups such as 4-ethyl-indanyl.
  • aryl moiety or the alkyl moiety or both are optionally substituted with other groups, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups.
  • Aralkenyl group are alkenyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to an aryl group as defined above.
  • Heterocyclyl groups include aromatic and non-aromatic ring compounds (heterocyclic rings) containing 3 or more ring members, of which one or more is a heteroatom such as, but not limited to, N, O, S, or P.
  • heterocyclyl groups include 3 to 20 ring members, whereas other such groups have 3 to 15 ring members.
  • At least one ring contains a heteroatom, but every ring in a polycyclic system need not contain a heteroatom.
  • a dioxolanyl ring and a benzdioxolanyl ring system are both heterocyclyl groups within the meaning herein.
  • a heterocyclyl group designated as a C 2 -heterocyclyl can be a 5-membered ring with two carbon atoms and three heteroatoms, a 6-membered ring with two carbon atoms and four heteroatoms and so forth.
  • a C4-heterocyclyl can be a 5-membered ring with one heteroatom, a 6-membered ring with two heteroatoms, and so forth.
  • the number of carbon atoms plus the number of heteroatoms sums up to equal the total number of ring atoms.
  • a saturated heterocyclic ring refers to a heterocyclic ring containing no unsaturated carbon atoms.
  • heterocyclyl group includes fused ring species including those having fused aromatic and non-aromatic groups.
  • the phrase also includes polycyclic ring systems containing a heteroatom such as, but not limited to, quinuclidyl and also includes heterocyclyl groups that have substituents, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups, bonded to one of the ring members.
  • a heterocyclyl group as defined herein can be a heteroaryl group or a partially or completely saturated cyclic group including at least one ring heteroatom.
  • Heterocyclyl groups include, but are not limited to, pyrrolidinyl, furanyl, tetrahydrofuranyl, dioxolanyl, piperidinyl, piperazinyl, morpholinyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl, dihydrobenzofuranyl, indolyl, dihydroindolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, i
  • thianaphthalenyl purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, quinoxalinyl, and quinazolinyl groups.
  • Heterocyclyl groups can be substituted.
  • Representative substituted heterocyclyl groups can be 1 mono-substituted or substituted more than once, including but not limited to, rings containing at least one heteroatom which are mono, di, tri, tetra, penta, hexa, or higher-substituted with substituents such as those listed above, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, and alkoxy groups.
  • Heteroaryl groups are aromatic ring compounds containing 5 or more ring members, of which, one or more is a heteroatom such as, but not limited to, N, O, and S.
  • a heteroaryl group designated as a C 2 -heteroaryl can be a 5-membered ring with two carbon atoms and three heteroatoms, a 6-membered ring with two carbon atoms and four heteroatoms and so forth.
  • a C4-heteroaryl can be a 5-membered ring with one heteroatom, a 6-membered ring with two heteroatoms, and so forth. The number of carbon atoms plus the number of heteroatoms sums up to equal the total number of ring atoms.
  • Heteroaryl groups include, but are not limited to, groups such as pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, 1 azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, : isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, and
  • heteroaryl and heteroaryl groups include fused ring compounds such as wherein at least one ring, but not necessarily all rings, are aromatic, including !tetrahydroquinolinyl, tetrahydroisoquinolinyl, indolyl and 2,3-dihydro indolyl.
  • the term also includes heteroaryl groups that have other groups bonded to one of the ring members, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups.
  • Representative substituted heteroaryl groups can be substituted one or more times with groups such as those listed above.
  • aryl and heteroaryl groups include but are not limited to phenyl, biphenyl, indenyl, naphthyl (1-naphthyl, 2-naphthyl), N-hydroxytetrazolyl, N- hydroxytriazolyl, N-hydroxyimidazolyl, anthracenyl (1-anthracenyl, 2-anthracenyl, 3- anthracenyl), thiophenyl (2-thienyl, 3-thienyl), furyl (2-furyl, 3-furyl) , indolyl, oxadiazolyl, isoxazolyl, quinazolinyl, fluorenyl, xanthenyl, isoindanyl, benzhydryl ⁇ acridinyl, thiazolyl, pyrrolyl (2 -pyrrolyl), pyrazolyl (3-pyrazolyl), imidazolyl (1 -imididazolyl
  • Heterocyclylalkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to a heterocyclyl group as defined above.
  • Representative heterocyclyl alkyl groups include, but are not limited to, furan-2-yl methyl, furan-3-yl methyl, pyridine-2-yl methyl (a-picolyl), pyridine-3-yl methyl ( ⁇ -picolyl), pyridine-4-yl methyl ( ⁇ -picolyl), tetrahydrofuran-2-yl ethyl, and indol-2-yl propyl.
  • Heterocyclylalkyl groups can be substituted on the heterocyclyl moiety, the alkyl moiety, or both.
  • Heteroarylalkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to a heteroaryl group as defined above. Heteroarylalkyl groups can be substituted on the heteroaryl moiety, the alkyl moiety, or both.
  • ring system as the term is used herein is meant a moiety comprising one, two, three or more rings, which can be substituted with non-ring groups or with other ring systems, or both, which can be fully saturated, partially unsaturated, fully unsaturated, or aromatic, and when the ring system includes more than a single ring, the rings can be fused, bridging, or spirocyclic.
  • spirocyclic is meant the class of structures wherein two rings are fused at a single tetrahedral carbon atom, as is well known in the art.
  • a "monocyclic, bicyclic or polycyclic, aromatic or partially aromatic ring” as the term is used herein refers to a ring system including an unsaturated ring possessing 4n+2 pi electrons, or a partially reduced (hydrogenated) form thereof.
  • the aromatic or partially aromatic ring can include additional fused, bridged, or spiro rings that are not themselves aromatic or partially aromatic.
  • naphthalene and tetrahydronaphthalene are both a "monocyclic, bicyclic or polycyclic, aromatic or partially aromatic ring" within the meaning herein.
  • a benzo-[2.2.2]-bicyclooctane is also a "monocyclic, bicyclic or polycyclic, aromatic or partially aromatic ring" within the meaning herein, containing a phenyl ring fused to a bridged bicyclic system.
  • a fully saturated ring has no double bonds therein, and is carbocyclic or heterocyclic depending on the presence of heteroatoms within the meaning herein.
  • alkoxy refers to an oxygen atom connected to an alkyl group, including a cycloalkyl group, as are defined above.
  • linear alkoxy groups include but are not limited to methoxy, ethoxy, n-propoxy, n-butoxy, n-pentyloxy, n-hexyloxy, and the like.
  • branched alkoxy include but are not limited to isopropoxy, sec-butoxy, tert-butoxy, isopentyloxy, isohexyloxy, and the like.
  • cyclic alkoxy include but are not limited to cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
  • aryloxy and arylalkoxy refer to, respectively, an aryl group bonded to an oxygen atom and an aralkyl group bonded to the oxygen atom at the alkyl moiety. Examples include but are not limited to phenoxy, naphthyloxy, and benzyloxy.
  • acyl group refers to a group containing a carbonyl moiety wherein the group is bonded via the carbonyl carbon atom.
  • the carbonyl carbon atom is also bonded to another carbon atom, which can be part of an alkyl, aryl, aralkyl cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl group or the like.
  • the group is a "formyl” group, an acyl group as the term is defined herein.
  • An acyl group can include 0 to about 12-20 additional carbon atoms bonded to the carbonyl group.
  • An acyl group can include double or triple bonds within the meaning herein.
  • An acryloyl group is an example of an acyl group.
  • An acyl group can also include heteroatoms within the meaning here.
  • a nicotinoyl group (pyridyl-3 -carbonyl) group is an example of an acyl group within the meaning herein.
  • Other examples include acetyl, benzoyl, phenylacetyl, pyridylacetyl, cinnamoyl, and acryloyl groups and the like.
  • the group containing the carbon atom that is bonded to the carbonyl carbon atom contains a halogen, the group is termed a "haloacyl" group.
  • An example is a trifluoroacetyl group.
  • amine includes primary, secondary, and tertiary amines having, e.g., the formula N(group) 3 wherein each group can independently be H or non-H, such as alkyl, aryl, and the like.
  • Amines include but are not limited to RNH 2 , for example, alkylamines, arylamines, alkylarylamines; R 2 NH wherein each R is independently selected, such as dialkylamines, diarylamines, aralkylamines, heterocyclylamines and the like; and R 3 N wherein each R is independently selected, such as trialkylamines, dialkylarylamines, alkyldiarylamines, triarylamines, and the like.
  • the term "amine” also includes ammonium ions as used herein.
  • amino group is a substituent of the form -NH 2 , -NHR, -NR 2 , -NR 3 + , wherein each R is independently selected, and protonated forms of each. Accordingly, any compound substituted with an amino group can be viewed as an amine.
  • ammonium ion includes the unsubstituted ammonium ion N3 ⁇ 4 + , but unless otherwise specified, it also includes any protonated or quaternarized forms of amines.
  • trimethylammonium hydrochloride and tetram ethyl ammonium chloride are both ammonium ions, and amines, within the meaning herein.
  • amide includes C- and N-amide groups, i.e., -C(0)N R'R", and -NR'C(0)R” groups, respectively.
  • the R' and R" of the C-amide may join together to form a heterocyclic ring with the nitrogen atom.
  • Amide groups therefore include but are not limited to carbamoyl groups (-C(0)NH 2 ) and formamide groups (-NHC(O)H).
  • a "carboxamido” group is a group of the formula C(0)NR 2 , wherein R can be H, alkyl, aryl, etc.
  • urethane (or “carbamyl”) includes N- and O-urethane groups, i.e., -NRC(0)OR and -OC(0)NR 2 groups, respectively.
  • sulfonamide (or “sulfonamido”) includes S- and N-sulfonamide groups, i.e., -S0 2 NR 2 and -NRS0 2 R groups, respectively. Sulfonamide groups therefore include but are not limited to sulfamoyl groups (-S0 2 NH 2 ).
  • amidine or “amidino” includes groups of the formula -C(NR)NR 2 . Typically, an amidino group is -C(NH)NH 2 .
  • guanidine or "guanidino” includes groups of the formula -NRC(NR)NR 2 .
  • a guanidino group is -NHC(NH)NH 2 .
  • Halo include fluorine, chlorine, bromine and iodine.
  • a "salt" as is well known in the art includes an organic compound such as a carboxylic acid, a sulfonic acid, or an amine, in ionic form, in combination with a counterion.
  • acids in their anionic form can form salts with cations such as metal cations, for example sodium, potassium, and the like; with ammonium salts such as NH 4 + or the cations of various amines, including tetraalkyl ammonium salts such as tetramethylammonium and alkyl ammonium salts such as tromethamine salts, or other cations such as trimethylsulfonium, and the like.
  • a “pharmaceutically acceptable” or “pharmacologically acceptable” salt is a salt formed from an ion that has been approved for human consumption and is generally non-toxic, such as a chloride salt or a sodium salt.
  • a “zwitterion” is an internal salt such as can be formed in a molecule that has at least two ionizable groups, one forming an anion and the other a cation, which serve to balance each other. For example, amino acids such as glycine can exist in a zwitterionic form.
  • a “zwitterion” is a salt within the meaning herein.
  • the compounds of the present invention may take the form of salts.
  • the term “salts" embraces addition salts of free acids or free bases which are compounds of the invention.
  • Salts can be "pharmaceutically-acceptable salts.”
  • pharmaceutically-acceptable salt refers to salts which possess toxicity profiles within a range that affords utility in pharmaceutical applications. Pharmaceutically unacceptable salts may nonetheless possess properties such as high crystallinity, which have utility in the practice of the present invention, such as for example utility in process of synthesis, purification or formulation of compounds of the invention.
  • Suitable pharmaceutically-acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include hydrochloric, hydrobromic, hydnodic, nitric, carbonic, sulfuric, and phosphoric acids.
  • organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, trifluoromethanesulfonic, 2-hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, ⁇ -hydroxybutyric, sal
  • Suitable pharmaceutically acceptable base addition salts of compounds of the invention include, for example, metallic salts including alkali metal, alkaline earth metal and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts.
  • Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, A -dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine.
  • Examples of pharmaceutically unacceptable base addition salts include lithium salts and cyanate salts.
  • salts may be useful, for example as intermediates in the synthesis of compounds, for example in their purification by recrystallization. All of these salts may be prepared by conventional means from the corresponding compound by reacting, for example, the appropriate acid or base with the compound.
  • pharmaceutically acceptable salts refers to nontoxic inorganic or organic acid and/or base addition salts, see, for example, Lit et al., Salt Selection for Basic Drugs (1986), IntJ. Pharm., 33, 201-217, incorporated by reference herein
  • Nonlimiting examples of potential salts of this invention include but are not limited to hydrochloride, citrate, glycolate, fumarate, malate, tartrate, mesylate, esylate, cinnamate, isethionate, sulfate, phosphate, diphosphate, nitrate, hydrobromide, hydroiodide, succinate, formate, acetate, dichloroacetate, lactate, 7-toluenesulfonate, pamitate, pidolate, pamoate, salicylate, 4-aminosalicylate, benzoate, 4-acetamido benzoate, glutamate, aspartate, glycolate, adipate, alginate, ascorbate, besylate, camphorate, camphorsulfonate, camsylate, caprate, caproate, cyclamate, laurylsulfate, edisylate, gentisate, galactarate, gluceptate,
  • a "hydrate” is a compound that exists in a composition with water molecules.
  • the composition can include water in stoichiometic quantities, such as a monohydrate or a dihydrate, or can include water in random amounts.
  • a "hydrate” refers to a solid form, i.e., a compound in water solution, while it may be hydrated, is not a hydrate as the term is used herein.
  • a "homolog" of a compound of the invention is a compound having one or more atoms of the compound replaced by an isotope of such atom.
  • homologs include compounds with deuterium in place of some hydrogen atoms of the compound such as compounds of the invention in which the methyl groups of the isopropoxy moiety of Formulas I-R and I-S are fully or partially deuterated (e.g., (D 3 C) 2 C-0-).
  • Isotopic substitutions which may be made in the formation of homologs of the invention include nonradioactive (stable) atoms such as deuterium and carbon 13, as well as radioactive (unstable) atoms such as tritium, carbon 14, iodine 123, iodine 125, etc.
  • a “solvate” is a similar composition except that a solvent other that water replaces the water.
  • a solvent other that water replaces the water.
  • methanol or ethanol can form an “alcoholate”, which can again be stoichiometic or non-stoichiometric.
  • a “solvate” refers to a solid form, i.e., a compound in solution in a solvent, while it may be solvated, is not a solvate as the term is used herein.
  • a "prodrug” as is well known in the art is a substance that can be administered to a patient where the substance is converted in vivo by the action of biochemicals within the patients body, such as enzymes, to the active pharmaceutical ingredient.
  • prodrugs include esters of carboxylic acid groups, which can be hydrolyzed by endogenous esterases as are found in the bloodstream of humans and other mammals.
  • prodrugs within the scope of this invention include:
  • the hydroxyl group may be modified to form an ester, carbonate, or carbamate. Examples include acetate, pivalate, methyl and ethyl carbonates, and dimethylcarbamate.
  • the ester may also be derived from amino acids such as glycine, serine, or lysine.
  • the compound may be modified to form an amide. Examples include acetamide or derivatization with amino acids such as glycine, serine, or lysine.
  • Certain compounds of the invention and their salts may exist in more than one crystal form and the present invention includes each crystal form and mixtures thereof.
  • the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water to form hydrates or adducts with alcohols such as Ci-4-alkanols, and the like.
  • compounds of this invention can be isolated in association with solvent molecules by crystallization from evaporation of an appropriate solvent.
  • Such solvents include but are not limited to toluene, tetrahydrofuran, dioxane, dimethylformamide, acetonitrile, acetates such as methyl acetate, ethyl acetate, butyl acetate, isobutyl acetate, propyl- and isopropyl acetate, ethers such as diethyl ether and ethyl ether, alcohols such as methanol, ethanol, 1- or 2-butanol, 1- or 2-propanol, pentanol, and dimethylsulfoxide.
  • a depiction for the compound by structure or name is considered to embrace the compound in any form (e.g., by itself, as a hydrate, solvate, or otherwise in a mixture).
  • SlPi compounds, their pharmaceutically acceptable salts or hydro lyzable esters of the present invention may be combined with a pharmaceutically acceptable carrier to provide pharmaceutical compositions useful for treating the biological conditions or disorders noted herein in mammalian species, and more preferably, in humans.
  • a pharmaceutically acceptable carrier employed in these pharmaceutical compositions may vary depending upon the type of administration desired (e.g. intravenous, oral, topical, suppository, or parenteral).
  • compositions in oral liquid dosage forms e.g. suspensions, elixirs and solutions
  • typical pharmaceutical media such as water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like
  • carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like can be employed.
  • compositions of the compounds of the invention alone or in combination with another SI Pi inhibitor or another type of therapeutic agent, or both.
  • compounds of the invention include stereoisomers, tautomers, solvates, hydrates, salts including pharmaceutically acceptable salts, and mixtures thereof.
  • Compositions containing a compound of the invention can be prepared by conventional techniques, e.g. as described in Remington: The Science and Practice of Pharmacy, 19th Ed., 1995, incorporated by reference herein.
  • the compositions can appear in conventional forms, for example capsules, tablets, aerosols, solutions, suspensions or topical applications.
  • compositions include a compound of the invention and a pharmaceutically acceptable excipient which can be a carrier or a diluent.
  • the active compound will usually be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier which can be in the form of an ampoule, capsule, sachet, paper, or other container.
  • the active compound can be solid, semi-solid, or liquid material that acts as a vehicle, excipient, or medium for the active compound.
  • the active compound can be adsorbed on a granular solid carrier, for example contained in a sachet.
  • suitable carriers are water, salt solutions, alcohols, polyethylene glycols, polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatin, lactose, terra alba, sucrose, dextrin, magnesium carbonate, sugar, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid or lower alkyl ethers of cellulose, silicic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose and polyvinylpyrrolidone.
  • the carrier or diluent can include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax.
  • the formulations can be mixed with auxiliary agents which do not deleteriously react with the active compounds.
  • Such additives can include wetting agents, emulsifying and suspending agents, salt for influencing osmotic pressure, buffers and/or coloring substances preserving agents, sweetening agents or flavoring agents.
  • the compositions can also be sterilized if desired.
  • the route of administration can be any route which effectively transports the active compound of the invention which inhibits the enzymatic activity of the focal adhesion kinase to the appropriate or desired site of action, such as oral, nasal, pulmonary, buccal, subdermal, intradermal, transdermal or parenteral, e.g., rectal, depot, subcutaneous, intravenous, intraurethral, intramuscular, intranasal, ophthalmic solution or an ointment, the oral route being preferred.
  • the carrier will typically comprise sterile water, although other ingredients that aid solubility or serve as preservatives can also be included.
  • injectable suspensions can also be prepared, in which case appropriate liquid carriers, suspending agents and the like can be employed.
  • the compounds of the present invention can be formulated using bland, moisturizing bases such as ointments or creams.
  • the preparation can be tabletted, placed in a hard gelatin capsule in powder or pellet form or it can be in the form of a troche or lozenge. If a liquid carrier is used, the preparation can be in the form of a syrup, emulsion, soft gelatin capsule or sterile injectable liquid such as an aqueous or non-aqueous liquid suspension or solution.
  • Injectable dosage forms generally include aqueous suspensions or oil suspensions which can be prepared using a suitable dispersant or wetting agent and a suspending agent Injectable forms can be in solution phase or in the form of a suspension, which is prepared with a solvent or diluent.
  • Acceptable solvents or vehicles include sterilized water, Ringer's solution, or an isotonic aqueous saline solution.
  • sterile oils can be employed as solvents or suspending agents.
  • the oil or fatty acid is non-volatile, including natural or synthetic oils, fatty acids, mono-, di- or tri-glycerides.
  • the formulation can also be a powder suitable for reconstitution with an appropriate solution as described above. Examples of these include, but are not limited to, freeze dried, rotary dried or spray dried powders, amorphous powders, granules, precipitates, or particulates.
  • the formulations can optionally contain stabilizers, pH modifiers, surfactants, bioavailability modifiers and combinations of these.
  • the compounds can be formulated for parenteral administration by injection such as by bolus injection or continuous infusion.
  • a unit dosage form for injection can be in ampoules or in multi-dose containers.
  • the formulations of the invention can be designed to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art.
  • the formulations can also be formulated for controlled release or for slow release.
  • compositions contemplated by the present invention can include, for example, micelles or liposomes, or some other encapsulated form, or can be administered in an extended release form to provide a prolonged storage and/or delivery effect. Therefore, the formulations can be compressed into pellets or cylinders and implanted intramuscularly or subcutaneously as depot injections. Such implants can employ known inert materials such as silicones and biodegradable polymers, e.g., polylactide-polyglycolide. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • the preparation can contain a compound of the invention which inhibits the enzymatic activity of the focal adhesion kinase, dissolved or suspended in a liquid carrier, preferably an aqueous carrier, for aerosol application.
  • a liquid carrier preferably an aqueous carrier
  • the carrier can contain additives such as solubilizing agents, e.g., propylene glycol, surfactants, absorption enhancers such as lecithin (phosphatidylcholine) or cyclodextrin, or preservatives such as parabens.
  • injectable solutions or suspensions preferably aqueous solutions with the active compound dissolved in polyhydroxylated castor oil.
  • Dosage forms can be administered daily, or more than once a day, such as twice or thrice daily. Alternatively dosage forms can be administered less frequently than daily, such as every other day, or weekly, if found to be advisable by a prescribing physician.
  • An embodiment of the invention also encompasses prodrugs of a compound of the invention which on administration undergo chemical conversion by metabolic or other physiological processes before becoming active pharmacological substances. Conversion by metabolic or other physiological processes includes without limitation enzymatic (e.g, specific enzymatically catalyzed) and non-enzymatic (e.g., general or specific acid or base induced) chemical transformation of the prodrug into the active pharmacological substance.
  • prodrugs will be functional derivatives of a compound of the invention which are readily convertible in vivo into a compound of the invention. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs, ed. H. Bundgaard, Elsevier, 1985.
  • methods of making a composition of a compound described herein including formulating a compound of the invention with a pharmaceutically acceptable carrier or diluent.
  • the pharmaceutically acceptable carrier or diluent is suitable for oral administration.
  • the methods can further include the step of formulating the composition into a tablet or capsule.
  • the pharmaceutically acceptable carrier or diluent is suitable for parenteral administration.
  • the methods further include the step of lyophilizing the composition to form a lyophilized preparation.
  • the compounds of the invention can be used therapeutically in combination with i) one or more other SI Pi inhibitors and/or ii) one or more other types of protein kinase inhibitors and/or one or more other types of therapeutic agents which can be administered orally in the same dosage form, in a separate oral dosage form (e.g., sequentially or non- sequentially) or by injection together or separately (e.g., sequentially or non-sequentially).
  • the invention provides combinations, comprising:
  • Combinations of the invention include mixtures of compounds from (a) and (b) in a single formulation and compounds from (a) and (b) as separate formulations. Some combinations of the invention can be packaged as separate formulations in a kit. In some embodiments, two or more compounds from (b) are formulated together while a compound of the invention is formulated separately.
  • the present invention encompasses orally bioavailable compounds that specifically agonize SIPj without binding (S1P 2 , S1P 3 and S1P 4 ), or having significant specificity over (S1P 5 ), other EDG receptors.
  • a selective SI Pi agonist can be used to treat diseases with an autoimmune, hyperactive immune-response, angiogenesis or inflammatory components, but would not be limited to such conditions.
  • Selective SlPi agonists have advantages over current therapies by increasing the therapeutic window because of reduced toxicity due to engagement of other EDG receptors.
  • the present invention encompasses compounds that bind with high affinity and specificity to the SlPi receptor in an agonist manner. Upon ligation of the SlPi receptor with agonist, signaling proceeds through G a ,, inhibiting the generation of cAMP by adenylate cyclase.
  • the present invention provides a method for activating or agonizing (i.e., to have an agonic effect, to act as an agonist) a sphingosine-1 -phosphate receptor subtype, such as SI Pi, with a compound of the invention.
  • the method involves contacting the receptor with a suitable concentration of an inventive compound to bring about activation of the receptor.
  • the contacting can take place in vitro, for example in carrying out an assay to determine the SIP receptor activation activity of an inventive compound undergoing experimentation related to a submission for regulatory approval.
  • the method for activating an SIP receptor can also be carried out in vivo, that is, within the living body of a mammal, such as a human patient or a test animal.
  • the inventive compound can be supplied to the living organism via one of the routes as described above, e.g., orally, or can be provided locally within the body tissues, for example by injection of a tumor within the organism. In the presence of the inventive compound, activation of the receptor takes place, and the effect thereof can be studied.
  • An embodiment of the present invention provides a method of treatment of a malcondition in a patient for which activation of an SIP receptor, such as SI Pi, is medically indicated, wherein the patient is administered the inventive compound in a dosage, at a frequency, and for a duration to produce a beneficial effect on the patient.
  • the inventive compound can be administered by any suitable means, examples of which are described above.
  • Reagents (i) KH 2 P0 4 , H 2 0 2 , NaC10 2 , CH 3 CN; (ii) H 2 NNHCSNH 2 , POCl 3 CuBr 2 , isoamylnitrite, CH 3 CN.
  • Reagents (i) (5)-2-methyl-CBS-oxazaborolidine, BH 3 -Me 2 S, toluene, DCM; (ii) PG- Cl, (where PG is protecting group), e.g. TBSCl, imidazole, DMF; (iii) bis(pinacolato)diboron, PdCl 2 (dppf).CH 2 Cl 2 , KOAc, 1,4-dioxane. [0302] The (5 -enantiomer was prepared in same manner as outlined in Scheme 5 by the use of (i?)-2-methyl-CBS-oxazaborolidine in Step i.
  • the racemic material can be prepared in an analogous manner using NaBH 4 as reducing agent in Step i.
  • Reagents (i) K 2 C0 3 , Pd(PPh 3 ) 4 , DME, H 2 0; (ii) NaOi ' Pr, z ' PrOH; (iii) deprotection, e.g. TBAF, THF or HCl, 1,4-dioxane.
  • Reagents (i) K 2 C0 3 , Pd(PPh 3 ) 4 , DME, H 2 0; (ii) NaO/Pr, PrOH; (iii) 4N HC1, 1,4- dioxane; (iv) (a) R'-LG or R"-LG, where LG represents a leaving group, 2 C0 3 , CH 3 CN; (b) R 3 -C0 2 H or R 4 -C0 2 H , HOBt, EDC, DMF or R 3 -COCl or R 4 -C0 2 H , TEA, DCM; (c) R 3 - S0 2 C1 or R 5 -S0 2 C1, TEA, DCM (d) R 4 -CHO, HO Ac, NaB3 ⁇ 4 or NaCNBH 3 or Na(OAc) 3 BH, MeOH; (e) R 3 -OCOCl or R -OCOC1, DIEA, DMF; (f) HN(R 7
  • Reagents (i) (3-cyano-4-isopropoxyphenyl)boronic acid, K 2 C0 3 , Pd(PPh 3 ) 4 , DME, H 2 0; (ii) (R)-, (5)-, or racemic tert-butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan- 2-yl)-2,3-dihydro-lH-inden-l-yl)oxy)silane, K 2 C0 3 , Pd(PPh 3 ) 4 , DME, H 2 0; (iii) TBAF, THF.
  • Reagents (i) (ii) (i?)-, (S)-, or racemic tert-butyldimethyl((4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-2,3-dihydro-lH-inden-l-yl)oxy)silane, 2 C0 3 , Pd(PPh 3 ) 4 , DME, H 2 0; (ii) (3-cyano-4-isopropoxyphenyl)boronic acid, K 2 C0 3 , Pd(PPh 3 ) 4 , DME, H 2 0; (iii) TBAF, THF.
  • Reagents (i) (S,S) Noyori Catalyst (Chloro ⁇ [15,25))-(-)-2-amino-l,2- diphenylethyl](4-toluene sulfonyl)amidi ⁇ (p-cymene) ruthenium (II)), 5:1 HC0 2 H/NEt 3 , 30°C; (ii) DBU, DPPA, Toluene; (iii) Pt0 2 , H 2 , Boc 2 0, MeOH; (iv) pinacolborane, PdCl 2 (dppf)-CH 2 Cl 2 , KOAc, Dioxane, 80°C.
  • Reagents (i) 5-(5-bromo-l,3,4-thiadiazol-2-yl)-2-fluorobenzonitrile, Pd(PPh 3 ) 4 , K 2 C0 3 , 3:1 DME/H 2 0, 90°C; (ii) NaO'Pr, IP A, 60°C; (iii) 4M HCl in dioxane; (iv) (a) R'-LG or R"-LG, where LG represents a leaving group, 2 C0 3 , CH 3 CN or (b) R'-C0 2 H or R 2 - C0 2 H, HOBt, EDC, DMF or R'-COCl or R 2 -COCl, TEA, DCM or (c) R -S0 2 C1 or R 3 - S0 2 C1, TEA, DCM or (d) R 2 -CHO, HO Ac, NaBH 4 or NaCNBH 3 or Na(OAc) 3 BH, MeOH or (e)
  • Reagents (i) 5-(5-bromothiazol-2-yl)-2-fluorobenzonitrile, Pd(PPh 3 ) 4 , K 2 C0 3 , 3:1 DME/H 2 0, 90°C; (ii) NaOiPr, IP A, 60°C; (iii) 4M HCl in dioxane; (iv) sulfamide, DIEA, dioxane, 100°C.
  • Reagents (i) bis(pinacolato)diboron, PdCl 2 (dppf).CH 2 Cl 2 , OAc, 1,4-dioxane.
  • Reagents (i) 3 P0 4 :3H 2 0, Pd(PPh 3 ) 4 , DME, H 2 0; (ii) K 3 P0 4 :3H 2 0, 2-isopropoxy-5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzonitrile, Pd(PPh 3 ) 4 , DMF, H 2 0 (iii) 4N HC1, 1,4-dioxane; (iv) (a) R ⁇ LG, where LG represents a leaving group, 2 C0 3 , BU 4 NI, CH 3 CN or (b) R 1 -C0 2 H, HOBt, EDC, DMF or R 2 ⁇ COCl, TEA, DCM or (c) R -S0 2 C1, TEA, DCM or (d) R'-CHO, HOAC, NaBH 4 or NaCNBH 3 or Na(OAc) 3 BH, MeOH or (e
  • the (R)-enantiomers can be prepared in same manner as outlined in Scheme 16 by the use of (i?)-tert-butyl (4-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)- 2,3-dihydro- lH-inden-l-yl)carbamate in Step i.
  • the gradient was 20-100% with mobile phase B over 2.5 min then held at 100% for 2.5 mins.
  • the flow rate was 1 mL/min.
  • Method 1 40-95% over 0.5 min, hold at 95% for 8.5 min, then return to 40% over 2 min, with a flow rate of 1 mL/min.
  • Final compounds were checked for purity using Method 2: 5% for 1 min, 5-95% over 9 min, then hold at 95% for 5 min, with a flow rate of 1 mL/min.
  • Method 3 20-100% over 2.5 min then held at 100% for 4.5 min, with the flow rate of 1 mL/min.
  • Enantiomeric excess was determined by integration of peaks that were separated on a Chiralpak AD-H, 250 x 4.6 mm column at a flow rate of 1 mL/min and an isocratic mobile phase. Unless otherwise indicated, the chiral data provided uses this method. Alternatively, chiral separations were performed under the following conditions, denoted as Chiral Method 1: Chiralpak AY-H, 250 x 4.6 mm column at a flow rate of 1 mL/min and an isocratic mobile phase. Chiral Method 2: Chiralcel OZ-3, 150 x 4.6 mm at flow rate of 1 mL min and an isocratic mobile phase.
  • EA ethyl acetate
  • TEA triethylamine
  • DEA diethyl amine
  • HOBt hydroxybenzotriazole
  • IP A isopropanol
  • DMF dimethylformamide
  • DMA dimethyl acetamide
  • Racemic ( ⁇ )-te ⁇ butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3- dihydro-1 H-inden- l-yl)oxy)silane IND INT-8 was prepared in an analogous fashion from IND INT-5.
  • a 20 mL microwave vial was charged sequentially with heterocyclic bromide (1 eq), (R)- (S)- or racemic indanol dioxaborolane (IND INT-6, 7 or 8, 1 eq), DME:H 2 0 (3:1, 0.05 M) and potassium carbonate (3 eq).
  • the mixture was degassed by bubbling N 2 gas through the stirring solution for 10 min.
  • Pd(PPh 3 )4 (0.07 eq) was added and the mixture degassed for additional 2 min.
  • the vial as was capped and subjected to microwave irradiation at 100°C until reaction completed (40-60 min). Additional bromide was added if needed.
  • the vial was cooled to room temperature, diluted with EA (10 x volume), washed with water and brine, dried over MgS0 4 , and concentrated.
  • the crude product was purified by silica gel column chromatography (EA / hexanes).
  • reaction mixture was degassed by bubbling N 2 gas through the stirring solution for 10 min.
  • Pd(PPh 3 ) 4 was added and mixture degassed for additional 2 min.
  • the vial was subjected to microwave irradiation at 100°C for 40 min.
  • the reaction mixture was cooled to room temperature, diluted with EA (10 mL), and washed with water and brine.
  • (R)-5-(5-( 1 -((tert-butyldimethylsilyl)oxy)-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3 ,4- tliiadiazol-2-yl)-2-isopropoxybenzonitrile was prepared in an analogous fashion using (R)-5- (5-( 1 -((tert-butyldimethylsilyl)oxy)-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3,4-thiadiazol-2-yl)-2- fluorobenzonitrile.
  • a reaction pressure flask was charged sequentially with the iheterocyclic bromide (1 eq), (R)- or (5)-Boc-protected indane amine (1 eq), DME:H 2 0 (3:1, 0.07 M) and potassium carbonate (3 eq).
  • the mixture was degassed by bubbling N 2 gas through the stirring solution for 20 min.
  • Pd(PPh 3 ) 4 (0.07 eq) was added and the mixture was degassed for additional 5 min.
  • the reaction flask was capped tightly and the mixture was heated at 85°C for 12-24 h.
  • the reaction was cooled to room temperature, diluted with water (2x volume), and stirred for 30 min.
  • the resulting solid was filtered, washed with hexanes, and dried under high vacuum.
  • the crude product was purified by silica gel column chromatography (EA / hexanes) or used in the next experiment without purification.
  • (i?)-2-isopropoxy-5-(5-(l-((2-(memylsulfonyl)emyl)amino)-2,3-dihydro-lH- inden-4-yl)-l,3,4-thiadiazol-2-yl)benzonitrile 32 was prepared in an analogous fashion using (i?)-5-(5-(l -amino-2,3-dihydro- 1 H-inden-4-yl)-l ,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 5.
  • reaction mixture was heated at 90°C for 24 h.
  • the reaction was diluted with DCM (5 mL) and washed with saturated aqueous ammonium chloride (2 x 5 mL) and saturated aqueous sodium bicarbonate (2 x 5 mL) and then dried.
  • the crude reaction was purified by a silica gel column (MeOH/DCM) to yield 44 mg (61%) of (5)-2-isopropoxy-5-(5-(l-((2- (methylsulfonyl)emyl)amino)-2,3-dihydro-lH-inden-4-yl)tWazol-2-yl)benzonitrile 221 as brown liquid.
  • N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl)methanesulfonamide 34 was prepared in an analogous fashion using (R)-5- (5-(l- amino-2,3 -dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 5.
  • N-((/-)-4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH -inden-1 -yl)-2- ((i?)-3-hydroxypyrrolidin-l-yl)ethanesulfonamide 38 was prepared in an analogous fashion using (7?)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)- 2,3 -dihydro- 1 H-inden- 1 -yl)ethenesulfonamide.
  • N-((5)-4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- 1 - yl)-2- ((i?)-3-hydroxypiperidin-l-yl)ethanesulfonarnide 141 was prepared in an analogous fashion using ( 1 S)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- 1 -yl)ethenesulfonamide.
  • a microwave vial was charged with 5-bromo-2-isopropoxybenzonitrile (200 mg, 0.83 mmol), thiophen-2-ylboronic acid (106.5 mg, 0.83 mmol), potassium carbonate (345.3 mg, 2.49 mmol) and 3:1 mixture of dimethylethylene glycol / H 2 0 (2 mL).
  • the reaction mixture was degassed by bubbling N 2 gas through the stirred solution for 10 min.
  • Pd(PPh 3 )4 (20.4 mg, 0.02 mmol) was added and the solution degassed for additional 2 min.
  • the vial was subjected to microwave irradiation at 100°C for 30 min.
  • EA ethyl acetate
  • TEA triethylamine
  • DEA diethyl amine
  • HOBt jV-hydroxybenzotriazole
  • EDC l-ethyl-3-(3- dimethylaminopropyl) carbodiimide hydrochloride
  • isopropanol
  • JV- dimethylformamide DMF
  • DMA dimethyl acetamide
  • DMA dimethyl acetamide
  • DBU diphenylphosphoryl azide
  • DPP A Di-tert-butyl dicarbonate
  • Boc 2 0 1,2- dimethoxyethane
  • DME 1,2- dimethoxyethane
  • DIEA N,N-Diisopropylethylamine
  • (S)-tert-buty ⁇ (5-(2-(3-cyano-4-fluorophenyl)thiazol-5-yl)-l ,2,3,4- tetrahydronaphthalen-l-yl)carbamate can be prepared in an analogous fashion from (S)-tert- butyl (5-(4,4,5,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl) carbamate THN INT-8.
  • (S)-tert-butyl (5- (2- (3- cyano- 4-isopropoxyphenyl)thiazol- 5-yl) -1,2,3,4- tetrahydronaphthalen-l-yl)carbamate can be prepared in an analogous fashion from (S)-tert- butyl (5- (2- (3- cyano- 4-fluorophenyl) thiazol-5-yl)- 1,2,3 ,4-tetrahydronaphthalen-l- yl)carbamate.
  • (.S)-2-isopropoxy-5-(5-(5-((pyrid -3-ylmemyl)amino)- 5,6,7,8- tetrahydronaphthalen- l-yl)-l,3,4-thiadiazol-2-yl)benzonitrile 271 was prepared in an analogous fashion from (5)- 5- (5- (5- amino- 5,6,7,8- tetrahydronaphthalen- 1- yl)- 1,3,4- thiadiazol- 2- yl)- 2-isopropoxybenzonitrile 229.
  • (5)-5-(5-(5-((2- (dimemylamino)ethyl)amino)- 5,6,7,8-tetrahydronaphthalen-l-yl)- 1,3,4- thiadiazol-2-yl)- 2-isopropoxybenzonitrile can be prepared in a similar fashion from (5)-5-(5-(5-ammo-5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 229.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Pulmonology (AREA)
  • Neurosurgery (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Transplantation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Heterocyclic Compounds Containing Sulfur Atoms (AREA)
  • Thiazole And Isothizaole Compounds (AREA)
  • Nitrogen- Or Sulfur-Containing Heterocyclic Ring Compounds With Rings Of Six Or More Members (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Compounds that selectively modulate the sphingosine 1 phosphate receptor are provided including compounds which modulate subtype 1 of the S1P receptor. Methods of chiral synthesis of such compounds is provided. Uses, methods of treatment or prevention and methods of preparing inventive compositions including inventive compounds are provided in connection with the treatment or prevention of diseases, malconditions, and disorders for which modulation of the sphingosine 1 phosphate receptor is medically indicated.

Description

SELECTIVE HETEROCYCLIC SPHINGOSI E 1 PHOSPHATE RECEPTOR
MODULATORS
RELATED APPLICATIONS
[0100] This application claims priority to application serial no. 61/485,872, filed May 13, 2011, which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
[0101] The invention relates to compounds which are agonists of the sphingosine 1- phosphate receptor subtype 1, methods of their synthesis and methods of their therapeutic and/or prophylactic use.
BACKGROUND
[0102] The SlPi/EDGi receptor is a G-protein coupled receptor (GPCR) and is a member of the endothelial cell differentiation gene (EDG) receptor family. Endogenous ligands for EDG receptors include lysophospholipids, such as sphingosine- 1 -phosphate (SIP). Like all GPCRs, ligation of the receptor propagates second messenger signals via activation of G- proteins (alpha, beta and gamma).
[0103] Development of small molecule SIPi agonists and antagonists has provided insight into some physiological roles of the SIPj/SlP-receptor signaling system. Agonism of the SI Pi receptor perturbs lymphocyte trafficking, sequestering them in lymph nodes and other secondary lymphoid tissue. This leads to rapid and reversible lymphopenia, and is probably due to receptor ligation on both lymphatic endothelial cells and lymphocytes themselves (Rosen et al, Immunol. Rev., 195:160-177, 2003). A clinically valuable consequence of lymphocyte sequestration is exclusion of them from sights of inflammation and/or auto-immune reactivity in peripheral tissues.
[0104] Agonism of SlPj has also been reported to promote survival of oligodendrocyte progenitors (Miron et al, Ann. Neurol, 63:61-71, 2008). This activity, in conjunction with lymphocyte sequestration would be useful in treating inflammatory and autoimmune conditions of the central nervous system. SUMMARY OF THE INVENTION
[0105] The present invention is directed to heterocyclic compounds adapted to act as agonists of SIP receptor subtype 1, SlPj; methods of preparation and methods of use, such as in treatment of a malcondition mediated by SIPi activation, or when activation of SIPj is medically indicated.
[0106J Certain embodiments of the present invention comprise a compound having the structure of Formula (I) or a pharmaceutically acceptable salt, ester, prodrug, homolog, tautomer, stereoisomer, or hydrate, or solvate thereof:
Figure imgf000003_0001
(I)
[0107] A dashed line signifies that a single bond or a double bond can be present, provided that there are two double bonds and three single bonds in the ring comprising A1, A , and A . A , A , and A each independently can be CH or S or N; provided that one of A , A2, and A3 is S.
[0108] R1 can be di-substituted phenyl or di-substituted pyridinyl where the phenyl and pyridinyl substituents ca each be independently any of halo, nitro, cyano, perfluromethyl, fluorinated methyl, and Ci-4-alkoxy. When R1 is di-substituted phenyl, such phenyl is para- substituted with C -alkoxy.
[01 2 can be
Figure imgf000003_0002
wherein a wavy line indicates a point of attachment. [0110] X can be -NR'R" or -OR'";
R' can be H, C1-4 alkyl, n-hydroxy C alkyl, -S02-R3, or -CO-R3. R" can be H, -S02-R5 , C alkyl optionally substituted with 1 or more R4, or a ring moiety optionally substituted with R6 wherein such ring moiety is pyridinyl, oxazolyl, piperidinyl, cyclohexyl, morpholinyl, thiazolyl, pyrazolyl, pyrrolidinyl, imidazolyl, or phenyl. R'" can be H, C alkyl, or -CO-R3. R' and R" taken together with the nitrogen atom to which they are bound can form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle is optionally singly or multiply substituted with substituents independently selected from the group consisting of -OH, oxo, -NH2, n-hydroxy-CM alkyl, -COOH, -(CH2)m-COOH, - (CH2)m-COOR3, -N(R3R3), and -(CH2)m-CO-N(R7R7). Each R3 can be independently CM alkyl or H. Each R4 can be independently H, halo, OH, oxo, =NH, NH2, -COOH, F, -NHR3, - NiRV -SCh-R3, -S02-N(R7R7), -N(R3)-S02-R3, -COOR3, -OCO-R3, -CO-N(R7R7), - N(R3)-COR3, Ci-3 alkyl, Ci-3 alkoxy, and a ring moiety optionally substituted with R6 wherein such ring moiety is pyridinyl, oxazolyl, piperazinyl, piperidinyl, morpholinyl, pyrrolidinyl, pyrazolyl, imidazolyl, benzimidazolyl, azetidinyl, cyclobutinyl, or phenyl. Each R5 can be independently R4, CM alkyl, C3-6 cycloalkyl, or CM alkyl optionally substituted with 1 or more R4. Each R6 can be independently halo, OH, -NH2, -NHR3, -N(R3R3), -COOH, - COOR3, -NHCO-R3 . Each R7 can be independently C1-4 alkyl or H, or two R7 taken together with the nitrogen atom to which they are bound can form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle can be optionally substituted with -OH, -NH2, -N(R3R3), n-hydroxy C1-4 alkyl, -(CH2)m-COOH, -(CH2)m-COOR3. Each m can be independently 0, 1, 2, or 3.
[0111] In certain embodiments, a pharmaceutical composition comprising a compound of the invention and a suitable excipient is provided.
[0112] In certain embodiments a method of use of an inventive compound comprising preparation of a medicament is provided.
[0113] In certain combinations a pharmaceutical combination comprising a compound of the invention and a second medicament is provided. In various embodiments the second medicament is medically indicated for the treatment of multiple sclerosis, transplant rejection, acute respiratory distress syndrome or adult respiratory distress syndrome.
[0114] In certain embodiments, a method of activation or agonism of a sphingosine-1- phosphate receptor subtype 1 comprising contacting the receptor subtype 1 with a compound of claim 1 is provided. In various embodiments, the compound of claim 1 activates or agonizes the sphingosine-1 -phosphate receptor subtype 1 to a greater degree than the compound activates or agonizes a sphingosin-1 -phosphate receptor subtype 3.
[0115] In certain embodiments a method of treatment of a malcondition in a patient for which activation or agonism of an SlPj receptor is medically indicated, is provided. In various embodiment, selective activation or agonism of an SI Pj receptor, such as with respect to an S1P3 receptor, is medically indicated. In various embodiments, the malcondition comprises multiple sclerosis, transplant rejection, or acute respiratory distress syndrome.
[0116] In certain embodiments, a method is provided for chiral synthesis of certain compounds including compounds of the invention. In certain other embodiments the invention provides certain intermediate compounds associated with such methods of chiral synthesis.
DETAILED DESCRIPTION OF THE INVENTION
[0117] Certain embodiments of the present invention comprise a compound having the structure of Formula (I) or a pharmaceutically acceptable salt, ester, prodrug, homolog, tautomer, stereoisomer, or hydrate, or solvate thereof:
Figure imgf000005_0001
(I)
[0118] A dashed line signifies that a single bond or a double bond can be present, provided that there are two double bonds and three single bonds in the ring comprising A1, A , and A . A , A , and A each independently can be CH or S or N; provided that one of A , A2, and A3 is S.
[0119] Rl can be di-substituted phenyl or di-substituted pyridinyl where the phenyl and pyridinyl substituents ca each be independently any of halo, nitro, cyano, perfluromethyl, fluorinated methyl, and C1-4-alkoxy. When R1 is di-substituted phenyl, such phenyl is para- substituted with Ci-4-alkoxy.
[0120] R2 can be
Figure imgf000006_0001
wherein a wavy line indicates a point of attachment.
[0121] X can be -NR'R" or -OR'";
R' can be H, CM alkyl, n-hydroxy CM alkyl, -S02-R3, or -CO-R3. R" can be H, -S02-R5 , C alkyl optionally substituted with 1 or more R4, or a ring moiety optionally substituted with R6 wherein such ring moiety is pyridinyl, oxazolyl, piperidinyl, cyclohexyl, mo holinyl, thiazolyl, pyrazolyl, pyrrolidinyl, imidazolyl, or phenyl. R'" can be H, C alkyl, or -CO-R . R' and R" taken together with the nitrogen atom to which they are bound can form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle is optionally singly or multiply substituted with substituents independently selected from the group consisting of -OH, oxo, -NH2, n-hydroxy-C^ alkyl, -COOH, -(CH2)M-COOH, - (CH2)M-COOR3, -N(R3R3), and -(CH2)M-CO-N(R7R7). Each R3 can be independently CM alkyl or H. Each R4 can be independently H, halo, OH, oxo, =NH, NH2, -COOH, F, -NHR3, - N(R7R7),-S02-R3, -S02-N(R7R7), -N(R3)-S02-R3, -COOR3, -OCO-R3, -CO-N(R7R7), - N(R3)-COR3, C1-3 alkyl, C1-3 alkoxy, and a ring moiety optionally substituted with R6 wherein such ring moiety is pyridinyl, oxazolyl, piperazinyl, piperidinyl, morpholinyl, pyrrolidinyl, pyrazolyl, imidazolyl, benzimidazolyl, azetidinyl, cyclobutinyl, or phenyl. Each R5 can be independently R4, C alkyl, C3-6 cycloalkyl, or C alkyl optionally substituted with 1 or more R4. Each R6 can be independently halo, OH, -NH2, -NHR3, -N(R3R3), -COOH, - COOR3, -NHCO-R3. Each R7 can be independently CM alkyl or H, or two R7 taken together with the nitrogen atom to which they are bound can form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle can be optionally substituted with -OH, -NH2, -N(R3R3), n-hydroxy CM alkyl, -(CH2)M-COOH, -(CH2)M-COOR3. Each m can be independently 0, 1, 2, or 3.
[0122] In certain embodiments, the compounds of the invention have the structure of a specific of Formula I or a pharmaceutically acceptable salt, ester, prodrug, homolog, hydrate or solvate thereof. In certain embodiments the invention provides compounds which are substantially enantiomerically pure. In certain such embodiments, the compounds are enantiomerically pure with respect to a chiral carbon on an indanyl or tetrahydronaphthalenyl moiety.
[0123] In certain embodiments the invention provides compounds which have an EC50 as an agonist of the wild type SIP receptor subtype 1 which is at least ten times smaller than the EC50 of such compound as an agonist of a mutant SIP receptor subtype 1 having a single mutation with respect to wild type SIP receptor subtype 1 such that the 101st amino acid residue is changed from asparagine to alanine.
[0124] In certain embodiments the invention provides compounds which have an EC50 as an agonist of the wild type SIP receptor subtype 1 which is at least twenty times smaller than the EC50 of such compound as an agonist of a mutant SIP receptor subtype 1 having a single mutation with respect to wild type SIP receptor subtype 1 such that the 101st amino acid residue is changed from asparagine to alanine.
[0125] In certain embodiments the invention provides compounds which have a therapeutic index of at least 5 as measured in rats following 5 or 14 days of dosing of rats with the compound where the therapeutic index is calculated as a ratio of (i) the highest dose of such compound which achieves less than or equal to a ten percent increase in the ratio of lung to terminal body weight at the conclusion of such 5 or 14 days of dosing, to (ii) the dose of such compound achieving 50% lymphopenia in rats. In certain embodiments, such therapeutic index is at least 10 and in certain embodiments the therapeutic index is at least 20. In certain embodiments, the therapeutic index for a compound is at least five times greater than the therapeutic index for the enantiomer of such compound.
[0126] In certain embodiments the invention provides compounds which have a therapeutic index of at least 5 as measured in rats following 5 or 14 days of dosing of rats with the compound where the therapeutic index is calculated as a ratio of (i) the highest dose of such compound which achieves less than or equal to a ten percent increase in the ratio of lung to terminal body weight at the conclusion of such 5 or 14 days of dosing, to (ii) the dose of such compound achieving 50% lymphopenia in rats. In certain embodiments, such therapeutic index is at least 10 and in certain embodiments the therapeutic index is at least 20. In certain embodiments, the therapeutic index for a compound is greater than the therapeutic index for the enantiomer of such compound. In certain embodiments, the therapeutic index for a compound is at least 150% of the therapeutic index for the enantiomer of such compound. [0127] In certain embodiments the invention provides compounds where the structure of Formula I is selected from the roup consisting of formulas a-i through a-x:
Figure imgf000008_0001
a-i a-ii a-iii a-iv
Figure imgf000008_0002
a-v a-vi a-vii a-viii
Figure imgf000008_0003
a-ix a-x
[0128] In certain embodiments the invention provides compounds where A1 is S, in other embodiments the invention provides compounds where A is S and in other embodiments the
1 9 invention provides compounds where A is S. In certain embodiments A is N and A is CH
9 9
or N; in certain such embodiments A is CH and in others A is N.
[0129] In certain es compounds where R1 is
Figure imgf000008_0004
R is C2-4 alkyl; and Y is -CN, -CI, -O-R , or -CF3. In certain such embodiments R isopropyl or ethyl. In certain embodiments Y is -CN or -0-C2H5.
[0130] In certain embodiments the invention provides compounds where R is
Figure imgf000008_0005
In certain of such embodiments the invention provides compounds where R2 is
Figure imgf000009_0001
In certain of such embodiments the compound is substantially enantiomerically pure.
[0131] In certain embodiments the invention provides compounds where Y is CI, in other embodiments the invention provides compounds where Y is CF3 and in other embodiments the invention provides compounds where Y is CN.
[0132] In certain embodiments the invention provides compounds where X is -NR'R", in other embodiments the invention provides compounds where X is -OR'". In certain embodiments the invention provides compounds where X is -OR'". In certain embodiments the invention provides compounds where X is -OH and in other embodiments the invention provides compounds where X is -OCO-R .
[0133] In certain embodiments the invention provides compounds where R3 is C1-3 alkyl; in other embodiments the invention provides compounds where R' is H.
[0134] In certain embodiments the invention provides compounds where R' is -COR3; in other embodiments the invention provides compounds where R' is S02-R3. In certain embodiments the invention provides compounds where R" is H.
[0135] In certain embodiments the invention provides compounds where R" is -S02-R5; in other embodiments the invention provides compounds where R" is C1-4 alkyl where the Ci. 4 alkyl is optionally substituted with 1 or more substituents defined by R4. In certain embodiments the invention provides compounds where R" is -(CR^^n-R4 and each Ra and each Rb can be independently any of H, hydroxyl and methyl or where Ra and Rb are bound to the same carbon they can be taken together to form oxo (i.e. with the carbon to which they are bound forming a carbonyl moiety). In certain such embodiments n can be 0, 1, 2, or 3 and in certain embodiments n is 2. In certain such embodiments R can be-OH, -NH2, - NHR3, -N(R7R7), or -COOH.
[0136] In certain embodiments the invention provides compounds where R5 is C alkyl optionally substituted with 1 or more R4. In certain embodiments the invention provides compounds where R4 is OH; in other embodiments the invention provides compounds where R4 is Ci_3 alkoxy. In certain embodiments the invention provides compounds where R5 is (CH2)2-OR3.
[0137] In certain embodiments the invention provides compounds where Y is CN and X is -NH-S02-R . In certain embodiments the invention provides compounds where R is - C2H5-N((R7R7) or -CH2-CO-N(R7R7). In certain embodiments the invention provides compounds where Y is CN and X is -NH-CO-N(R7R7).
[0138] In certain embodiments X is -NH2 and in certain of such embodiments Y is CN.
[0139] In certain embodiments the invention provides compounds having the following formula:
Figure imgf000010_0001
wherein
A2 is CH or N;
Rm is H, -S02-R5, or CH alkyl optionally substituted with 1 or more R4;
[0140] each R4 is independently H, OH, oxo, NH2, -COOH, S02-R3, -N(R3)-S02-R3, - COOR3, -C0-N(R7R7), N(R7R7), C1-3 alkyl, C1-3 alkoxy; or a ring moiety selected from pyridinyl, oxazolyl, pyrrolidinyl, or imidazolyl.
[0141] In certain embodiments the invention provides compounds having the following formula:
Figure imgf000011_0001
Figure imgf000011_0002
[0144] In certain embodiments the invention provides compounds where RM is -S02-R5. In certain embodiments, R5 is NH2, or C alkyl optionally substituted with 1 or more R4 In certain embodiments, R4 is OH, C1-3 alkoxy, -COOH, -COOR3 , CO-N(R7R7) or N(R7R7).
[0145] In certain embodiments the invention provides compounds where RM is is C1-4 alkyl optionally substituted with 1 or more R4. In certain embodiments, the invention provides compounds where at least one of the 1 or more R4 is oxo. In certain embodiments, the invention provides compounds where one of the 1 or more R4 is OH, or C1-3 alkoxy.
[0146] In certain embodiments the invention provides one or more of compounds 1-227:
Figure imgf000011_0003
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001

Figure imgf000020_0001

Figure imgf000021_0001
20
Figure imgf000022_0001
21
Figure imgf000023_0001
22
Figure imgf000024_0001
Figure imgf000024_0002
Figure imgf000024_0003
Figure imgf000024_0004
Figure imgf000024_0005

Figure imgf000025_0001

Figure imgf000026_0001
Figure imgf000026_0002
Figure imgf000026_0003
Figure imgf000026_0004
Figure imgf000026_0005

Figure imgf000027_0001

Figure imgf000028_0001

Figure imgf000029_0001
Figure imgf000029_0002
Figure imgf000029_0003
Figure imgf000029_0004
Figure imgf000029_0005

Figure imgf000030_0001

Figure imgf000031_0001
Figure imgf000031_0002
Figure imgf000031_0003
Figure imgf000031_0004
Figure imgf000031_0005
30
Figure imgf000032_0001
31
Figure imgf000033_0001
32
Figure imgf000034_0001
or any pharmaceutically acceptable salt, tautomer, stereoisomer, solvate, hydrate, or prodrug thereof. In certain of such embodiments, the invention provides a compound selected from compounds 43, 46, 47, 56, 58, 166, 172, 186 and 246 or any pharmaceutically acceptable salt, ester, tautomer, stereoisomer, solvate, hydrate, homolog, or prodrug thereof. In certain of such embodiments, the invention provides compound 43, 46, 166 or 246 or any pharmaceutically acceptable salt, ester, tautomer, solvate, hydrate, homolog, or prodrug thereof.
[0147] In certain embodiments the invention provides one or more of compounds 307- 314:
Figure imgf000035_0001
Figure imgf000035_0002
Figure imgf000035_0003
Figure imgf000035_0004
and or any pharmaceutically acceptable salt, tautomer, stereoisomer, solvate, hydrate, or prodrug thereof.
[0148] In certain embodiments the invention provides one or more of compounds 228- 306:
Figure imgf000036_0001
Figure imgf000037_0001

Figure imgf000038_0001
Figure imgf000038_0002

Figure imgf000039_0001

Figure imgf000040_0001

Figure imgf000041_0001
40
Figure imgf000042_0001
41
Figure imgf000043_0001
or any pharmaceutically acceptable salt, tautomer, stereoisomer, solvate, hydrate, or prodrug thereof. [0149] In certain embodiments, an invention compound of Formula I is provided wherein the compound has at least one chiral center and is substantially enantiomerically pure.
[0150] In other embodiments, a pharmaceutical composition comprising an invention compound of Formula I and a suitable excipient is provided.
[0151] In other embodiments, a pharmaceutical combination comprising an invention compound and a second medicament is provided. In still other embodiments, a pharmaceutical combination comprising an invention compound and a second medicament is provided wherein the second medicament is medically indicated for the treatment of multiple sclerosis, transplant rejection, or adult respiratory distress syndrome.
[0152] In certain embodiments, a method of use of an invention compound for preparation of a medicament is provided.
[0153] In certain embodiments a method of activation or agonism of a sphingosine-1 - phosphate receptor subtype 1 by contacting the receptor subtype 1 with an effective amount of an invention compound. In further embodiments, a method of activation or agonism of a sphingosine-1 -phosphate receptor subtype 1 by contacting the receptor subtype 1 with an effective amount of an invention compound is provided, wherein the compound activates or agonizes the sphingosine-1 -phosphate receptor subtype 1 to a greater extent than the compound activates or agonizes a sphingosine-1 -phosphate receptor subtype 3. In further embodiments, a method of activation or agonism of a sphingosine-1 -phosphate receptor subtype 1 by contacting the receptor subtype 1 with an effective amount of an invention compound is provided, wherein the sphingosine-1 -phosphate receptor subtype 1 is disposed within a living mammal.
[0154] In certain embodiments, a method is provided for treatment of a malcondition in a patient for which activation or agonism of an sphingosine-1 -phosphate receptor subtype 1 is medically indicated, by administering an effective amount of an invention compound to the patient at a frequency and for a duration of time sufficient to provide a beneficial effect to the patient. In further embodiments, a method is provided for treatment of a malcondition in a patient for which activation or agonism of an sphingosine-1 -phosphate receptor subtype 1 is medically indicated, by administering an effective amount of an invention compound to the patient at a frequency and for a duration of time sufficient to provide a beneficial effect to the patient, wherein selective activation or agonism of an SIP subtype 1 receptor with respect to other subtypes of SIP receptor is medically indicated. In yet further embodiments, a method is provided for treatment of a malcondition in a patient for which activation or agonism of an sphingosine-1 -phosphate receptor subtype 1 is medically indicated, by administering an effective amount of an invention compound to the patient at a frequency and for a duration of time sufficient to provide a beneficial effect to the patient, wherein the malcondition comprises rejection of transplanted organs or tissue; graft-versus-host diseases brought about by transplantation; autoimmune syndromes including rheumatoid arthritis; acute respiratory distress syndrome; adult respiratory distress syndrome; influenza; cancer; systemic erythematosus; Hashimoto's thyroiditis; lymphocytic thyroiditis; multiple sclerosis; myasthenia gravis; type I and II diabetes; uveitis; posterior uveitis; uveitis associated with Behcet's disease; uveomeningitis syndrome; allergic encephalomyelitis; chronic allograft vasculopathy; post-infectious autoimmune diseases including rheumatic fever and postinfectious glomerulonephritis; inflammatory and hyperproliferative skin diseases; cutaneous manifestations of immunologically-mediated disorders; psoriasis; atopic dermatitis; osteomyelitis; contact dermatitis; eczematous dermatitis; seborrhoeic dermatitis; lichen planus; pemphigus; bullous pemphigoid; epidermolysis bullosa; urticaria; angioedema; vasculitis; erythema; cutaneous eosinophilia; acne; alopecia areata; keratoconjunctivitis; vernal conjunctivitis; keratitis; herpetic keratitis; dystrophia epithelialis corneae; corneal leukoma; ocular pemphigus; Mooren's ulcer; ulcerative keratitis; scleritis; Graves' ophthalmopathy; Vogt-Koyanagi-Harada syndrome; sarcoidosis; pollen allergies; reversible obstructive airway disease; bronchial asthma; allergic asthma; intrinsic asthma; extrinsic asthma; dust asthma; chronic or inveterate asthma; late asthma and airway hyper- responsiveness; bronchitis; gastric ulcers; ischemic bowel diseases; inflammatory bowel diseases; necrotizing enterocolitis; intestinal lesions associated with thermal burns; celiac diseases; proctitis; eosinophilic gastroenteritis; mastocytosis; Crohn's disease; ulcerative colitis; vascular damage caused by ischemic diseases and thrombosis; atherosclerosis; fatty heart; myocarditis; cardiac infarction; arteriosclerosis; aortitis syndrome; cachexia due to viral disease; vascular thrombosis; migraine; rhinitis; eczema; interstitial nephritis; IgA- induced nephropathy; Goodpasture's syndrome; hemolytic-uremic syndrome; diabetic nephropathy; glomerulosclerosis; glomerulonephritis; multiple myositis; Guillain-Barre syndrome; Meniere's disease; polyneuritis; multiple neuritis; mononeuritis; radiculopathy; hyperthyroidism; Basedow's disease; thyrotoxicosis; pure red cell aplasia; aplastic anemia; hypoplastic anemia; idiopathic thrombocytopenic purpura; autoimmune hemolytic anemia; agranulocytosis; pernicious anemia; megaloblastic anemia; anerythroplasia; osteoporosis; sarcoidosis; fibroid lung; idiopathic interstitial pneumonia; dermatomyositis; leukoderma vulgaris; ichthyosis vulgaris; photoallergic sensitivity; cutaneous T cell lymphoma; polyarteritis nodosa; Huntington's chorea; Sydenham's chorea; myocardosis; scleroderma; Wegener's granuloma; Sjogren's syndrome; adiposis; eosinophilic fascitis; lesions of gingiva, periodontium, alveolar bone, substantia ossea dentis; male pattern alopecia or alopecia senilis; muscular dystrophy; pyoderma; Sezar s syndrome; chronic adrenal insufficiency; Addison's disease; ischemia-reperfusion injury of organs which occurs upon preservation; endotoxin shock; pseudomembranous colitis; colitis caused by drug or radiation; ischemic acute renal insufficiency; chronic renal insufficiency; lung cancer; malignancy of lymphoid origin; acute or chronic lymphocytic; leukemias; lymphoma; psoriasis; inflammatory lung injury, pulmonary emphysema; cataracta; siderosis; retinitis pigmentosa; senile macular degeneration; vitreal scarring; inflammatory eye disease; corneal alkali burn; dermatitis erythema; ballous dermatitis; cement dermatitis; gingivitis; periodontitis; sepsis; pancreatitis; carcinogenesis; metastasis of carcinoma; hypobaropathy; autoimmune hepatitis; primary biliary cirrhosis; sclerosing cholangitis; partial liver resection; acute liver necrosis; cirrhosis; alcoholic cirrhosis; hepatic failure; fulminant hepatic failure; late-onset hepatic failure; "acute-on-chronic" liver failure. In yet further embodiments, the malcondition is one or more of rejection of transplanted organs or tissue; graft- versus-host diseases brought about by transplantation; autoimmune syndromes including rheumatoid arthritis, multiple sclerosis, myasthenia gravis; pollen allergies; type I diabetes; prevention of psoriasis; Crohn's disease; ulcerative colitis, acute respiratory distress syndrome; adult respiratory distress syndrome; influenza; post-infectious autoimmune diseases including rheumatic fever and post-infectious glomerulonephritis; and metastasis of carcinoma. In yet further empbodiments the malcondition is one of influenza, ulcerative colitis, multiple sclerosis, transplant rejection, acute respiratory distress syndrome or adult respiratory distress syndrome.
[0155] In certain embodiments, methods are provided for use of an invention compound for preparation of a medicament adapted for treatment of a disorder or a malcondition wherein activation or inhibition of a sphingosine-1 -phosphate receptor subtype 1 is medically indicated.
[0156] In certain embodiments the invention provides a method for the chiral synthesis of a compound comprising an indane moiety having a chiral carbon in the five-member ed ring of the indane moiety where the compound is enantiomerically enriched with respect to the chiral carbon. In such embodiments, the method of the invention provides the steps of
(i) providing a compound comprising an indane moiety where the ring carbon of the five-membered ring of the indane moiety where chiral substitution is desired is oxo substituted at such carbon, and wherein a carbon of the phenyl ring is halo substituted; (ii) reacting such compound with a chiral reagent selected from the group consisting of a Corey Bakshita Shibata-oxazaborolidine and a chiral sulfanamide of the form RS(=0)NH2 where R is selected from the group consisting of t-butyl, branched C2-6 alkyl and C3-8 cycloalkyl; and
(iii) forming the chiral center at the indane moiety carbon previously bound to the oxo group by either reacting such compound with a suitable reducing agent along with the chiral reagent in step (ii) or reacting the result of the reaction of such compound with a suitable reducing agent.
[0157] In certain embodiments R is t-butyl, sec-butyl, isopropyl, cyclopropyl, adamantyl, C3-6 branched alkyl, or optionally bridged C3-8 cycloalkyl. In certain of such embodiments the chiral reagent is the Corey Bakshita Shibata-oxazaborolidine and the compound comprising an indane moiety is enantiomerically enriched with respect to a carbon-oxygen bond on a ring carbon of the five-membered ring of the indane moiety. In certain of such embodiments a suitable reducing reagent includes a borohydride such as BH3-DMS or NaB¾.
[0158] In further embodiments, the chiral reagent is (i?)-(-)-(2)-methyl-CBS- oxazaborolidine or (5 -(-)-(2)-methyl-CBS-oxazaborolidine.
[0159] In certain of such embodiments the compound comprising an indane moiety provided in step (i) is contacted with the chiral reagent to form in step (ii) Formula VI-R or VI-S:
Figure imgf000047_0001
wherein Z is CI, Br or I.
[0160] In certain embodiments the method further comprises the step of protecting the hydroxy group of Formula VI-R or VI-S by treating Formula VI-R or VI-S with a protecting agent to form Formula -R or Vla-S:
Figure imgf000047_0002
wherein PG is a protecting group.
[0161] Protecting groups can render chemical functionality inert to specific reaction conditions and can be appended to and removed from such functionality in a molecule without substantially damaging the remainder of the molecule. Practitioners in the art would be familiar with suitable protecting groups for use in the synthetic methods of the invention. See, e.g., Greene and Wuts, Protective Groups in Organic Synthesis, 2nd ed., John Wiley & Sons, New York, 1991. In certain embodiments such protecting agent is t-butyldimethylsilyl chloride (TBSC1).
[0162] In certain embodiments the method further comprises the step of reacting Formula VTa-R or Vla-S with boronic acid or bis(pinacolato)diboron to form a boronic acid or boronate ester of F
Figure imgf000048_0001
[0163] In certain embodiments the chiral reagent is RS(=0)NH2 and the compound comprising an indane moiety is enantiomerically enriched with respect to a carbon-nitrogen bond on a ring carbon of the five-membered ring of the indane moiety. In further embodiments the chiral reagent is t-Bu-S(=0)NH2.
[0164] In certain embodiments the compound comprising an indane moiety provided in step (i) is contacted with the chiral reagent to form in step (ii) Form -R or VII-S:
Figure imgf000048_0002
wherein Z is CI, Br or I.
[0165] In certain embodiments a compound of Formula VIII-R or VHI-S is formed in step (iii):
Figure imgf000049_0001
[0166] In certain embodiments the method further comprises the step of contacting Formula VIII-R or VIII-S with 1 ,4-dioxane in the presence of an acid to form Formula Vlb- R or VIb-S or Formul -R or IX-S:
Figure imgf000049_0002
[0167] In certain embodiments the method further comprises the step of protecting the amino group by treating Formula IX-R or IX-S with a protecting agent to form Formula IXa- R or IXa-S:
Figure imgf000049_0003
[0168] In certain of such embodiments the protecting agent is di-tert-butyldicarbonate.
[0169] In certain embodiments the method further comprises the step of reacting Formula IXa-R or IXa-S with boronic acid or bis(pinacolato)diboron to form a boronic acid or boronate ester of
Figure imgf000049_0004
[0170] In certain embodiments the method further comprises the step of reacting Formula VIb-R, Formula VIb-S, Formula IXb-R or Formula IXb-S with Formula XI:
Figure imgf000050_0001
[0171] to form Formula XII-R or XH-S:
Figure imgf000050_0002
wherein each A and each A is independently N, or CH; R is di-substituted phenyl or di-substituted pyridinyl where the phenyl and pyridinyl substituents are each independently selected from the group consisting of halo, nitro, cyano, perfluromethyl, fluorinated methyl, and Ci-4-alkoxy; provided that if R1 is di- substituted phenyl, such phenyl is para-substituted with d-4-alkoxy; and X is NH or O.
[0172] In further embodiments R1 is di-substituted phenyl where the phenyl substituents are F and Y, wherein Y is -CN, -CI, or -CF3. In still further embodiment Y is -CN.
[0173] In certain embodiments the method further comprises the step of reacting Formula XII-R -S with /PrOH in the presence of NaO/Pr to from Formula XIII-R or XIH-S:
Figure imgf000050_0003
S.
[0174] In certain embodiments the method further comprises the step of deprotecting the hydroxyl group wherein X is O, or the amino group wherein X is NH, by treating Formula XIII-R or XIII-S with a deprotecting agent. In further embodiments the method further comprises the step of converting the deprotected amino group to a secondary amine.
[0175] In certain embodiments A1 is N and A2 is N. In certain of such embodiments Formula XI is prepared following the process comprising the step of a) treating a di-substituted benzaldehyde with potassium phosphate monobasic to form a di-substituted benzoic acid; b) contacting the di-substituted benzoic acid with H2NNHCSNH2 to form an amino- 1,3-4-thiadizole having a di-substituted phenyl group substituted on the thiadiazole moiety; and c) treating the amino- 1,3-4-thiadizole in step b) with a mixture of copper bromide and isoamylnitrite.
[0176] In certain embodiments A1 is N and A2 is CH. In certain of such embodiments Formula XI is prepared following the process comprising the step of
a) contacting 2-bromotbiazole with a (di-substituted phenyl)boronic acid to form a 2-(di-substituted phenyl)thiazole; and b) treating the 2-(di-substituted phenyl)thiazole with NBS.
[0177] In certain embodiments A1 is CH and A2 is N. In certain of such embodiments Formula XI is prepared following the process comprising the step of
a) contacting 5-(tributylstannyl)thiazole with an iodobenzene having two other substituents to form a 5-(di-substituted phenyl)thiazole; and b) treating the 2-(di-substituted phenyl)thiazole with NBS.
[0178] In certain embodiments, the method of the invention provides the steps of
(i) providing the compound
Figure imgf000051_0001
(ϋ) reacting such compound with a chiral reagent selected from the group consisting of a Corey Bakshita Shibata-oxazaborolidine and a chiral sulfanamide of the form RS(=0)NH2 where R is a bulky group [e.g. t-butyl, branched alkyl or cycloalkyl]; and
(iii) forming a chiral center at the indane moiety carbon previously bound to the oxo group by either reacting such compound with a suitable reducing agent along with the chiral reagent in step (ii) or reacting the result of the reaction of such compound with a suitable reducing agent.
[0179] In certain of such embodiments, the chiral reagent is a Corey Bakshita Shibata- oxazaborolidine and X is -OR'". In further embodiments, the chiral reagent is (R)-(-)-(2)- methyl-CBS-oxazaborolidine or (S)-(-)-(2)-methyl-CBS-oxazaborolidine.
[0180] In certain of such embodiments the chiral reagent is RS(=0)NH2 where R is branched alkyl or cycloalkyl and X is -NR' R". In further such embodiments, the chiral reagent is t-Bu-S(=0)NH2.
[0181] In certain of such embodiments a suitable reducing reagent includes a borohydride such as BH3-DMS or NaB¾.
[0182] Additional steps for the preparation of such compounds can be adapted from the synthetic methods disclosed herein including recrystallization and other processes for purification.
[0183] In certain of such embodiments the invention provides a method of synthesizing a chiral compound of the invention by (i) providing a compound comprising an indane moiety where the ring carbon of the five-membered ring of the indane moiety where chiral substitution is desired is oxo substituted at such carbon; (ii) reacting such compound with a chiral reagent selected from the group consisting of a Corey Bakshita Shibata- oxazaborolidine and a chiral sulfanamide of the form RS(=0)NH2 where R is a bulky group [e.g. t-butyl or other branched alkyl or cycloalkyl]; and (iii) forming a chiral center at the indane moiety carbon previously bound to the oxo group by either reacting such compound with a suitable reducing agent along with the chiral reagent in step (ii) or reacting the result of the reaction of such compound with a suitable reducing agent.
[0184] Additional steps for the preparation of such compounds can be adapted from the synthetic methods disclosed herein including recrystallization and other processes for purification.
[0185] In certain of such embodiments the invention provides a method of synthesizing a chiral compound of the invention by (i) providing a compound comprising an indane moiety where the ring carbon of the five-membered ring of the indane moiety where chiral substitution is desired is oxo substituted at such carbon; (ii) reacting such compound with a chiral reagent selected from the group consisting of a Corey Bakshita Shibata- oxazaborolidine and a chiral sulfinamide of the form RS(=0)NH2 where R is a bulky group [e.g. t-butyl or other branched alkyl or cycloalkyl]; and (iii) forming a chiral center at the indane moiety carbon previously bound to the oxo group by either reacting such compound with a suitable reducing agent along with the chiral reagent in step (ii) or reacting the result of the reaction of such compound with a suitable reducing agent.
[0186] In certain of such embodiments, the invention provides a method for chiral synthesis of a chiral compound comprising an indane moiety having a chiral carbon in the five-membered ring of the indane moiety or a chiral compound comprising an oxadiazole- indane moiety having a chiral carbon in the five-membered ring of the indane moiety where the chiral compound has an enantiomeric enrichment of at least 75%, 85%, 90%, 95%, 98%, or 99%.
[0187] In certain of such embodiments, the invention provides a method for synthesis of a chiral compound of the invention having an enantiomeric enrichment of at least 75%, 85%, 90%, 95%, 98%, or 99%.
[0188] In certain embodiments, a method for the synthesis of a compound comprising an indane moiety having a chiral carbon in the five-membered ring of the indane moiety where the compound is enantiomerically enriched with respect to the chiral carbon is provided. In certain embodiments, a method comprising a step of providing a compound of the structures described herein is provided.
[0189] In certain embodiments the invention provides a method for the chiral synthesis of a compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six- membered saturated ring of the tetrahydronaphthalene moiety where the compound is enantiomerically enriched with respect to the chiral carbon, the method comprising the steps of
(i) providing a compound comprising a tetrahydronaphthalene moiety where the ring carbon of the six-membered saturated ring of the tetrahydronaphthalene moiety where chiral substitution is desired is oxo substituted at such carbon;
(ii) reacting such compound with a chiral reagent to form a chiral center at the tetrahydronaphthalene moiety carbon previously bound to the oxo group.
[0190] In certain embodiments the chiral reagent is RuCl(p-cymene)[(i?,^)-Ts-DPEN] or RuCl(p-cymene)[(S, ^-Ts-DPEN] . [0191] In certain of such embodiments the compound comprising a tetrahydronaphthalene moiety provided in step (i) is contacted with the chiral reagent to form in step (ii) For -R or XTV-S:
Figure imgf000054_0001
[0192] In certain embodiments the method further comprises the step of reversing the chiral configuration of the chiral carbon in the six-membered saturated ring of the tetrahydronaphthalene moiety that was previously bound to the oxo group by treating Formula XIV-R or XIV-S with diphenylphosphoryl azide (DPP A) to form an azido tetrah drona hthalene of Formula XV-S r -R:
Figure imgf000054_0002
where the azido substituent in the six-membered saturated ring of the tetrahydronaphthalene moiety replacing the hydroxy substituent and the chiral carbon that is bound to the azido substituent has a reverse chiral configuration of the chiral carbon when it was previously bound to the hydroxy substituent .
[0193] In certain embodiments the method further comprises the step of converting the azido tetrahydronaphthalene of Formula XV-S or XV-R to an amine substituted tetrahydronaphthalene of Formula XVI-S or XVI-R:
Figure imgf000054_0003
wherein Xm is NH-PG, NH2 or NRTT; R' is H, C1-4 alkyl, n-hydroxy C alkyl, -S02-R3, or -CO-R3; R" is H, -SO2-R5 , C alkyl optionally substituted with 1 or more R4, or a ring moiety optionally substituted with R6 wherein such ring moiety is pyridinyl, oxazolyl, piperidinyl, cyclohexyl, morpholinyl, thiazolyl, pyrazolyl, pyrrolidinyl, imidazolyl, or phenyl; and PG is a protecting group. In certain of such embodiments, Xm is NH-PG.
[0194] In certain embodiments, the step of converting the azido tetrahydronaphthalene of Formula XV-S or XV-R to an amine substituted tetrahydronaphthalene of Formula XVI-S or XVI-R comprises contacting the azido tetrahydronaphthalene of Formula XV-S or XV-R with a carbonate ester. In certain of such embodiments, the carbonate ester is di-tert-butyl dicarbonate.
[0195] In certain embodiments the method further comprises the step of reacting Formula XVI-S or XVI-R with a boronic acid to form a boronic acid of Formula XVII-S or XVII-R:
Figure imgf000055_0001
In certain of such embodiments, the boronic acid is pinacolborane or bis(pinacolato)diboron.
[0196] In certain embodiments the method further comprises the step of forming a substituted 1,3,4-thiadiazole on the tetrahydronaphthalene moiety by reacting an intermediate of XVII-S or XVII-R with a substituted 2-bromo- 1,3,4-thiadiazole of Formula XVTV and a coupling reagent to form a compound of Formula XW-S or XW-R:
Figure imgf000055_0002
wherein Ym is F or -ORa; and Ra is lower alkyl. In certain embodiments, the coupling reagent comprises a palladium. In certain embodiments, the coupling reagent comprises tetrakis (triphenylphosphine) palladium.
[0197] In certain specific embodiments the method provides compound of Formula XW-S -R:
Figure imgf000055_0003
[0198] In certain embodiments the invention provides the method of synthesizing a chiral compound where the compound provided in step (i) is
Figure imgf000056_0001
[0199] In certain embodiments the invention provides the method where the resulting compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six- membered saturated ring of the tetrahydronaphthalene moiety is enantiomerically enriched at least 90%. In certain of such embodiments the resulting compound is enantiomerically enriched at least 95%. In certain of such embodiments the resulting compound is enantiomerically enriched at least 98%. In certain of such embodiments the resulting compound is enantiomerically enriched at least 99%.
[0200] In certain embodiments, the invention provides compounds which can be intermediates in the herein described methods for chiral syntheses. In certain of such embodiments, the invention provides one or more of the following intermediate compounds:
Figure imgf000057_0001
[0201] In certain of such embodimetnns, the invention provides a method for the synthesis of a compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six-membered saturated ring of the tetrahydronaphthalene moiety where the compound is enantiomerically enriched with respect to the chiral carbon, the method comprising a step of providing an intermediate compound.
[0202] In certain of such embodiments, the invention provides a method for chiral synthesis of a chiral compound comprising an tetrahydronaphthalene moiety having a chiral carbon in the six-membered ring of the tetrahydronaphthalene moiety or a chiral compound comprising an 1,3,4-thiadiazole-tetrahydronaphthalene moiety having a chiral carbon in the six-membered ring of the tetrahydronaphthalene moiety where the chiral compound has an enantiomeric enrichment of at least 75%, 85%, 90%, 95%, 98%, or 99%.
[0203] In certain of such embodiments, the invention provides a method for synthesis of a chiral compound of the invention having an enantiomeric enrichment of at least 75%, 85%, 90%, 95%, 98%, or 99%.
[0204] In certain embodiments, a method for the synthesis of a compound comprising an tetrahydronaphthalene moiety having a chiral carbon in the six-membered ring of the tetrahydronaphthalene moiety where the compound is enantiomerically enriched with respect to the chiral carbon is provided. In certain embodiments, a method comprising a step of providing a compound of the structures described herein is provided.
[0205] As used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise.
[0206] As used herein, "individual" (as in the subject of the treatment) means both mammals and non-mammals. Mammals include, for example, humans; non-human primates, e.g. apes and monkeys; cattle; horses; sheep; and goats. Non-mammals include, for example, fish and birds.
[0207] The term " SlPt" as used herein refers to subtype 1 of a sphingosine-1 -phosphate receptor, while other sphingosine-1 -phosphate receptor subtypes are referred to in a corresponding manner, for example, sphingosine-1 -phosphate receptor subtype 3 is referred to as " S1P3".
[0208] A "receptor", as is well known in the art, is a biomolecular entity usually comprising a protein that specifically binds a structural class of ligands or a single native ligand in a living organism, the binding of which causes the receptor to transduce the binding signal into another kind of biological action, such as signaling a cell that a binding event has occurred, which causes the cell to alter its function in some manner. An example of transduction is receptor binding of a ligand causing alteration of the activity of a "G-protein" in the cytoplasm of a living cell. Any molecule, naturally occurring or not, that binds to a receptor and activates it for signal transduction, is referred to as an "agonist" or "activator." Any molecule, naturally occurring or not, that binds to a receptor, but does not cause signal transduction to occur, and which can block the binding of an agonist and its consequent signal transduction, is referred to as an "antagonist."
[0209] An "SIPj compound" or "SIPi agonist" or "SlPj activator" or "S
Figure imgf000059_0001
inhibitor" or "SI Pi antagonist" as the terms are used herein refer to compounds that interact in some way with the SIP receptor subtype 1. They can be agonist or activators, or they can be antagonists or inhibitors. An "SIPi compound" of the invention can be selective for action on subtype 1 of the SIP receptor family; for example a compound of the invention can act at a lower concentration on subtype 1 of the S IP receptor family than on other subtypes of the SIP receptor family; more specifically, an "SIPi compound" of the invention can selectively act on subtype 1 receptors compared to its action on subtype 3, or "S1P3" receptors.
[0210] In certain embodiments, compounds of the invention are orthostatic agonists. In certain other embodiments, compounds of the invention are allostenc agonists. Receptor agonists may be classified as either orthosteric or allosteric. An orthosteric agonist binds to a site in the receptor that significantly overlaps with the binding of the natural ligand and replicates the key interactions of the natural ligand with the receptor. An orthosteric agonist will activate the receptor by a molecular mechanism similar to that of the natural ligand, will be competitive for the natural ligand, and will be competitively antagonized by pharmacological agents that are competitive antagonists for the natural ligand. An allosteric agonist binds to a site in the receptor that makes some significant interactions that are partly or wholly non-overlapping with the natural ligand. Allosteric agonists are true agonists and not allosteric potentiators. Consequently, they activate receptor signaling alone and without a requirement for a sub-maximal concentration of the natural ligand. Allosteric agonists may be identified when an antagonist known to be competitive for the orthosteric ligand shows non-competitive antagonism. The allosteric agonist site can also be mapped by receptor mutagenesis. The introduction of single point mutations in receptors that retain receptor activation by allosteric agonist, while diminishing or abolishing signaling induced by orthosteric agonist or vice versa provide formal evidence for differences in binding interactions. Orthosteric agonists may destabilize GPCR structure and conformation, while allosteric agonists may either stabilize or destabilize GPCR structure and conformation. Allosteric agonists, by virtue of their different interactions with receptor, may be pharmaceutically useful because the allosteric site may confer additional opportunities for agonist potency and selectivity within a related family of receptor subtypes that share a similar orthosteric ligand. In addition, the allosteric site may require very different physical and chemical properties of an agonist compared to the orthosteric ligand. These chemico- physical properties, which include hydrophobicity, aromaticity, charge distribution and solubility may also provide advantages in generating agonists of varying pharmacokinetic, oral bioavailability, distributional and metabolism profiles that facilitate the development of effective pharmaceutical substances.
[0211] "Substantially" as the term is used herein means completely or almost completely; for example, a composition that is "substantially free" of a component either has none of the component or contains such a trace amount that any relevant functional property of the composition is unaffected by the presence of the trace amount, or a compound is "substantially pure" is there are only negligible traces of impurities present.
[0212] Substantially enantiomerically pure means a level of enantiomeric enrichment of one enantiomer with respect to the other enantiomer of at least 90%, 95%, 98%, 99%, 99.5% or 99.9%.
[0213] "Treating" or "treatment" within the meaning herein refers to an alleviation of symptoms associated with a disorder or disease, or inhibition of further progression or worsening of those symptoms, or prevention or prophylaxis of the disease or disorder.
[0214] The expression "effective amount", when used to describe use of a compound of the invention in providing therapy to a patient suffering from a disorder or malcondition mediated by a sphingosine-l-phospate receptor of subtype 1 refers to the amount of a compound of the invention that is effective to bind to as an agonist or as an antagonist a SIPi receptor in the individual's tissues, wherein the SlPj is implicated in the disorder, wherein such binding occurs to an extent sufficient to produce a beneficial therapeutic effect on the patient. Similarly, as used herein, an "effective amount" or a "therapeutically effective amount" of a compound of the invention refers to an amount of the compound that alleviates, in whole or in part, symptoms associated with the disorder or condition, or halts or slows further progression or worsening of those symptoms, or prevents or provides prophylaxis for the disorder or condition. In particular, a "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result by acting as an agonist of sphingosine-1 -phosphate receptor subtype 1 (SI Pi) activity. A therapeutically effective amount is also one in which any toxic or detrimental effects of compounds of the invention are outweighed by the therapeutically beneficial effects. For example, in the context of treating a malcondition mediated by activation of SI Pi, a therapeutically effective amount of an SI Pi agonist of the invention is an amount sufficient to control the malcondition, to mitigate the progress of the malcondition, or to relieve the symptoms of the malcondition. Examples of malconditions that can be so treated include multiple sclerosis, transplant rejection, adult respiratory distress syndrome.
[0215] Diseases, disorders and conditions which may be treated by compounds of the invention include rejection of transplanted organs or tissue; graft- versus-host diseases brought about by transplantation; autoimmune syndromes including rheumatoid arthritis; acute respiratory distress syndrome; adult respiratory distress syndrome; influenza; cancer; systemic erythematosus; Hashimoto's thyroiditis; lymphocytic thyroiditis; multiple sclerosis; myasthenia gravis; type I and II diabetes; uveitis; posterior uveitis; uveitis associated with Behcet's disease; uveomeningitis syndrome; allergic encephalomyelitis; chronic allograft vasculopathy; post-infectious autoimmune diseases including rheumatic fever and postinfectious glomerulonephritis; inflammatory and hyperproliferative skin diseases; cutaneous manifestations of immunologically-mediated disorders; psoriasis; atopic dermatitis; osteomyelitis; contact dermatitis; eczematous dermatitis; seborrhoeic dermatitis; lichen planus; pemphigus; bullous pemphigoid; epidermolysis bullosa; urticaria; angioedema; vasculitis; erythema; cutaneous eosinophilia; acne; alopecia areata; keratoconjunctivitis; vernal conjunctivitis; keratitis; herpetic keratitis; dystrophia epithelialis corneae; corneal leukoma; ocular pemphigus; Mooren's ulcer; ulcerative keratitis; scleritis; Graves' ophthalmopathy; Vogt- oyanagi-Harada syndrome; sarcoidosis; pollen allergies; reversible obstructive airway disease; bronchial asthma; allergic asthma; intrinsic asthma; extrinsic asthma; dust asthma; chronic or inveterate asthma; late asthma and airway hyper- responsiveness; bronchitis; gastric ulcers; ischemic bowel diseases; inflammatory bowel diseases; necrotizing enterocolitis; intestinal lesions associated with thermal burns; celiac diseases; proctitis; eosinophilic gastroenteritis; mastocytosis; Crohn's disease; ulcerative colitis; vascular damage caused by ischemic diseases and thrombosis; atherosclerosis; fatty heart; myocarditis; cardiac infarction; arteriosclerosis; aortitis syndrome; cachexia due to viral disease; vascular thrombosis; migraine; rhinitis; eczema; interstitial nephritis; IgA- induced nephropathy; Goodpasture's syndrome; hemolytic-uremic syndrome; diabetic nephropathy; glomerulosclerosis; glomerulonephritis; multiple myositis; Guillain-Barre syndrome; Meniere's disease; polyneuritis; multiple neuritis; mononeuritis; radiculopathy; hyperthyroidism; Basedow's disease; thyrotoxicosis; pure red cell aplasia; aplastic anemia; hypoplastic anemia; idiopathic thrombocytopenic purpura; autoimmune hemolytic anemia; agranulocytosis; pernicious anemia; megaloblastic anemia; anerythroplasia; osteoporosis; sarcoidosis; fibroid lung; idiopathic interstitial pneumonia; dermatomyositis; leukoderma vulgaris; ichthyosis vulgaris; photoallergic sensitivity; cutaneous T cell lymphoma; polyarteritis nodosa; Huntington's chorea; Sydenham's chorea; myocardosis; scleroderma; Wegener's granuloma; Sjogren's syndrome; adiposis; eosinophilic fascitis; lesions of gingiva, periodontium, alveolar bone, substantia ossea dentis; male pattern alopecia or alopecia senilis; muscular dystrophy; pyoderma; Sezar s syndrome; chronic adrenal insufficiency; Addison's disease; ischemia-reperfusion injury of organs which occurs upon preservation; endotoxin shock; pseudomembranous colitis; colitis caused by drug or radiation; ischemic acute renal insufficiency; chronic renal insufficiency; lung cancer; malignancy of lymphoid origin; acute or chronic lymphocytic; leukemias; lymphoma; psoriasis; inflammatory lung injury, pulmonary emphysema; cataracta; siderosis; retinitis pigmentosa; senile macular degeneration; vitreal scarring; inflammatory eye disease; corneal alkali burn; dermatitis erythema; ballous dermatitis; cement dermatitis; gingivitis; periodontitis; sepsis; pancreatitis; carcinogenesis; metastasis of carcinoma; hypobaropathy; autoimmune hepatitis; primary biliary cirrhosis; sclerosing cholangitis; partial liver resection; acute liver necrosis; cirrhosis; alcoholic cirrhosis; hepatic failure; mlminant hepatic failure; late-onset hepatic failure; "acute-on-chronic" liver failure. Particularly preferred diseases and conditions which may be treated with compounds of the invention comprise the group consisting of rejection of transplanted organs or tissue; graft-versus-host diseases brought about by transplantation; autoimmune syndromes including rheumatoid arthritis, multiple sclerosis, myasthenia gravis; pollen allergies; type I diabetes; prevention of psoriasis; Crohn's disease; ulcerative colitis, acute respiratory distress syndrome; adult respiratory distress syndrome; influenza; postinfectious autoimmune diseases including rheumatic fever and post-infectious glomerulonephritis; and metastasis of carcinoma.
[0216] Furthermore, compounds of Formula I-R or I-S are also useful, in combination with one or several immunosuppressant agents, for the treatment of diseases, disorders and conditions associated with an activated immune system and selected from the list as above- mentioned. According to a preferred embodiment of the invention, said immunosuppressant agent is selected from the group comprising or consisting of cyclosporin, daclizumab, basiliximab, everolimus, tacrolimus (FK506), azathiopirene, leflunomide, 15- deoxyspergualin, or other immunosuppressant drugs
[0217] All chiral, diastereomeric, racemic forms of a structure are intended, unless a particular stereochemistry or isomeric form is specifically indicated. Compounds used in the present invention can include enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions, at any degree of enrichment. Both racemic and diastereomeric mixtures, as well as the individual optical isomers can be synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these are all within the scope of certain embodiments of the invention.
[0218] The isomers resulting from the presence of a chiral center comprise a pair of non-superimposable isomers that are called "enantiomers." Single enantiomers of a pure compound are optically active, i.e., they are capable of rotating the plane of plane polarized light. Single enantiomers are designated according to the Cahn-Ingold-Prelog system. Once the priority ranking of the four groups is determined, the molecule is oriented so that the lowest ranking group is pointed away from the viewer. Then, if the descending rank order of the other groups proceeds clockwise, the molecule is designated (R) and if the descending rank of the other groups proceeds counterclockwise, the molecule is designated (S). In the examples, the Cahn-Ingold-Prelog ranking is A > B > C > D. The lowest ranking atom, D is oriented away from the viewer.
Figure imgf000063_0001
(.^-configuration (5)-configuration
[0219] "Isolated optical isomer" means a compound which has been substantially purified from the corresponding optical isomer(s) of the same formula. Preferably, the isolated isomer is at least about 80%, more preferably at least 90% pure, even more preferably at least 98% pure, most preferably at least about 99% pure, by weight.
Rotational Isomerism
[0220] It is understood that due to chemical properties {i.e., resonance lending some double bond character to the C-N bond) of restricted rotation about the amide bond linkage (as illustrated below) it is possible to observe separate rotamer species and even, under some circumstances, to isolate such species, example shown below. It is further understood that certain structural elements, including steric bulk or substituents on the amide nitrogen, may enhance the stability of a rotamer to the extent that a compound may be isolated as, and exist indefinitely, as a single stable rotamer. The present invention therefore includes any possible stable rotamers of compounds of the invention which are biologically active in the treatment of a disease, disorder or condition for which a compound of the invention may be effective as described herein.
Figure imgf000064_0001
Regioisomerism
[0221] The preferred compounds of the present invention have a particular spatial arrangement of substituents on the aromatic rings, which is related to the structure activity relationship demonstrated by the compound class. Often such substitution arrangement is denoted by a numbering system; however, numbering systems are often not consistent between different ring systems. In six-membered aromatic systems, the spatial arrangements are specified by the common nomenclature "para" for 1,4-substitution, "meta" for 1,3 -substitution and "ortho" for 1,2-substitution as shown below.
Figure imgf000064_0002
'para" "meta" "ortho"
[0222] All structures encompassed within a claim are "chemically feasible", by which is meant that the structure depicted by any combination or subcombination of optional substituents meant to be recited by the claim is physically capable of existence with at least some stability as can be determined by the laws of structural chemistry and by experimentation. Structures that are not chemically feasible are not within a claimed set of compounds.
[0223] In general, "substituted" refers to an organic group as defined herein in which one or more bonds to a hydrogen atom contained therein are replaced by one or more bonds to a non-hydrogen atom such as, but not limited to, a halogen (i.e., F, CI, Br, and I); an oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy groups, aralkyloxy groups, oxo(carbonyl) groups, carboxyl groups including carboxylic acids, carboxylates, and carboyxlate esters; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfoxide groups, sulfone groups, sulfonyl groups, and sulfonamide groups; a nitrogen atom in groups such as amines, hydroxylamines, nitriles, nitro groups, N-oxides, hydrazides, azides, and enamines; and other heteroatoms in various other groups. Non-limiting examples of substituents that can be bonded to a substituted carbon (or other) atom include F, CI, Br, I, OR', OC(0)N(R')2, CN, CF3, OCF3, R', O, S, C(O), S(O), methylenedioxy, ethylenedioxy, N(R')2, SR', SOR', S02R\ S02N(R')2, S03R*, C(0)R', C(0)C(0)R', C(0)CH2C(0)R', C(S)R*, C(0)OR', OC(0)R', C(0)N(R')2, OCiOWh, C(S)N(R')2, (CH2)0-2NHC(O)R', (CH2)0- 2N(R')N(R')2, N(R')N(R')C(0)R', N(R')N(R')C(0)OR', N(R')N(R')CON(R')2, N(R')S02R', N^SC^R'^, N(R')C(0)OR', N(R')C(0)R', N(R')C(S)R\ N(R')C(0)N(R')2, N(R')C(S)N(R')2, N(COR')COR*, N(OR')R\ C(=NH)N(R')2, C(0)N(OR')R', or C(=NOR')R' wherein R' can be hydrogen or a carbon-based moiety, and wherein the carbon-based moiety can itself be further substituted.
[0224] Substituted alkyl, alkenyl, alkynyl, cycloalkyl, and cycloalkenyl groups as well as other substituted groups also include groups in which one or more bonds to a hydrogen atom are replaced by one or more bonds, including double or triple bonds, to a carbon atom, or to a heteroatom such as, but not limited to, oxygen in carbonyl (oxo), carboxyl, ester, amide, imide, urethane, and urea groups; and nitrogen in imines, hydroxyimines, oximes, hydrazones, amidines, guanidines, and nitriles. The substituents of the substituted groups can further be substituted with alkyl, alkenyl, cycloalkyl, aryl, heteroaryl, and alkynyl groups as defined herein, which can themselves be further substituted. For example, an CM alkyl group can be substituted with an amide, and the amide can further be substituted with another C alkyl, which can further be substituted.
[0225] Substituted ring groups such as substituted aryl, heterocyclyl and heteroaryl groups also include rings and fused ring systems in which a bond to a hydrogen atom is replaced with a bond to a carbon atom. Therefore, substituted aryl, heterocyclyl and heteroaryl groups can also be substituted with alkyl, alkenyl, cycloalkyl, aryl, heteroaryl, and alkynyl groups as defined herein, which can themselves be further substituted.
[0226] The term "heteroatoms" as used herein refers to non-carbon and non-hydrogen atoms, capable of forming covalent bonds with carbon, and is not otherwise limited. Typical heteroatoms are N, O, and S. When sulfur (S) is referred to, it is understood that the sulfur can be in any of the oxidation states in which it is found, thus including sulfoxides (R-S(O)- R') and sulfones (R-S(0)2-R'), unless the oxidation state is specified; thus, the term "sulfone" encompasses only the sulfone form of sulfur; the term "sulfide" encompasses only the sulfide (R-S-R1) form of sulfur. When the phrases such as "heteroatoms selected from the group consisting of O, NH, NR' and S," or "[variable] is O, S . . ." are used, they are understood to encompass all of the sulfide, sulfoxide and sulfone oxidation states of sulfur. [0227] Alkyl groups include straight chain and branched alkyl groups and cycloalkyl groups having from 1 to about 20 carbon atoms (Ci-2o alkyl), and typically from 1 to 12 carbons (C1-12 alkyl)or, in some embodiments, from 1 to 8 carbon atoms (C1-8 alkyl) or, in some embodiments, from 1 to 4 carbon atoms (C1-4 alkyl) or, in some embodiments, from 1 to 3 carbon atoms (Ci-3 alkyl). Examples of straight chain alkyl groups include, but are not limited to methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, and n-octyl groups. Examples of branched alkyl groups include, but are not limited to, isopropyl, iso-butyl, sec- butyl, t-butyl, neopentyl, isopentyl, and 2,2-dimethylpropyl groups. Representative substituted alkyl groups can be substituted one or more times with any of the groups listed above, for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups. The group "n-hydroxy C^ alkyl" represents an C1- alkyl substituted with a terminal hydroxy group.
[0228] Cycloalkyl groups are alkyl groups forming a ring structure, which can be substituted or unsubstituted. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl groups. In some embodiments, the cycloalkyl group has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms range from 3 to 5, 3 to 6, or 3 to 7. Cycloalkyl groups further include polycyclic cycloalkyl groups such as, but not limited to, norbomyl, adamantyl, bornyl, camphenyl, isocamphenyl, and carenyl groups, and fused rings such as, but not limited to, decalinyl, and the like. Cycloalkyl groups also include rings that are substituted with straight or branched chain alkyl groups as defined above. Representative substituted cycloalkyl groups can be mono-substituted or substituted more than once, such as, but not limited to, 2,2-, 2,3-, 2,4- 2,5- or 2,6-disubstituted cyclohexyl groups or mono-, di- or tri-substituted norbomyl or cycloheptyl groups, which can be substituted with, for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups.
[0229] The terms "carbocyclic" and "carbocycle" denote a ring structure wherein the atoms of the ring are carbon. In some embodiments, the carbocycle has 3 to 8 ring members, whereas in other embodiments the number of ring carbon atoms is 4, 5, 6, or 7. Unless specifically indicated to the contrary, the carbocyclic ring can be substituted with as many as N substituents wherein N is the size of the carbocyclic ring with for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups.
[0230] (Cycloalkyl)alkyl groups, also denoted cycloalkylalkyl, are alkyl groups as defined above in which a hydrogen or carbon bond of the alkyl group is replaced with a bond to a cycloalkyl group as defined above. [0231] Alkenyl groups include straight and branched chain and cyclic alkyl groups as defined above, except that at least one double bond exists between two carbon atoms. Thus, alkenyl groups have from 2 to about 20 carbon atoms, and typically from 2 to 12 carbons or, in some embodiments, from 2 to 8 carbon atoms. Examples include, but are not limited to -CH=CH(CH3), -CH=C(CH3)2, -C(CH3)-CH2, -C(CH3)=CH(CH3), -C(CH2CH3)=CH2, vinyl, cyclohexenyl, cyclopentenyl, cyclohexadienyl, butadienyl, pentadienyl, and hexadienyl among others.
[0232] The term "cycloalkenyl" alone or in combination denotes a cyclic alkenyl group wherein at least one double bond is present in the ring structure. Cycloalkenyl groups include cycloalkyl groups having at least one double bond between two adjacent carbon atoms. Thus for example, cycloalkenyl groups include but are not limited to cyclohexenyl, cyclopentenyl, and cyclohexadienyl groups.
[0233] (Cycloalkenyl)alkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of the alkyl group is replaced with a bond to a cycloalkenyl group as defined above.
[0234] Alkynyl groups include straight and branched chain alkyl groups, except that at least one triple bond exists between two carbon atoms. Thus, alkynyl groups have from 2 to about 20 carbon atoms, and typically from 2 to 12 carbons or, in some embodiments, from 2 to 8 carbon atoms. Examples include, but are not limited to -C≡CH, -C≡ (CH3), - C≡C(CH2CH3), -CH2C≡CH, -CH2C≡C(CH3), and -CH2C≡C(CH2C¾), among others.
[0235] Aryl groups are cyclic aromatic hydrocarbons that do not contain heteroatoms. Thus aryl groups include, but are not limited to, phenyl, azulenyl, heptalenyl, biphenyl, indacenyl, fluorenyl, phenanthrenyl, triphenylenyl, pyrenyl, naphthacenyl, chrysenyl, biphenylenyl, anthracenyl, and naphthyl groups. In some embodiments, aryl groups contain 6-14 carbons in the ring portions of the groups. The phrase "aryl groups" includes groups containing fused rings, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like), and also includes substituted aryl groups that have other groups, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups, bonded to one of the ring atoms. Representative substituted aryl groups can be mono-substituted or substituted more than once, such as, but not limited to, 2-, 3-, 4-, 5-, or 6-substituted phenyl or naphthyl groups, which can be substituted with groups including but not limited to those listed above. [0236] Aralkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to an aryl group as defined above. Representative aralkyl groups include benzyl and phenylethyl groups and fused (cycloalkylaryl)alkyl groups such as 4-ethyl-indanyl. The aryl moiety or the alkyl moiety or both are optionally substituted with other groups, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups. Aralkenyl group are alkenyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to an aryl group as defined above.
[0237] Heterocyclyl groups include aromatic and non-aromatic ring compounds (heterocyclic rings) containing 3 or more ring members, of which one or more is a heteroatom such as, but not limited to, N, O, S, or P. In some embodiments, heterocyclyl groups include 3 to 20 ring members, whereas other such groups have 3 to 15 ring members. At least one ring contains a heteroatom, but every ring in a polycyclic system need not contain a heteroatom. For example, a dioxolanyl ring and a benzdioxolanyl ring system (methylenedioxyphenyl ring system) are both heterocyclyl groups within the meaning herein. A heterocyclyl group designated as a C2-heterocyclyl can be a 5-membered ring with two carbon atoms and three heteroatoms, a 6-membered ring with two carbon atoms and four heteroatoms and so forth. Likewise a C4-heterocyclyl can be a 5-membered ring with one heteroatom, a 6-membered ring with two heteroatoms, and so forth. The number of carbon atoms plus the number of heteroatoms sums up to equal the total number of ring atoms. A saturated heterocyclic ring refers to a heterocyclic ring containing no unsaturated carbon atoms.
[0238] The phrase "heterocyclyl group" includes fused ring species including those having fused aromatic and non-aromatic groups. The phrase also includes polycyclic ring systems containing a heteroatom such as, but not limited to, quinuclidyl and also includes heterocyclyl groups that have substituents, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups, bonded to one of the ring members. A heterocyclyl group as defined herein can be a heteroaryl group or a partially or completely saturated cyclic group including at least one ring heteroatom. Heterocyclyl groups include, but are not limited to, pyrrolidinyl, furanyl, tetrahydrofuranyl, dioxolanyl, piperidinyl, piperazinyl, morpholinyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl, dihydrobenzofuranyl, indolyl, dihydroindolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, i
thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, quinoxalinyl, and quinazolinyl groups. Heterocyclyl groups can be substituted. Representative substituted heterocyclyl groups can be 1 mono-substituted or substituted more than once, including but not limited to, rings containing at least one heteroatom which are mono, di, tri, tetra, penta, hexa, or higher-substituted with substituents such as those listed above, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, and alkoxy groups.
[0239] Heteroaryl groups are aromatic ring compounds containing 5 or more ring members, of which, one or more is a heteroatom such as, but not limited to, N, O, and S. A heteroaryl group designated as a C2-heteroaryl can be a 5-membered ring with two carbon atoms and three heteroatoms, a 6-membered ring with two carbon atoms and four heteroatoms and so forth. Likewise a C4-heteroaryl can be a 5-membered ring with one heteroatom, a 6-membered ring with two heteroatoms, and so forth. The number of carbon atoms plus the number of heteroatoms sums up to equal the total number of ring atoms. Heteroaryl groups include, but are not limited to, groups such as pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, 1 azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl,: isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, and quinazolinyl groups. The terms "heteroaryl" and "heteroaryl groups" include fused ring compounds such as wherein at least one ring, but not necessarily all rings, are aromatic, including !tetrahydroquinolinyl, tetrahydroisoquinolinyl, indolyl and 2,3-dihydro indolyl. The term also includes heteroaryl groups that have other groups bonded to one of the ring members, including but not limited to alkyl, halo, amino, hydroxy, cyano, carboxy, nitro, thio, or alkoxy groups. Representative substituted heteroaryl groups can be substituted one or more times with groups such as those listed above.
[0240] Additional examples of aryl and heteroaryl groups include but are not limited to phenyl, biphenyl, indenyl, naphthyl (1-naphthyl, 2-naphthyl), N-hydroxytetrazolyl, N- hydroxytriazolyl, N-hydroxyimidazolyl, anthracenyl (1-anthracenyl, 2-anthracenyl, 3- anthracenyl), thiophenyl (2-thienyl, 3-thienyl), furyl (2-furyl, 3-furyl) , indolyl, oxadiazolyl, isoxazolyl, quinazolinyl, fluorenyl, xanthenyl, isoindanyl, benzhydryl^ acridinyl, thiazolyl, pyrrolyl (2 -pyrrolyl), pyrazolyl (3-pyrazolyl), imidazolyl (1 -imidazolyl, 2-imidazolyl, 4-imidazolyl, 5 -imidazolyl), triazolyl (1,2,3-triazol-l-yl, l,2,3-triazol-2-yl l,2,3-triazol-4-yl, l,2,4-triazol-3-yl), oxazolyl (2 -oxazolyl, 4-oxazolyl, 5-oxazolyl), thiazolyl (2-thiazolyl, 4- thiazolyl, 5-thiazolyl), pyridyl (2-pyridyl, 3-pyridyl, 4-pyridyl), pyrimidinyl (2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl), pyrazinyl, pyridazinyl (3- pyridazinyl, 4- pyridazinyl, 5 -pyridazinyl), quinolyl (2-quinolyl, 3-quinolyl, 4-quinolyl, 5-quinolyl, 6- quinolyl, 7-quinolyl, 8-quinolyl), isoquinolyl (1 -isoquinolyl, 3-isoquinolyl, 4-isoquinolyl, 5- isoquinolyl, 6-isoquinolyl, 7-isoquinolyl, 8-isoquinolyl), benzo[b]furanyl (2-benzo[b]furanyl, 3-benzo[b]furanyl, 4-benzo[b]furanyl, 5-benzo[b]furanyl, 6-benzo[b]furanyl, 7- benzo[b]furanyl), 2,3-dihydro-benzo[b]furanyl (2-(2,3-dihydro-benzo[b]furanyl), 3-(2,3- dihydro-benzo[b]furanyl), 4-(2,3-dihydro-benzo[b]furanyl), 5-(2,3-dihydro-benzo[b]furanyl),
6- (2,3-dihydro-benzo[b]furanyl), 7-(2,3-dihydro-benzo[b]furanyl), benzo[b]thiophenyl (2- benzo[b]thiophenyl, 3-benzo[b]tbiophenyl, 4-benzo[b]thiophenyl, 5-benzo[b]thiophenyl, 6- benzo[b]thiophenyl, 7-benzo[b]thiophenyl), 2,3-dihydro-benzo[b]thiophenyl, (2-(2,3- dihydro-benzo[b]thiophenyl), 3-(2,3-dihydro-benzo[b]thiophenyl), 4-(2,3-dihydro- benzo[b]thiophenyl), 5-(2,3-dihydro-benzo[b]thiophenyl), 6-(2,3-dihydro- benzo[b]thiophenyl), 7-(2,3-dihydro-benzo[b]thiophenyl), indolyl (1-indolyl, 2-indolyl,
3- indolyl, 4-indolyl, 5-indolyl, 6-indolyl, 7-indolyl), indazole (1-indazolyl, 3-indazolyl,
4- indazolyl, 5-indazolyl, 6-indazolyl, 7-indazolyl), benzimidazolyl (1-benzimidazolyl, 2-benzimidazolyl, 4-benzimidazolyl, 5-benzimidazolyl, 6-benzimidazolyl, 7-benzimidazolyl, 8-benzimidazolyl), benzoxazolyl (1-benzoxazolyl, 2-benzoxazolyl), benzothiazolyl (1- benzothiazolyl, 2-benzothiazolyl, 4-benzothiazolyl, 5-benzothiazolyl, 6-benzothiazolyl,
7- benzothiazolyl), carbazolyl (1-carbazolyl, 2-carbazolyl, 3-carbazolyl, 4-carbazolyl), 5H-dibenz[b,f]azepine (5H-dibenz[b,f]azepin-l-yl, 5H-dibenz[b,f]azepine-2-yl, 5H-dibenz[b,f]azepine-3-yl, 5H-dibenz[b,f]azepine-4-yl, 5H-dibenz[b,f]azepine-5-yl), 10,1 l-dihydro-5H-dibenz[b,f]azepine (10,1 l-dihydro-5H-dibenz[b,f]azepine-l-yl, 10, 11 -dihydro-5H-dibenz[b,f]azepine-2-yl, 10, 11 -dihydro-5H-dibenz[b,f]azepine-3-yl, 10, 11 -dihydro-5H-dibenzp3,f]azepine-4-yl, 10, 11 -dihydro-5H-dibenz[b,f]azepine-5-yl), and the like.
[0241] Heterocyclylalkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to a heterocyclyl group as defined above. Representative heterocyclyl alkyl groups include, but are not limited to, furan-2-yl methyl, furan-3-yl methyl, pyridine-2-yl methyl (a-picolyl), pyridine-3-yl methyl (β-picolyl), pyridine-4-yl methyl (γ-picolyl), tetrahydrofuran-2-yl ethyl, and indol-2-yl propyl. Heterocyclylalkyl groups can be substituted on the heterocyclyl moiety, the alkyl moiety, or both. [0242] Heteroarylalkyl groups are alkyl groups as defined above in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to a heteroaryl group as defined above. Heteroarylalkyl groups can be substituted on the heteroaryl moiety, the alkyl moiety, or both.
[0243] By a "ring system" as the term is used herein is meant a moiety comprising one, two, three or more rings, which can be substituted with non-ring groups or with other ring systems, or both, which can be fully saturated, partially unsaturated, fully unsaturated, or aromatic, and when the ring system includes more than a single ring, the rings can be fused, bridging, or spirocyclic. By "spirocyclic" is meant the class of structures wherein two rings are fused at a single tetrahedral carbon atom, as is well known in the art.
[0244] A "monocyclic, bicyclic or polycyclic, aromatic or partially aromatic ring" as the term is used herein refers to a ring system including an unsaturated ring possessing 4n+2 pi electrons, or a partially reduced (hydrogenated) form thereof. The aromatic or partially aromatic ring can include additional fused, bridged, or spiro rings that are not themselves aromatic or partially aromatic. For example, naphthalene and tetrahydronaphthalene are both a "monocyclic, bicyclic or polycyclic, aromatic or partially aromatic ring" within the meaning herein. Also, for example, a benzo-[2.2.2]-bicyclooctane is also a "monocyclic, bicyclic or polycyclic, aromatic or partially aromatic ring" within the meaning herein, containing a phenyl ring fused to a bridged bicyclic system. A fully saturated ring has no double bonds therein, and is carbocyclic or heterocyclic depending on the presence of heteroatoms within the meaning herein.
[0245] The term "alkoxy" refers to an oxygen atom connected to an alkyl group, including a cycloalkyl group, as are defined above. Examples of linear alkoxy groups include but are not limited to methoxy, ethoxy, n-propoxy, n-butoxy, n-pentyloxy, n-hexyloxy, and the like. Examples of branched alkoxy include but are not limited to isopropoxy, sec-butoxy, tert-butoxy, isopentyloxy, isohexyloxy, and the like. Examples of cyclic alkoxy include but are not limited to cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
[0246] The terms "aryloxy" and "arylalkoxy" refer to, respectively, an aryl group bonded to an oxygen atom and an aralkyl group bonded to the oxygen atom at the alkyl moiety. Examples include but are not limited to phenoxy, naphthyloxy, and benzyloxy.
[0247] An "acyl" group as the term is used herein refers to a group containing a carbonyl moiety wherein the group is bonded via the carbonyl carbon atom. The carbonyl carbon atom is also bonded to another carbon atom, which can be part of an alkyl, aryl, aralkyl cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl group or the like. In the special case wherein the carbonyl carbon atom is bonded to a hydrogen, the group is a "formyl" group, an acyl group as the term is defined herein. An acyl group can include 0 to about 12-20 additional carbon atoms bonded to the carbonyl group. An acyl group can include double or triple bonds within the meaning herein. An acryloyl group is an example of an acyl group. An acyl group can also include heteroatoms within the meaning here. A nicotinoyl group (pyridyl-3 -carbonyl) group is an example of an acyl group within the meaning herein. Other examples include acetyl, benzoyl, phenylacetyl, pyridylacetyl, cinnamoyl, and acryloyl groups and the like. When the group containing the carbon atom that is bonded to the carbonyl carbon atom contains a halogen, the group is termed a "haloacyl" group. An example is a trifluoroacetyl group.
[0248] The term "amine" includes primary, secondary, and tertiary amines having, e.g., the formula N(group)3 wherein each group can independently be H or non-H, such as alkyl, aryl, and the like. Amines include but are not limited to RNH2, for example, alkylamines, arylamines, alkylarylamines; R2NH wherein each R is independently selected, such as dialkylamines, diarylamines, aralkylamines, heterocyclylamines and the like; and R3N wherein each R is independently selected, such as trialkylamines, dialkylarylamines, alkyldiarylamines, triarylamines, and the like. The term "amine" also includes ammonium ions as used herein.
[0249] An "amino" group is a substituent of the form -NH2, -NHR, -NR2, -NR3 +, wherein each R is independently selected, and protonated forms of each. Accordingly, any compound substituted with an amino group can be viewed as an amine.
[0250] An "ammonium" ion includes the unsubstituted ammonium ion N¾+, but unless otherwise specified, it also includes any protonated or quaternarized forms of amines. Thus, trimethylammonium hydrochloride and tetram ethyl ammonium chloride are both ammonium ions, and amines, within the meaning herein.
[0251] The term "amide" (or "amido") includes C- and N-amide groups, i.e., -C(0)N R'R", and -NR'C(0)R" groups, respectively. The R' and R" of the C-amide may join together to form a heterocyclic ring with the nitrogen atom. Amide groups therefore include but are not limited to carbamoyl groups (-C(0)NH2) and formamide groups (-NHC(O)H). A "carboxamido" group is a group of the formula C(0)NR2, wherein R can be H, alkyl, aryl, etc.
[0252] The term "urethane" (or "carbamyl") includes N- and O-urethane groups, i.e., -NRC(0)OR and -OC(0)NR2 groups, respectively. [0253] The term "sulfonamide" (or "sulfonamido") includes S- and N-sulfonamide groups, i.e., -S02NR2 and -NRS02R groups, respectively. Sulfonamide groups therefore include but are not limited to sulfamoyl groups (-S02NH2).
[0254] The term "amidine" or "amidino" includes groups of the formula -C(NR)NR2. Typically, an amidino group is -C(NH)NH2.
[0255] The term "guanidine" or "guanidino" includes groups of the formula -NRC(NR)NR2. Typically, a guanidino group is -NHC(NH)NH2.
[0256] "Halo," "halogen," and "halide" include fluorine, chlorine, bromine and iodine.
[0257] The terms "comprising," "including," "having," "composed of," are open-ended terms as used herein, and do not preclude the existence of additional elements or components. In a claim element, use of the forms "comprising," "including," "having," or "composed of means that whatever element is comprised, had, included, or composes is not necessarily the only element encompassed by the subject of the clause that contains that word.
[0258] A "salt" as is well known in the art includes an organic compound such as a carboxylic acid, a sulfonic acid, or an amine, in ionic form, in combination with a counterion. For example, acids in their anionic form can form salts with cations such as metal cations, for example sodium, potassium, and the like; with ammonium salts such as NH4 + or the cations of various amines, including tetraalkyl ammonium salts such as tetramethylammonium and alkyl ammonium salts such as tromethamine salts, or other cations such as trimethylsulfonium, and the like. A "pharmaceutically acceptable" or "pharmacologically acceptable" salt is a salt formed from an ion that has been approved for human consumption and is generally non-toxic, such as a chloride salt or a sodium salt. A "zwitterion" is an internal salt such as can be formed in a molecule that has at least two ionizable groups, one forming an anion and the other a cation, which serve to balance each other. For example, amino acids such as glycine can exist in a zwitterionic form. A "zwitterion" is a salt within the meaning herein. The compounds of the present invention may take the form of salts. The term "salts" embraces addition salts of free acids or free bases which are compounds of the invention. Salts can be "pharmaceutically-acceptable salts." The term "pharmaceutically-acceptable salt" refers to salts which possess toxicity profiles within a range that affords utility in pharmaceutical applications. Pharmaceutically unacceptable salts may nonetheless possess properties such as high crystallinity, which have utility in the practice of the present invention, such as for example utility in process of synthesis, purification or formulation of compounds of the invention. [0259] Suitable pharmaceutically-acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include hydrochloric, hydrobromic, hydnodic, nitric, carbonic, sulfuric, and phosphoric acids. Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, trifluoromethanesulfonic, 2-hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic, β-hydroxybutyric, salicylic, galactaric and galacturonic acid. Examples of pharmaceutically unacceptable acid addition salts include, for example, perchlorates and tetrafluoroborates.
[0260] Suitable pharmaceutically acceptable base addition salts of compounds of the invention include, for example, metallic salts including alkali metal, alkaline earth metal and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts. Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, A -dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. Examples of pharmaceutically unacceptable base addition salts include lithium salts and cyanate salts. Although pharmaceutically unacceptable salts are not generally useful as medicaments, such salts may be useful, for example as intermediates in the synthesis of compounds, for example in their purification by recrystallization. All of these salts may be prepared by conventional means from the corresponding compound by reacting, for example, the appropriate acid or base with the compound. The term "pharmaceutically acceptable salts" refers to nontoxic inorganic or organic acid and/or base addition salts, see, for example, Lit et al., Salt Selection for Basic Drugs (1986), IntJ. Pharm., 33, 201-217, incorporated by reference herein
[0261] Nonlimiting examples of potential salts of this invention include but are not limited to hydrochloride, citrate, glycolate, fumarate, malate, tartrate, mesylate, esylate, cinnamate, isethionate, sulfate, phosphate, diphosphate, nitrate, hydrobromide, hydroiodide, succinate, formate, acetate, dichloroacetate, lactate, 7-toluenesulfonate, pamitate, pidolate, pamoate, salicylate, 4-aminosalicylate, benzoate, 4-acetamido benzoate, glutamate, aspartate, glycolate, adipate, alginate, ascorbate, besylate, camphorate, camphorsulfonate, camsylate, caprate, caproate, cyclamate, laurylsulfate, edisylate, gentisate, galactarate, gluceptate, gluconate, glucuronate, oxoglutarate, hippurate, lactobionate, malonate, maleate, mandalate, napsylate, napadisylate, oxalate, oleate, sebacate, stearate, succinate, thiocyanate, undecylenate, and xinafoate.
[0262] A "hydrate" is a compound that exists in a composition with water molecules. The composition can include water in stoichiometic quantities, such as a monohydrate or a dihydrate, or can include water in random amounts. As the term is used herein a "hydrate" refers to a solid form, i.e., a compound in water solution, while it may be hydrated, is not a hydrate as the term is used herein.
[0263J A "homolog" of a compound of the invention is a compound having one or more atoms of the compound replaced by an isotope of such atom. For example, homologs include compounds with deuterium in place of some hydrogen atoms of the compound such as compounds of the invention in which the methyl groups of the isopropoxy moiety of Formulas I-R and I-S are fully or partially deuterated (e.g., (D3C)2C-0-). Isotopic substitutions which may be made in the formation of homologs of the invention include nonradioactive (stable) atoms such as deuterium and carbon 13, as well as radioactive (unstable) atoms such as tritium, carbon 14, iodine 123, iodine 125, etc.
[0264] A "solvate" is a similar composition except that a solvent other that water replaces the water. For example, methanol or ethanol can form an "alcoholate", which can again be stoichiometic or non-stoichiometric. As the term is used herein a "solvate" refers to a solid form, i.e., a compound in solution in a solvent, while it may be solvated, is not a solvate as the term is used herein.
[0265] A "prodrug" as is well known in the art is a substance that can be administered to a patient where the substance is converted in vivo by the action of biochemicals within the patients body, such as enzymes, to the active pharmaceutical ingredient. Examples of prodrugs include esters of carboxylic acid groups, which can be hydrolyzed by endogenous esterases as are found in the bloodstream of humans and other mammals.
[0266] Any compound which can be converted in vivo to the active drug by chemical or biochemical transformations functions as a prodrug. Prodrugs of claimed compounds are covered under this invention.
[0267] Some examples of prodrugs within the scope of this invention include:
i. If the compound contains a hydroxyl group, the hydroxyl group may be modified to form an ester, carbonate, or carbamate. Examples include acetate, pivalate, methyl and ethyl carbonates, and dimethylcarbamate. The ester may also be derived from amino acids such as glycine, serine, or lysine. ii. If the compound contains an amine group, the amine group may be modified to form an amide. Examples include acetamide or derivatization with amino acids such as glycine, serine, or lysine.
[0268] Certain compounds of the invention and their salts may exist in more than one crystal form and the present invention includes each crystal form and mixtures thereof. In addition, the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water to form hydrates or adducts with alcohols such as Ci-4-alkanols, and the like. Furthermore, compounds of this invention can be isolated in association with solvent molecules by crystallization from evaporation of an appropriate solvent. Such solvents include but are not limited to toluene, tetrahydrofuran, dioxane, dimethylformamide, acetonitrile, acetates such as methyl acetate, ethyl acetate, butyl acetate, isobutyl acetate, propyl- and isopropyl acetate, ethers such as diethyl ether and ethyl ether, alcohols such as methanol, ethanol, 1- or 2-butanol, 1- or 2-propanol, pentanol, and dimethylsulfoxide. In general, a depiction for the compound by structure or name is considered to embrace the compound in any form (e.g., by itself, as a hydrate, solvate, or otherwise in a mixture).
[0269] In addition, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group. For example, if X is described as selected from the group consisting of bromine, chlorine, and iodine, claims for X being bromine and claims for X being bromine and chlorine are fully described. Moreover, where features or aspects of the invention are described in terms of Markush groups, those skilled in the art will recognize that the invention is also thereby described in terms of any combination of individual members or subgroups of members of Markush groups. Thus, for example, if X is described as selected from the group consisting of bromine, chlorine, and iodine, and Y is described as selected from the group consisting of methyl, ethyl, and propyl, claims for X being bromine and Y being methyl are fully described.
COMPOSITIONS AND COMBINATION TREATMENTS
[0270] The SlPi compounds, their pharmaceutically acceptable salts or hydro lyzable esters of the present invention may be combined with a pharmaceutically acceptable carrier to provide pharmaceutical compositions useful for treating the biological conditions or disorders noted herein in mammalian species, and more preferably, in humans. The particular carrier employed in these pharmaceutical compositions may vary depending upon the type of administration desired (e.g. intravenous, oral, topical, suppository, or parenteral).
[0271] In preparing the compositions in oral liquid dosage forms (e.g. suspensions, elixirs and solutions), typical pharmaceutical media, such as water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like can be employed. Similarly, when preparing oral solid dosage forms (e.g. powders, tablets and capsules), carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like can be employed.
[0272] Another aspect of an embodiment of the invention provides compositions of the compounds of the invention, alone or in combination with another SI Pi inhibitor or another type of therapeutic agent, or both. As set forth herein, compounds of the invention include stereoisomers, tautomers, solvates, hydrates, salts including pharmaceutically acceptable salts, and mixtures thereof. Compositions containing a compound of the invention can be prepared by conventional techniques, e.g. as described in Remington: The Science and Practice of Pharmacy, 19th Ed., 1995, incorporated by reference herein. The compositions can appear in conventional forms, for example capsules, tablets, aerosols, solutions, suspensions or topical applications.
[0273] Typical compositions include a compound of the invention and a pharmaceutically acceptable excipient which can be a carrier or a diluent. For example, the active compound will usually be mixed with a carrier, or diluted by a carrier, or enclosed within a carrier which can be in the form of an ampoule, capsule, sachet, paper, or other container. When the active compound is mixed with a carrier, or when the carrier serves as a diluent, it can be solid, semi-solid, or liquid material that acts as a vehicle, excipient, or medium for the active compound. The active compound can be adsorbed on a granular solid carrier, for example contained in a sachet. Some examples of suitable carriers are water, salt solutions, alcohols, polyethylene glycols, polyhydroxyethoxylated castor oil, peanut oil, olive oil, gelatin, lactose, terra alba, sucrose, dextrin, magnesium carbonate, sugar, cyclodextrin, amylose, magnesium stearate, talc, gelatin, agar, pectin, acacia, stearic acid or lower alkyl ethers of cellulose, silicic acid, fatty acids, fatty acid amines, fatty acid monoglycerides and diglycerides, pentaerythritol fatty acid esters, polyoxyethylene, hydroxymethylcellulose and polyvinylpyrrolidone. Similarly, the carrier or diluent can include any sustained release material known in the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax. [0274] The formulations can be mixed with auxiliary agents which do not deleteriously react with the active compounds. Such additives can include wetting agents, emulsifying and suspending agents, salt for influencing osmotic pressure, buffers and/or coloring substances preserving agents, sweetening agents or flavoring agents. The compositions can also be sterilized if desired.
[0275] The route of administration can be any route which effectively transports the active compound of the invention which inhibits the enzymatic activity of the focal adhesion kinase to the appropriate or desired site of action, such as oral, nasal, pulmonary, buccal, subdermal, intradermal, transdermal or parenteral, e.g., rectal, depot, subcutaneous, intravenous, intraurethral, intramuscular, intranasal, ophthalmic solution or an ointment, the oral route being preferred.
[0276] For parenteral administration, the carrier will typically comprise sterile water, although other ingredients that aid solubility or serve as preservatives can also be included. Furthermore, injectable suspensions can also be prepared, in which case appropriate liquid carriers, suspending agents and the like can be employed.
[0277] For topical administration, the compounds of the present invention can be formulated using bland, moisturizing bases such as ointments or creams.
[0278] If a solid carrier is used for oral administration, the preparation can be tabletted, placed in a hard gelatin capsule in powder or pellet form or it can be in the form of a troche or lozenge. If a liquid carrier is used, the preparation can be in the form of a syrup, emulsion, soft gelatin capsule or sterile injectable liquid such as an aqueous or non-aqueous liquid suspension or solution.
[0279] Injectable dosage forms generally include aqueous suspensions or oil suspensions which can be prepared using a suitable dispersant or wetting agent and a suspending agent Injectable forms can be in solution phase or in the form of a suspension, which is prepared with a solvent or diluent. Acceptable solvents or vehicles include sterilized water, Ringer's solution, or an isotonic aqueous saline solution. Alternatively, sterile oils can be employed as solvents or suspending agents. Preferably, the oil or fatty acid is non-volatile, including natural or synthetic oils, fatty acids, mono-, di- or tri-glycerides.
[0280] For injection, the formulation can also be a powder suitable for reconstitution with an appropriate solution as described above. Examples of these include, but are not limited to, freeze dried, rotary dried or spray dried powders, amorphous powders, granules, precipitates, or particulates. For injection, the formulations can optionally contain stabilizers, pH modifiers, surfactants, bioavailability modifiers and combinations of these. The compounds can be formulated for parenteral administration by injection such as by bolus injection or continuous infusion. A unit dosage form for injection can be in ampoules or in multi-dose containers.
[0281] The formulations of the invention can be designed to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art. Thus, the formulations can also be formulated for controlled release or for slow release.
[0282] Compositions contemplated by the present invention can include, for example, micelles or liposomes, or some other encapsulated form, or can be administered in an extended release form to provide a prolonged storage and/or delivery effect. Therefore, the formulations can be compressed into pellets or cylinders and implanted intramuscularly or subcutaneously as depot injections. Such implants can employ known inert materials such as silicones and biodegradable polymers, e.g., polylactide-polyglycolide. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides).
[0283] For nasal administration, the preparation can contain a compound of the invention which inhibits the enzymatic activity of the focal adhesion kinase, dissolved or suspended in a liquid carrier, preferably an aqueous carrier, for aerosol application. The carrier can contain additives such as solubilizing agents, e.g., propylene glycol, surfactants, absorption enhancers such as lecithin (phosphatidylcholine) or cyclodextrin, or preservatives such as parabens.
[0284] For parenteral application, particularly suitable are injectable solutions or suspensions, preferably aqueous solutions with the active compound dissolved in polyhydroxylated castor oil.
[0285] Dosage forms can be administered daily, or more than once a day, such as twice or thrice daily. Alternatively dosage forms can be administered less frequently than daily, such as every other day, or weekly, if found to be advisable by a prescribing physician.
[0286] An embodiment of the invention also encompasses prodrugs of a compound of the invention which on administration undergo chemical conversion by metabolic or other physiological processes before becoming active pharmacological substances. Conversion by metabolic or other physiological processes includes without limitation enzymatic (e.g, specific enzymatically catalyzed) and non-enzymatic (e.g., general or specific acid or base induced) chemical transformation of the prodrug into the active pharmacological substance. In general, such prodrugs will be functional derivatives of a compound of the invention which are readily convertible in vivo into a compound of the invention. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs, ed. H. Bundgaard, Elsevier, 1985.
[0287] In another embodiment, there are provided methods of making a composition of a compound described herein including formulating a compound of the invention with a pharmaceutically acceptable carrier or diluent. In some embodiments, the pharmaceutically acceptable carrier or diluent is suitable for oral administration. In some such embodiments, the methods can further include the step of formulating the composition into a tablet or capsule. In other embodiments, the pharmaceutically acceptable carrier or diluent is suitable for parenteral administration. In some such embodiments, the methods further include the step of lyophilizing the composition to form a lyophilized preparation.
[0288] The compounds of the invention can be used therapeutically in combination with i) one or more other SI Pi inhibitors and/or ii) one or more other types of protein kinase inhibitors and/or one or more other types of therapeutic agents which can be administered orally in the same dosage form, in a separate oral dosage form (e.g., sequentially or non- sequentially) or by injection together or separately (e.g., sequentially or non-sequentially).
[0289] Accordingly, in another embodiment the invention provides combinations, comprising:
a) a compound of the invention as described herein; and
b) one or more compounds comprising:
i) other compounds of the present invention,
ii) other medicaments adapted for treatment of a malcondition for which activation of SlPt is medically indicated, for example multiple sclerosis, transplant rejection, or adult respiratory distress syndrome.
[0290] Combinations of the invention include mixtures of compounds from (a) and (b) in a single formulation and compounds from (a) and (b) as separate formulations. Some combinations of the invention can be packaged as separate formulations in a kit. In some embodiments, two or more compounds from (b) are formulated together while a compound of the invention is formulated separately.
[0291] The dosages and formulations for the other agents to be employed, where applicable, will be as set out in the latest edition of the Physicians' Desk Reference, incorporated herein by reference. METHODS OF TREATMENT
[0292] In certain embodiments, the present invention encompasses orally bioavailable compounds that specifically agonize SIPj without binding (S1P2, S1P3 and S1P4), or having significant specificity over (S1P5), other EDG receptors. A selective SI Pi agonist can be used to treat diseases with an autoimmune, hyperactive immune-response, angiogenesis or inflammatory components, but would not be limited to such conditions. Selective SlPi agonists have advantages over current therapies by increasing the therapeutic window because of reduced toxicity due to engagement of other EDG receptors.
[0293] In certain embodiments, the present invention encompasses compounds that bind with high affinity and specificity to the SlPi receptor in an agonist manner. Upon ligation of the SlPi receptor with agonist, signaling proceeds through Ga,, inhibiting the generation of cAMP by adenylate cyclase.
[0294] In certain embodiments, the present invention provides a method for activating or agonizing (i.e., to have an agonic effect, to act as an agonist) a sphingosine-1 -phosphate receptor subtype, such as SI Pi, with a compound of the invention. The method involves contacting the receptor with a suitable concentration of an inventive compound to bring about activation of the receptor. The contacting can take place in vitro, for example in carrying out an assay to determine the SIP receptor activation activity of an inventive compound undergoing experimentation related to a submission for regulatory approval.
[0295] In certain embodiments, the method for activating an SIP receptor, such as SI Pi, can also be carried out in vivo, that is, within the living body of a mammal, such as a human patient or a test animal. The inventive compound can be supplied to the living organism via one of the routes as described above, e.g., orally, or can be provided locally within the body tissues, for example by injection of a tumor within the organism. In the presence of the inventive compound, activation of the receptor takes place, and the effect thereof can be studied.
[0296] An embodiment of the present invention provides a method of treatment of a malcondition in a patient for which activation of an SIP receptor, such as SI Pi, is medically indicated, wherein the patient is administered the inventive compound in a dosage, at a frequency, and for a duration to produce a beneficial effect on the patient. The inventive compound can be administered by any suitable means, examples of which are described above. PREPARATION OF CERTAIN EMBODIMENTS
[0297] Scheme 1
Figure imgf000082_0001
Reagents: (i) KH2P04, H202, NaC102, CH3CN; (ii) H2NNHCSNH2, POCl3 CuBr2, isoamylnitrite, CH3CN.
[0298] Scheme 2:
BR /^S R1^S R1 S
Reagents: (i) Rl-boronic acid, K2C03, Pd(PPh3)4, DME, H20; (ii) NBS, DMF. [0299] Scheme 3:
Figure imgf000082_0002
Reagents: (i) R'-I, Pd(PPh3)2Cl2, THF; (ii) Br2, AcOK, AcOH. [0300] Scheme 4:
Figure imgf000082_0003
Reagents: (i) R'-Br, K2C03, Pd(PPh3)4, DME, H20; (ii) NBS, DMF. [0301] Scheme 5:
Figure imgf000082_0004
Reagents: (i) (5)-2-methyl-CBS-oxazaborolidine, BH3-Me2S, toluene, DCM; (ii) PG- Cl, (where PG is protecting group), e.g. TBSCl, imidazole, DMF; (iii) bis(pinacolato)diboron, PdCl2(dppf).CH2Cl2, KOAc, 1,4-dioxane. [0302] The (5 -enantiomer was prepared in same manner as outlined in Scheme 5 by the use of (i?)-2-methyl-CBS-oxazaborolidine in Step i. The racemic material can be prepared in an analogous manner using NaBH4 as reducing agent in Step i.
[0303] Scheme 6:
Figure imgf000083_0001
Reagents: (i) (/?)-2-methylpropane-2-sulfinamide, NaBH4, THF, toluene; (ii) 4N HCl, 1,4-dioxane; (iii) PG = di-tert-butyldicarbonate, triethylamine, DCM; (iv) bis(pinacolato)diboron, PdCl2(dppf).CH2Cl2, KOAc, 1,4-dioxane.
[0304] The (5)-enantiomer was prepared in same manner as outlined in Scheme 6 by the use of (S)-2-methylpropane-2-sulfinamide in Step i.
[0305] Scheme 7:
Figure imgf000083_0002
Reagents: (i) K2C03, Pd(PPh3)4, DME, H20; (ii) NaOi'Pr, z'PrOH; (iii) deprotection, e.g. TBAF, THF or HCl, 1,4-dioxane.
[0306] The (5)-enantiomers were prepared in same manner as outlined in Scheme 7 by the use of (5)-tert-butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3- dihydro- lH-inden-l-yl)oxy)silane in Step i. Racemic indanol was prepared in same manner using racemic ter/-butyldimethyl((4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-2,3- dihydro-lH-inden-l-yl)oxy)silane in Step i.
[0307] Scheme 8:
Figure imgf000084_0001
Reagents: (i) K2C03, Pd(PPh3)4, DME, H20; (ii) NaO/Pr, PrOH; (iii) 4N HC1, 1,4- dioxane; (iv) (a) R'-LG or R"-LG, where LG represents a leaving group, 2C03, CH3CN; (b) R3-C02H or R4-C02H , HOBt, EDC, DMF or R3-COCl or R4-C02H , TEA, DCM; (c) R3- S02C1 or R5-S02C1, TEA, DCM (d) R4-CHO, HO Ac, NaB¾ or NaCNBH3 or Na(OAc)3BH, MeOH; (e) R3-OCOCl or R -OCOC1, DIEA, DMF; (f) HN(R7R7), CDI, TEA, DCM; (g) H2NS02NH2, D, dioxane; (h) dimethyloxirane, D, EtOH; (x) (a) If R' or R" = H, then reactions (ix)(a-d) can be performed; (b) If R' or R" contains an ester then (i) hydrolysis NaOH, EtOH or (ii) reduction NaBFU, MeOH can be performed; (c) If R' or R" contains an acid then couplings HN(R7R7), HOBt, EDC, DMF can be performed; (d) If R' or R" contains
7 7
an appropriate activated alkene then Michael additions HN(R R ), DMF can be performed. [0308] The (5 -enantiomers were prepared in same manner as outlined in Scheme 8 by the use of (5)-tert-butyl (4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3-dihydro-lH- inden-l-yl)carbamate in Step i.
[0309] Scheme 9:
Figure imgf000085_0001
Reagents: (i) (3-cyano-4-isopropoxyphenyl)boronic acid, K2C03, Pd(PPh3)4, DME, H20; (ii) (R)-, (5)-, or racemic tert-butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan- 2-yl)-2,3-dihydro-lH-inden-l-yl)oxy)silane, K2C03, Pd(PPh3)4, DME, H20; (iii) TBAF, THF.
[0310] Scheme 10:
Figure imgf000085_0002
Reagents: (i) (ii) (i?)-, (S)-, or racemic tert-butyldimethyl((4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-2,3-dihydro-lH-inden-l-yl)oxy)silane, 2C03, Pd(PPh3)4, DME, H20; (ii) (3-cyano-4-isopropoxyphenyl)boronic acid, K2C03, Pd(PPh3)4, DME, H20; (iii) TBAF, THF.
[0311] Scheme 11:
Figure imgf000085_0003
Reagents: (i) (S,S) Noyori Catalyst (Chloro{[15,25))-(-)-2-amino-l,2- diphenylethyl](4-toluene sulfonyl)amidi}(p-cymene) ruthenium (II)), 5:1 HC02H/NEt3, 30°C; (ii) DBU, DPPA, Toluene; (iii) Pt02, H2, Boc20, MeOH; (iv) pinacolborane, PdCl2(dppf)-CH2Cl2, KOAc, Dioxane, 80°C. [0312] Similar compounds containing the (S)-enantiomer of the boc-protected amine were prepared in the same manner as outlined in Scheme 11 using (R,R) Noyori catalyst (Chloro {[li?,2R))-(-)-2-amino-l,2-diphenylethyl] (4-toluene sulfonyl)amidi}(p-cymene) ruthenium (II)) in Step i.
[0313] Scheme 12:
Figure imgf000086_0001
Reagents: (i) 5-(5-bromo-l,3,4-thiadiazol-2-yl)-2-fluorobenzonitrile, Pd(PPh3)4, K2C03, 3:1 DME/H20, 90°C; (ii) NaO'Pr, IP A, 60°C; (iii) 4M HCl in dioxane; (iv) (a) R'-LG or R"-LG, where LG represents a leaving group, 2C03, CH3CN or (b) R'-C02H or R2- C02H, HOBt, EDC, DMF or R'-COCl or R2-COCl, TEA, DCM or (c) R -S02C1 or R3- S02C1, TEA, DCM or (d) R2-CHO, HO Ac, NaBH4 or NaCNBH3 or Na(OAc)3BH, MeOH or (e) H2NS02NH2, Δ, dioxane; (v) (a) If R' or R" = H, then reactions (iii) (a-c) can be performed; (b) If R' or R" contains an ester then (i) hydrolysis NaOH, EtOH or (ii) reduction NaBH4, MeOH can be performed; (c) If R' or R"contains an acid then couplings HN(R R' '), HOBt, EDC, DMF can be performed.
[0314] Similar compounds containing the (5)-enantiomer of the amine were prepared in the same manner as outlined in Scheme 12 using (S)-tert-butyl (5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate in Step i. [03151 Scheme 13:
Figure imgf000087_0001
Reagents: (i) 5-(5-bromothiazol-2-yl)-2-fluorobenzonitrile, Pd(PPh3)4, K2C03, 3:1 DME/H20, 90°C; (ii) NaOiPr, IP A, 60°C; (iii) 4M HCl in dioxane; (iv) sulfamide, DIEA, dioxane, 100°C.
[0316] Similar compounds containing the (S)-enantiomer of the amine can be prepared in the same manner as outlined in Scheme 13 using (S)-tert-butyl (5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate in Step x.
[0317] Scheme 14:
Figure imgf000087_0002
Reagents: (i) bis(pinacolato)diboron, PdCl2(dppf).CH2Cl2, OAc, 1,4-dioxane.
[0318] Scheme 15:
Figure imgf000088_0001
Reagents: (i) K3P04:3H20, Pd(PPh3)4, DME, H20; (ii) K3P04:3H20, (S)-tert-bu y\ (4- (4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)- 2,3-dihydro-lH-inden-l-yl) carbamate, Pd(PPh3)4, DMF, H20 (iii) 4N HC1, 1,4-dioxane; (iv) (a) R -LG, where LG represents a leaving group, 2C03, BU4NI, CH3CN or (b) R1-C02H, HOBt, EDC, DMF or R2-COCl, TEA, DCM; or (c) R -S02C1, TEA, DCM or (d) R!-CHO, HOAc, NaB¾ or NaCNBH3 or Na(OAc)3BH, MeOH or (e) R^COCl, DffiA, DMF or (f) H R'R", CDI, TEA, DCM; (g) H2NS02NH2, Δ, dioxane or (h) dimethyloxirane, Δ, EtOH or (i) (methylsulfonyl)ethene, DIEA (v) (a) If R1 contains an ester then (i) hydrolysis using NaOH, EtOH or (ii) reduction using NaBH4, MeOH can be performed; (b) If R1 contains an acid then couplings using HNR'R", HOBt, EDC, DMF or HNR'R", HATU, DEA, DCM can be performed; (c) If R' contains an appropriate activated alkene then Michael additions using HNR'R", DMF can be performed.
[0319] The (i?)-enantiomers can be prepared in same manner as outlined in Scheme 15 by the use of (R)-tert-buty\ (4-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)- 2,3-dihydro-lH- inden-l-yl)carbamate in Step ii. [0320] Scheme 16:
Figure imgf000089_0001
Reagents: (i) 3P04:3H20, Pd(PPh3)4, DME, H20; (ii) K3P04:3H20, 2-isopropoxy-5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzonitrile, Pd(PPh3)4, DMF, H20 (iii) 4N HC1, 1,4-dioxane; (iv) (a) R^LG, where LG represents a leaving group, 2C03, BU4NI, CH3CN or (b) R1-C02H, HOBt, EDC, DMF or R2^COCl, TEA, DCM or (c) R -S02C1, TEA, DCM or (d) R'-CHO, HOAC, NaBH4 or NaCNBH3 or Na(OAc)3BH, MeOH or (e) R1- OCOC1, DIEA, DMF or (f) HNR'R", CDI, TEA, DCM or (g) H2NS02NH2, Δ, dioxane or (h) dimethyloxirane, Δ, EtOH or (i) (methylsulfonyl)ethene, DIEA (v) (a) If R1 contains an ester then (i) hydrolysis NaOH, EtOH or (ii) reduction NaBH4, MeOH can be performed; (b) If R1 contains an acid then couplings HNR'R", HOBt, EDC, DMF or HNR'R", HATU, DEA, DCM can be performed; (c) If R' contains an appropriate activated alkene then Michael additions HNR'R", DMF can be performed.
[0321] The (R)-enantiomers can be prepared in same manner as outlined in Scheme 16 by the use of (i?)-tert-butyl (4-(4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2-yl)- 2,3-dihydro- lH-inden-l-yl)carbamate in Step i.
EXAMPLES
PREPARATION OF COMPOUNDS WITH AN IN DA E MOIETY
General Methods
[0322] 1H NMR (400 MHz) and 13C NMR (100 MHz) were obtained in solution of deuteriochloroform (CDC13), deuteriomethanol (CD3OD) or dimethyl sulfoxide - D6 (DMSO). NMR spectra were processed using Mestrec 5.3.0 and 6.0.1. 13C NMR peaks that are bracketed are two rotomers of the same carbon. Mass spectra (LCMS) were obtained using an Agilent 1100/6110 HPLC system equipped with a Thompson ODS-A, 100 A, 5 μ (50 X 4.6 mm) column using water with 0.1% formic acid as the mobile phase A, and acetonitrile with 0.1% formic acid as the mobile phase B. The gradient was 20-100% with mobile phase B over 2.5 min then held at 100% for 2.5 mins. The flow rate was 1 mL/min. Unless otherwise indicated, the LCMS data provided uses this method. For more hydrophobic compounds, the following gradient was used, denoted as Method 1: 40-95% over 0.5 min, hold at 95% for 8.5 min, then return to 40% over 2 min, with a flow rate of 1 mL/min. Final compounds were checked for purity using Method 2: 5% for 1 min, 5-95% over 9 min, then hold at 95% for 5 min, with a flow rate of 1 mL/min. Method 3: 20-100% over 2.5 min then held at 100% for 4.5 min, with the flow rate of 1 mL/min. Enantiomeric excess was determined by integration of peaks that were separated on a Chiralpak AD-H, 250 x 4.6 mm column at a flow rate of 1 mL/min and an isocratic mobile phase. Unless otherwise indicated, the chiral data provided uses this method. Alternatively, chiral separations were performed under the following conditions, denoted as Chiral Method 1: Chiralpak AY-H, 250 x 4.6 mm column at a flow rate of 1 mL/min and an isocratic mobile phase. Chiral Method 2: Chiralcel OZ-3, 150 x 4.6 mm at flow rate of 1 mL min and an isocratic mobile phase. The pyridine, dichloromethane (DCM), tetrahydrofuran (THF), and toluene used in the procedures were from Aldrich Sure-Seal bottles kept under nitrogen (N2). All reactions were stirred magnetically and temperatures are external reaction temperatures. Chromatographies were carried out using a Combiflash Rf flash purification system (Teledyne Isco) equipped with Redisep (Teledyne Isco) silica gel (Si02) columns. Preparative HPLC purifications were done on Varian ProStar/PrepStar system using water containing 0.05% trifluoroacetic acid as mobile phase A, and acetonitrile with 0.05% trifluoroacetic acid as mobile phase B. The gradient was 10-80% with mobile phase B over 12 min, hold at 80% for 2 min, and then return to 10% over 2 min with flow rate of 22 mL/min. Other methods similar to this may have been employed. Fractions were collected using a Varian Prostar fraction collector and were evaporated using a Savant SpeedVac Plus vacuum pump. Compounds with salt-able centers were presumed to be the trifluoroacetic acid (TFA) salt. Microwave heating was performed using a Biotage Initiator microwave reactor equipped with Biotage microwave vessels. The following abbreviations are used: ethyl acetate (EA), triethylamine (TEA), diethyl amine (DEA), hydroxybenzotriazole (HOBt), l-emyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC), isopropanol (IP A), dimethylformamide (DMF), dimethyl acetamide (DMA). Norit is activated charcoal.
Experimental Procedures
[0323] 3-cyano-4-fluorobenzoic acid
Figure imgf000091_0001
[0324] To a solution of 3-cyano-4-fluorobenzaldehyde (45 g, 301 mmol) in CH3CN (450 mL) was added potassium phosphate monobasic (24 g, 176 mmol) in water (225 mL) and 30% hydrogen peroxide in water (30 mL). The reaction mixture was cooled to 0 °C and sodium chlorite (60 g, 663 mmol) in water (450 mL) was added dropwise over 2 h. The resulting yellow suspension was stirred at room temperature until production of oxygen ceased (4 h). Sodium sulfite (30 g, 238 mmol) in water (100 mL) was added and the reaction mixture stirred for 1 h. The reaction was quenched with 2N HC1 (500 mL) and the resulting solid was filtered and washed with water. The aqueous phase was extracted with EA (2 x 500 mL). The combined organic layers were washed with brine (200 mL), dried over MgS04, concentrated, and combined with the collected solid to produce a total of 48.5 g (97%) of crude 3-cyano-4-fluorobenzoic acid as a white solid. LCMS-ESI (m/z) calculated for C8H4FN02: 165.0; found 166.1 [M+H]+, tR = 2.54 min. 1H NMR (400 MHz, DMSO) δ 13.60 (s, 1H), 8.41 (dd, J = 6.3, 2.1 Hz, 1H), 8.30 (ddd, J = 8.8, 5.3, 2.2 Hz, 1H), 7.66 (t, J = 9.0 Hz, 1H).
[0325] 5 -(5 -amino- 1, 3, 4-thiadiazol-2-yl)-2-fluorobenzonitrile (TDZ INT- 1 )
Figure imgf000091_0002
[0326] To a stirred mixture of 3-cyano-4-fluorobenzoic acid (37.3 g, 225 mmol) and thiosemicarbazide (22.6 g, 248 mmol) was added POCl3 (148 mL) at 0°C. The reaction mixture was stirred at 0°C for 1 h and then heated to 85 °C for 6 h. The resulting yellow solution was cooled to room temperature and concentrated to 50% volume. The residue was cooled to 0 °C and water was added (300 mL) drop wise. {Caution: exothermic and violent reaction with gas evolution). The mixture was heated to 90°C for lh then cooled to room temperature. EA was added EA (300 mL) and the reaction mixture stirred for 10 min and before filtration. The collected solid was dispersed into water (270 mL), cooled to 0°C, and neutralized with 50% NaOH aqueous solution to pH8. The resulting solid was filtered, washed thoroughly with water, and dried under high vacuum to afford 26 g (52%) of 5-(5- ammo-l,3,4-t adiazol-2-yl)-2-fluorobenzonitrile TDZ INT-1 as pale yellow solid which was used in next experiment without purification. LCMS-ESI (m/z) calculated for C9H FN4S: 220.0; found 221.1 [M+H]+, tR = 2.44 min. 1H NMR (400 MHz, DMSO) δ 8.29 (dd, J= 6.1, 2.3 Hz, 1H), 8.19 (ddd, J = 8.9, 5.2, 2.4 Hz, 1H), 7.64 (t, J = 9.0 Hz, 1H), 7.58 (s, 2H). 13C NMR (101 MHz, DMSO) 6 169.82, 164.25, 161.68, 133.68, 131.65, 128.96, 117.96, 113.77, 101.59.
[0327] 5-(3,4-Diemoxyphmyl)-l,3,4-thiadiazol-2-amine TDZ INT-2 was synthesized in a similar manner as 5-(5-ammo-l,3,4-thiadiazol-2-yl)-2-fluorobenzonitrile TDZ INT-1 using 3,4-diethoxybenzoic acid. LCMS-ESI (m/z) calculated for C12H15N302S: 265.3; found 266.1. [M+H]+, ¾ = 2.58 min. !H NMR (400 MHz, DMSO) δ 7.45 - 7.31 (m, 1H), 7.23 (dd, J = 8.3, 2.1 Hz, 1H), 7.06 (d, J= 8.4 Hz, 1H), 4.31 - 3.94 (m, 4H), 3.4 (s, 2H), 1.42 (qd, J= 6.8, 3.3 Hz, 6H).
[0328] 5- -bromo-l,3,4-thiadiazol-2-yl)-2-fluorobenzonitrile (TDZ INT-3)
Figure imgf000092_0001
[0329] To a stirred solution of 5-(5-amino-l,3,4-thiadiazol-2-yl)-2-fluorobenzonitrile TDZ INT-1 (25 g, 113 mmol) and copper bromide (30.4 g, 136 mmol) in CH3CN (400 mL) was added isoamylnitrite (15.9 g, 136 mmol) and the mixture stirred at room temperature for 5 h. The reaction was partitioned between EA (2 x 250 mL) and IN HC1 (250 mL). The combined organic extracts were washed with brine, dried over MgS04, and concentrated. The crude product was crystallized from EA to afford 23.5 g (73%) of 5-(5-bromo-l,3,4- thiadiazol-2-yl)-2-fluorobenzonitrile TDZ INT-3 as a pale yellow solid. LCMS-ESI (m/z) calculated for: C9H3BrFN3S: 284.1; found 285.9 [M+H]+, tR = 3.27 min. 1H NMR (400 MHz, DMSO) δ 8.58 (dd, J= 6.0, 2.3 Hz, 1H), 8.40 (ddd, J= 8.9, 5.1, 2.4 Hz, 1H), 7.76 (t, J= 9.0 Hz, 1H); 13C NMR (101 MHz, DMSO) δ 168.61, 162.47, 140.32, 134.88, 133.38, 126.13, 117.88, 112.91.
[0330] 2-Bromo-5-(3,4-diethoxyphenyl)-l,3,4-thiadiazole TDZ INT-4 was synthesized in similar manner as described for the synthesis of 2-bromo-5-(3,4-diethoxyphenyl)- 1,3,4- thiadiazole TDZ INT-3 using 5-(3,4-diemoxyphenyl)-l,3,4-tWadiazol-2-amine. LCMS-ESI (m/z) calculated for: Ci2H13BrN202S: 328.0; found 329.1 [M+H]+, tR = 2.58 min. Ή NMR (400 MHz, CDC13) δ 7.47 (d, J = 2.1 Hz, 1H), 7.25 (d, J = 2.1 Hz, 1H), 6.84 (d, J - 8.4 Hz, 1H), 4.10 (dq, J= 8.9, 7.0 Hz, 4H), 1.42 (t, J= 7.0 Hz, 6H).
[0331] 2-fluoro-5- thiazol-2-yl)benzonitrile (THZ INT- 1 )
Figure imgf000093_0001
[0332] A solution of 2-bromothiazole (25 g, 153.4 mmol), (3-cyano-4- fluorophenyl)boronic acid (25.3 g, 153.3 mmol), 2C03 (63.6 g, 460 mmol) and 3:1 DME/H2O (205 mL) was purged with N2 for 1 h before the addition of Pd(PPh3)4 (9.2 g, 7.9 mmol). The mixture was further degassed with N2 for 5 min and then heated to 85°C for 7 h under N2. Upon cooling, the reaction mixture was diluted with EA (250 mL), washed with water (200 mL) and brine (200 mL), and dried over MgS04. The reaction mixture was filtered and concentrated to give beige solid. The crude product was purified by recrystallization from 20% EA / hexanes to afford 22 g (71 %) of 2-fluoro-5-(thiazol-2- yl)benzonitrile THZ ΓΝΤ-1 as a pale yellow solid. LCMS-ESI (m/z) calculated for C10H5FN2S: 204.2; found 205.0 [M+H]+, tR = 3.26 min. 1H NMR (400 MHz, CDC13) δ 8.21 - 8.16 (m, 1H), 8.15 - 8.08 (m, 1H), 7.86 - 7.81 (m, 1H), 7.36 - 7.32 (m, 1H), 7.27 - 7.21 (m, 1H). 13C NMR (101 MHz, CDCI3) δ 164.31, 162.22, 143.73, 132.68, 131.28, 128.34, 119.94, 116.98, 113.10.
[0333] 5-( -bromothiazol-2-yl)-2-fluorobenzonitrile (THZ INT-2)
Figure imgf000093_0002
[0334] To 2-fluoro-5-(thiazol-2-yl)benzonitrile (21.8 g, 106.7 mmol) in anhydrous DMF (200 mL) was added recrystallized N-bromosuccinimide (22.7 g, 128 mmol). The reaction mixture was stirred at room temperature for 23 h under N2. The reaction mixture was basified with IN NaOH and washed with EA and brine. The combined organic layers were dried over MgS04> filtered, and concentrated to yield orange oil. The crude product was purified by silica gel flash chromatography (20% EA/Hexanes) to produce 21 mg (70%) of 5-(5- bromothiazol-2-yl)-2-fluorobenzonitrile THZ ΓΝΤ-2 as half-white solid. LCMS-ESI (m/z) calculated for C10H4BrFN2S: 283.1; found 284.9 [M+H]+, tR = 3.82 min. 1H NMR (400 MHz, CDC13) δ 8.15 (dd, J = 5.9, 2.3 Hz, 1H), 8.08 (ddd, J= 8.8, 4.9, 2.3 Hz, 1H), 7.78 (d, J= 4.8 Hz, 1H), 7.31 (t, J- 8.6 Hz, 1H).
[0335] (S)-4-bro -2,3-dihydro-lH-inden-l-ol (IND INT-1)
Figure imgf000094_0001
[0336] To a 100 mL 3 -neck flask equipped with an internal thermometer and an addition funnel was added (i?)-(+)-2-methyl-CBS-oxazaborolidine (1.6 mL, 1M solution in toluene) and borane-dimethylsulfide (150 uL) under N2. The reaction was stirred at room temperature for 10 min then diluted with DCM (10 mL). Borane-dimethylsulfide (6.0 mL) was added and the reaction cooled to -20 °C. A solution of 4-Bromo-2,3-dihydro-lH-inden-l-one (2.5 g, 11.8 mmol) in DCM (10 mL) was added dropwise over 20 min while maintaining the reaction temperature at -20 ± 5°C. The reaction was stirred for 2 h after the addition was complete, then quenched by the dropwise addition of MeOH (10 mL). The reaction mixture was diluted with MeOH (20 mL) and the solvent distilled at atmospheric pressure. MeOH (30 mL) was added in two portions and the distillation was repeated twice. All the solvent was evaporated to give a solid which was purified by silica gel column chromatography (EA / hexanes) and recrystallization from 5:1 hexane/EA (30 mL) to provide 1.56 g (62%) of (S)-4- bromo-2,3-dihydro-lH-inden-l-ol as a white powder IND INT-1. LCMS-ESI (m/z) calculated for C9H9BrO: 213.1; found 196.9 [M-OH]+, tR = 3.06 min. 1H NMR (400 MHz, CDC13) δ 7.40 (d, J= 7.9 Hz, 1H), 7.33 (d, J= 7.5 Hz, 1H), 7.10 (t, J= 7.7 Hz, 1H), 5.29 (dd, J= 12.6, 6.9 Hz, 1H), 3.05 (ddd, J= 16.6, 8.7, 4.6 Hz, 1H), 2.87 - 2.71 (m, 1H), 2.50 (dddd, J= 13.2, 8.4, 7.0, 4.6 Hz, 1H), 1.94 (dddd, J= 13.5, 8.8, 6.6, 5.5 Hz, 1H), 1.80 (d, J= 7.1 Hz, 1H). 13C NMR (101 MHz, CDC13) δ 146.82, 143.50, 131.24, 128.58, 123.21, 120.25, 76.83, 34.69, 31.19. Chiral HPLC: (5)-4-bromo-2,3-dihydro-lH-inden-l-ol was eluted using 10% IPA in hexanes: >99.9% % ee, tR = 6.27 min.
[0337] (i?)-4-bromo-2,3-dihydro-lH-inden-l-ol IND INT-2 was prepared in an analogous manner using (5)-(-)-2-methyl-CBS-oxazaborolidine: 97.6 % ee, ¾ for ( ?)-enantiomer = 5.83 min.
[0338] (S)-((4-bromo-2,3-dihydro-lH-inden-l -yl)oxy)(tert-butyl)dimethylsilane (IND ΓΝΤ-3) BS
Figure imgf000095_0001
[0339] To a solution of (S)-4-bromo-2,3-dihydro-l H-inden- l-ol IND INT-1 (1.56 g, 7.3 mm) in DMF (5 mL) was added TBDMSCl (1.3 g, 8.7 mmol) and imidazole (1.24 g, 18.3 mmol) and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with saturated NaHC03 solution (30 mL) and extracted with EA (2 X 50 mL). The organic layers were washed with water and brine, and dried over MgS04. The crude product was purified by chromatography (EA / hexane) to afford 2.1 g (88 %) of (5)- ((4-bromo-2,3-dihydro-lH-inden-l-yl)oxy)(tert-butyl)dimethylsilane IND INT-3 as white solid. LCMS-ESI (m/z) calculated for CtsH^BrOSi: 327.3; no M+ observed, ¾ = 5.73 min (Method 2). Ή NMR (400 MHz, CDC13) δ 7.35 (d, J= 7.8 Hz, 1H), 7.22 (d, J= 7.4 Hz, 1H), 7.07 (t, J = 7.7 Hz, 1H), 5.28 (t, J = 7.1 Hz, 1H), 3.00 (ddd, J = 16.4, 9.1, 2.9 Hz, 1H), 2.73 (dt, J= 16.5, 8.3 Hz, 1H), 2.42 (dddd, J = 12.8, 8.0, 7.1, 3.0 Hz, 1H), 1.91 (dtd, J= 12.8, 8.9, 7.1 Hz, 1H), 0.98 - 0.88 (m, 9H), 0.14 (d, J = 7.4 Hz, 6H).
[0340] (i?)-((4-bromo-2,3 -dihydro- 1 H-inden- 1 -yl)oxy)(tert-butyl)dimethylsilane IND INT-4 was prepared in an analogous fashion using (i?)-4-bromo-2,3 -dihydro- 1 H-inden- l-ol.
[0341] (±)-4-bromo-2, 3-dihydro-lH-inden-l-ol (IND INT-5)
Figure imgf000095_0002
[0342] To a stirring solution of 4-bromoindanone (3 g, 14.2 mmol) in anhydrous EtOH (30mL) was added sodium borohydride (0.36 g, 9.5 mmol) and silica gel (2 g) at 0°C. The reaction was stirred at 0°C for 20 min and was allowed to stir at room temperature for 2 h. The reaction mixture was quenched with saturated NaHC03 solution (10 mL) and concentrated to remove EtOH. The aqueous layer was extracted with EA (3x20 mL) and the organic phase was dried over MgS04. After concentration, the crude product was purified by chromatography (EA / hexane) to yield (±)-4-bromo-2,3-dihydro-lH-inden-l-ol IND INT-5 (2.56 g, 85%) as a white solid. LCMS-ESI (m/z) calculated for C9H9BrO: 213.07; found
195.0 [M-H20]+, tR = 3.07 min. 1H NMR (400 MHz, CDC13) δ 7.35 (d, J= 7.9, 1H), 7.27 (d, J= 7.4, 1H), 7.05 (t, J - 7.7, 1H), 5.23 (t, J= 6.2, 1H), 3.00 (ddd, J= 16.6, 8.8, 4.6, 1H), 2.84 - 2.66 (m, 1H), 2.45 (dddd, J= 13.2, 8.4, 7.0, 4.6, 1H), 1.96 - 1.70 (m, 2H). [0343] (S)-tert-butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3- dihydro-lH-inden-l-yl)oxy)silane (IND INT-6)
Figure imgf000096_0001
[0344] A solution of (S)-((4-bromo-2,3-dihydro-lH-inden-l-yl)oxy)(tert- butyl)dimethylsilane IND INT-3 (0.2 mg, 0.61 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(l,3,2-dioxaborolane) (0.17 g, 0.67 mmol), and potassium acetate (1.8 g, 0.45 mmol) in anhydrous 1,4-dioxane (4 mL) was degassed by passing N2 through the solution for 10 min. PdCl2(dppf).CH2Cl2 (99 mg, 0.12 mmol) was added and the reaction mixture heated at 85 °C overnight. The solvent was removed under vacuum. The residue was dissolved in EA (10 mL), and filtered through celite. The filtrate was washed with water and brine, dried over MgSQ* and filtered. The crude product was purified by chromatography (EA / hexanes) to afford 26 mg (45%) (5)-tert-butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)- 2,3-dihydro-lH-inden-l-yl)oxy)silane IND INT-6 as a white solid. LCMS-ESI (m/z) calculated for C21H35B03Si: 374.4; found 245.0 [M-OTBS]+, tR = 6.57 min (Method 1). 1H NMR (400 MHz, CDC13) δ 7.66 (d, J= 7.2 Hz, 1H), 7.36 (dd, J = 8.7, 4.3 Hz, 1H), , 7.19 (dd, J= 9.4, 5.4 Hz, 1H), 5.21 (t, J= 7.0 Hz, 1H), 3.26 (ddd, J= 16.9, 8.9, 3.0 Hz, 1H), 2.86 (dt, J = 16.8, 8.3 Hz, 1H), 2.48 - 2.23 (m, 1H), 1.86 (dtd, J = 12.6, 8.8, 7.0 Hz, 1H), 1.38 - 1.23 (m, 12H), 1.00 - 0.81 (m, 9H), 0.22 - 0.07 (m, 6H). i3C NMR (101 MHz, CDC13) δ 149.59, 145.08, 134.83, 134.75, 126.92, 125.78, 83.39, 76.52, 36.29, 30.78, 25.96, 24.96, 18.28, - 4.29, -4.55.
[0345] (i?)-te^butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3- dihydro-lH-inden-l-yl)oxy)silane IND INT-7 was prepared in an analogous fashion using (#)-((4-bromo-2,3 -dihydro- 1 H-inden- 1 -yl)oxy)(tert-butyl)dimethylsilane IND INT-4. Racemic (±)-te^butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3- dihydro-1 H-inden- l-yl)oxy)silane IND INT-8 was prepared in an analogous fashion from IND INT-5.
[0346] General Procedure 1: Coupling of Heterocyclic Bromide to Indanol Boronate
[0347] A 20 mL microwave vial was charged sequentially with heterocyclic bromide (1 eq), (R)- (S)- or racemic indanol dioxaborolane (IND INT-6, 7 or 8, 1 eq), DME:H20 (3:1, 0.05 M) and potassium carbonate (3 eq). The mixture was degassed by bubbling N2 gas through the stirring solution for 10 min. Pd(PPh3)4 (0.07 eq) was added and the mixture degassed for additional 2 min. The vial as was capped and subjected to microwave irradiation at 100°C until reaction completed (40-60 min). Additional bromide was added if needed. The vial was cooled to room temperature, diluted with EA (10 x volume), washed with water and brine, dried over MgS04, and concentrated. The crude product was purified by silica gel column chromatography (EA / hexanes).
[0348] (S)-5-(5-(l-((tert-butyldimethylsilyl)oxy)-2 -dihydro-lH-inden^
thiadiazol- -yl)-2-fluorobenzonitnle
Figure imgf000097_0001
[0349] Prepared using General Procedure 1: A 20 mL microwave vial was charged with 5-(5-bromo-l,3,4-thiadiazol-2-yl)-2-fluorobenzonitrile TDZ INT-3 (30 mg, 0.1 mmol), (S)- tert-butyldimethyl((4-(4,4,5,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)-2,3 -dihydro- 1 H- inden- 1 - yl)oxy)silane IND ΓΝΤ-6 (43.6 mg, 0.11 mmol), potassium carbonate (44 mg, 0.32 mmol) and a 3:1 mixture of DME/H20 (2 mL). The reaction mixture was degassed by bubbling N2 gas through the stirring solution for 10 min. Pd(PPh3)4 was added and mixture degassed for additional 2 min. The vial was subjected to microwave irradiation at 100°C for 40 min. The reaction mixture was cooled to room temperature, diluted with EA (10 mL), and washed with water and brine. The organic layer dried over MgS04, concentrated, and purified by silica gel chromatography (EA / hexanes) to provide 25 mg (44%) of (5)-5-(5-(l-((tert- butyldimethylsilyl)oxy)-2,3-dihydro-lH-inden-4-yl)-l,3,4-thiadiazol-2-yl)-2- fluorobenzonitrile as a light yellow solid. LCMS-ESI (m/z) calculated for C24H26FN3OSSi: 451.15; found 452.1 [M+H]+, tR = 4.53 min (Method 1). !H NMR (400 MHz, CDC13) δ 8.34
- 8.25 (m, 2H), 7.85 (d, J= 7.6 Hz, 1H), 7.49 (d, J= 7.5 Hz, 1H), 7.44 - 7.34 (m, 2H), 5.34 (t, J= 7.1 Hz, 1H), 3.46 (ddd, J= 16.8, 9.0, 2.8 Hz, 1H), 3.13 (dt, J= 16.8, 8.3 Hz, 1H), 2.61
- 2.50 (m, 1H), 2.08 - 1.96 (m, 1H), 0.98 - 0.95 (m, 9H), 0.22 - 0.17 (m, 6H).
[0350] (^)-5-(5-(l-((tert-butyldimemylsilyl)oxy)-2,3-dihydro-lH-inden-4-yl)-l,3,4- thiadiazol-2-yl)-2-fiuorobenzonitrile was prepared in an analogous fashion using (R)-tert- butyldimethyl((4-(4,4,5,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)-2,3 -dihydro- 1 H-inden- 1 - yl)oxy)silane IND INT-7.
[0351] (S)-5-(5-(l-((tert-butyldimethylsilyl)oxy)-2 -dihydro-lH-m^
2-fluorobenzonitrile
Figure imgf000098_0001
[0352] Prepared using General Procedure 1. A solution of 5-(5-bromothiazol-2-yl)-2- fluorobenzonitrile THZ INT-2 (0.12 g, 0.42 mmol), (.S)-tert-butyldimethyl((4-(4,4,5,5- tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)-2,3-dihydro- 1 H-inden- 1 -yl)oxy)silane ESfD INT-6 (0.16 g, 0.42 mmol), potassium carbonate (0.176 g, 1.2 mmol) and 3:1 mixture of DME/H20 (2 mL) was degassed with N2 for 10 min before the addition of Pd(PPh3)4 (0.034 g, 0.03 mmol). The mixture reaction was degassed with N2 for additional 2 min and then heated under microwave at 90°C for 1.5 h. Upon cooling, the reaction mixture was diluted with EA (20 mL) and washed with brine (20 mL). The combined organic layers were dried over MgS04> filtered, and concentrated. The crude product was purified by silica gel flash chromatography (30% EA / hexanes) to produce 0.116 g (60 %) of (S)-5-(5-(l-((tert- butyldimethylsilyl)oxy)-2,3-dihydro-l H-inden-4-yl)thiazol-2-yl)-2-fluorobenzonitrile as a white solid. LCMS-ESI (m/z) calculated for C25H27FN2OSSi: 450.6; found 451.1 [M+H]+, tR = 4.86 min (Method 1). 1H NMR (400 MHz, CDCL3) δ 8.30-8.14 (m, 2H), 7.95 (s, 1H), 7.45 (dd, J= 7.0, 0.9, 1H), 7.32 (ddd, J= 23.9, 14.6, 11.0, 3H), 5.32 (t, J= 7.0, 1H), 3.19 (ddd, J = 15.9, 8.8, 2.7, 1H), 2.95 (dt, J = 16.1, 8.1, 1H), 2.59-2.40 (m, 1H), 2.08-1.89 (m, 1H), 0.94 (s, 9H), 0.17 (dd, J = 13.7, 7.8, 6H).
[0353] (i?)-5-(5-(l-((te^butyldimethylsilyl)oxy)-2,3-dihydro-lH-mden-4-yl)thiazol-2-yl) -2-fluorobenzonitrile was prepared in an analogous fashion using ( ?)-tert-butyldimethyl((4- (4,4,5,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)-2,3-dihydro- 1 H-inden- 1 -yl)oxy)silane.
[0354] General Procedure 2: Displacement of Fluorine with Isopropoxide
[0355] To a stirred solution of the (R)- or (^-fluorobenzene derivative (1 eq) in IP A (0.02 M) was added sodium isopropoxide (1.3 eq). The reaction was stirred at 60°C under N2 for 2 h or until reaction is complete. Upon cooling the solvent was evaporated to dryness and the product was purified by silica gel column chromatography (EA / hexanes).
[0356] (S)-5-(5-(l-((tert-butyldimethylsilyl)oxy)-2 -dihydro-lH nden^
thiadiazol-2-yl)-2-isopropoxybenzonitrile
Figure imgf000098_0002
[0357] Prepared using General Procedure 2: To a solution of (S)-5-(5-(l-((tert- butyldimethylsilyl)oxy)-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2- fluorobenzonitrile (21 mg , 0.04 mmol) in IPA (2 mL) was added sodium isopropoxide (5 mg, 0.06 mmol). The reaction mixture was heated at 60°C for 2 h. Upon cooling, the solvent was evaporated and the product was purified by a silica gel column chromatography (EA / hexanes) to afford (5)-5-(5-(l-((tert-butyldimethylsilyl)oxy)-2,3-dihydro-lH-inden-4-yl)- 1,3,4-thiadiazol -2-yl) -2-isopropoxybenzonitrile (15 mg, 68%). LCMS-ESI (m/z) calculated for C28H34N202SSi: 491.7, found 492.2 [M+H]+, tR = 5.17 min (Method 1).
[0358] (R)-5-(5-( 1 -((tert-butyldimethylsilyl)oxy)-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3 ,4- tliiadiazol-2-yl)-2-isopropoxybenzonitrile was prepared in an analogous fashion using (R)-5- (5-( 1 -((tert-butyldimethylsilyl)oxy)-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3,4-thiadiazol-2-yl)-2- fluorobenzonitrile.
[0359] (S)-5-(5-(l-(tert-butyldimethylsilyloxy)-2 -dihydro-lH nd^
isopropoxybenzonitrile
Figure imgf000099_0001
[0360] Prepared using General Procedure 2. To a solution of (5)-5-(5-(l-((tert- butyldimemylsilyl)oxy)-2,3-dihydro-lH-inden-4-yl)tWazol-2-yl)-2-fluorobenzonitrile (116 mg, 0.25 mmol) in IPA (2 mL) was added sodium isopropoxide (21.1 mg, 0.25 mmol). The reaction mixture was heated at 60°C for 2 h. Upon cooling, the solvent was evaporated and the product was purified by a silica gel column chromatography (EA / hexanes) to afford 151 mg (88 %) of (5)-5-(5-(l-(te^butyldime lsilyloxy)-2,3-dihydro-lH-inden-4-yl)thiazol-2- yl)-2- isopropoxy-benzonitrile LCMS-ESI (m/z) calculated for C28H34N202SSi: 490.7, found 491.1 [M+H]+, tR = 6.81 min (Method 1).
[0361] (i?)-5-(5-(l-(te^butyldimethylsilyloxy)-2,3-dihydro-lH-inden-4-yl)thiazol-2-yl)- 2-isopropoxybenzonitrile was prepared in an analogous fashion using (R)-5-(5-(l-((tert- butyldimethylsilyl)oxy)-2,3-dihydro-lH-inden-4-yl)thiazol-2-yl)-2-fluorobenzonitrile
[0362] General Procedure 3: Deprotection of Silyl protected Indanols
[0363] To a stirred solution of the (R)- or (5)-silyl protected indanol (1 eq) in anhydrous THF (0.06 M) was added 1 M tetrabutyl ammonium fluoride (5 eq) in THF and the mixture was stirred at room temperature under N2. Upon completion, the reaction mixture was diluted with EA (lOx volume), and washed thoroughly with NaHC03, water and brine, dried over MgS04, and concentrated. The crude product was purified by silica gel column chromatography (EA / hexanes).
[0364] Compounds 1-3, and 69-70 were prepared using a sequence of General Procedures 1-3.
[0365] (S)-5-(5-(l-hydroxy-2 -dihydro-lH-inden-4-yl)-l -thiadiaz^
Figure imgf000100_0001
[0366] To a stirred solution of (5)-5-(5-(l-((tert-butyldimethylsilyl)oxy)-2,3-dihydro-lH- inden-4-yl)-l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile (21 mg, 0.06 mmol) in anhydrous THF (1 mL) was added 1M tetrabutyl ammonium fluoride (0.3 mL, 0.3 mmol) and the reaction mixture stirred at room temperature overnight. The reaction mixture was diluted with EA (10 mL), washed with saturated NaHC03 and brine, and dried over MgS04. The product was purified by chromatography (EA / hexanes) to afford 8 mg (81%) of (5)-5-(5-(l- hydroxy-2,3-dihydro-lH-mden-4-yl)-l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile 1 as a white solid. LCMS-ESI (m/z) calculated for C21H19N302S: 377.1; found 378.1 [M+H]+, /R = 3.67 min. lH NMR (400 MHz, CDC13) δ 8.28 - 8.07 (m, 2H), 7.86 (d, J = 7.5 Hz, 1H), 7.57 (d, J = 7.5 Hz, 1H), 7.40 (t, J = 7.6 Hz, 1H), 7.08 (d, J = 9.0 Hz, 1H), 5.41 - 5.18 (m, 1H), 4.74 (dd, J = 12.2, 6.0 Hz, 1H), 3.48 (ddd, J = 17.1, 8.7, 4.6 Hz, 1H), 3.30 - 3.06 (m, 1H), 2.72 - 2.40 (m, 1H), 2.04 (ddd, J = 13.6, 8.7, 6.5 Hz, 1H), 1.64 (s, 2H), 1.44 (d, J = 6.1 Hz, 5H). 13C NMR (101 MHz, CDC13) δ 167.61, 166.22, 162.23, 147.58, 142.93, 134.04, 133.83, 129.81, 128.30, 127.45, 127.09, 123.37, 116.14, 114.45, 104.34, 77.23, 76.71, 73.09, 36.24, 31.69, 22.32.
[0367] (R)-5-(5-(\ -hydroxy-2,3-dihydro-lH-inden-4-yl) -1 ,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile 2 was prepared in an analogous fashion using (R)-5-(5-(l-((tert- butyldimethylsilyl)oxy)-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile.
[0368] (S)-5-(5-(l-hydroxy-2,3-dihydro-lH-inden-4-yl)thiazol-2-yl)-2- isopropoxybenzonitrile (Compound 70)
Figure imgf000100_0002
[0369] Prepared using General Procedure 3. To a solution of crude (S)-5-(5-(l-(tert- butyldimethylsilyloxy)-2,3-dmydro- 1 H-inden
(0.11 g, 0.22 mmol) in anhydrous THF (3 mL) was added 1.0 M solution of TBAF (1.0 mL) in THF. The reaction mixture was stirred at room temperature for 2 h. The solvent was concentrated under vacuum and the reside purified by a silica gel chromatography to afford 35 mg (41%) of (5)-5-(5-(l-hydroxy-2,3-dihydro-lH-inden-4-yl)thiazol-2-yl)-2- isopropoxybenzonitrile 70 as white solid. LCMS-ESI (m/z): calcd for: C22H 0N2O2S: 376.4; found 377.1 [M+H]+, tR = 3.66 min. 1H NMR (400 MHz, CDC13) δ 8.14 - 7.89 (m, 2H), 7.82 - 7.62 (m, 1H), 7.35 (dd, J - 7.5, 2.6 Hz, 2H), 7.22 (dd, J = 15.0, 7.5 Hz, 1H), 7.02 - 6.77 (m, 1H), 5.36 - 5.08 (m, 1H), 4.65 (hept, J = 6.0 Hz, 1H), 3.10 (ddd, J = 16.1, 8.5, 4.6 Hz, 1H), 2.93 - 2.80 (m, 1H), 2.68 - 2.54 (m, 1H), 2.44 (dddd, J = 11.7, 8.3, 7.0, 4.7 Hz, 1H), 2.01 - 1.77 (m, 1H), 1.41 - 1.29 (m, 6H). 13C NMR (101 MHz, CDC13) δ 164.98, 161.11, 146.82, 141.23, 140.87, 137.98, 132.11, 131.99, 128.40, 128.03, 127.91, 126.67, 124.62, 116.13, 113.98, 103.76, 76.43, 72.55, 35.89, 30.78, 22.01. Chiral HPLC: (S)-5-(5-(l- hydroxy-2,3-dihydro-lH-inden-4-yl)tWazol-2-yl)-2-isopropoxyberizonitrile was eluted using 15% IP A in hexanes: 100% ee; ¾ = 24.19 min.
[0370] (/?)-5-(5-(l-hydroxy-2,3-dihydro-lH-inden-4-yl)thiazol-2-yl)-2-isopropoxy benzonitrile 69 was prepared in an analogous fashion using (7?)-5-(5-(l-(tert- butyldimemylsilyloxy)-2,3-dmydro-lH-inden-4-yl)thiazol-2-yl)-2- isopropoxy-benzonitrile: 97% ee, tR for (i?)-enantiomer = 47.32 min.
[0371] (S,E)-N-(4-bromo-2,3-dihydro-lH-inden-l-ylidene)-2-methylpropane-2- sulfinamide (IND INT-9)
Figure imgf000101_0001
[0372] An oven dried 2L RB flask was charged with (,S)-2-memylpropane-2-sulfmarnide (31.5 g, 260 mmol), titanium tetraethoxide (81 g, 355 mmol) and anhydrous toluene (250 mL). The reaction mixture was heated at 90°C and a solution of 4-bromo-2,3-dihydro-lH- inden-l-one (50.0 g, 236 mmol) in anhydrous toluene was added dropwise over 90 min. The reaction mixture was then stirred at 90°C for 4 h and then overnight at 70°C. The crude (5',E)-N-(4-bromo-2,3-dihydro-lH-inden-l-ylidene)-2-methylpropane-2-sulfinamide IND ΓΝΤ-9 was used in the next experiment without purification. LCMS-ESI (m/z) calculated C,3Hi8BrNOS: 315.0; found 316.0 [M+H , tR = 3.65 min.
[0373] (i?,E)-N-(4-bromo-2,3-dihydro- 1 H-inden- 1 -ylidene)-2-methylpropane-2- sulfinamide IND INT- 10 was prepared in an analogous fashion using (7?)-2-methylpropane-2- sulfinamide.
[0374] (S)-N-((S)-4-bromo-2 -dihydro-lH-inden-l-yl)-2-methylpropane-2-sulfinamide (IND INT-l l)
Figure imgf000102_0001
[0375] To a stirred suspension of crude (S,E)-N-(4-bromo-2,3-dihydro-l H-inden- 1- ylidene)-2-methylpropane-2-sulfinamide IND ΓΝΤ-9 in toluene (250 mL) under N2I was added anhydrous THF (250 mL) and the reaction mixture was cooled to -78 °C. Sodium borohydride (26.8 g, 710 mmol) was added in four portions over 30 min (internal temperature maintained below -65 °C). The reaction mixture was stirred at -78 °C for 30 min before it was warmed to room temperature over 1 h and continued to stir for additional 1 h. The reaction mixture was filtered through Celite pad to remove Ti salts. The filtrate was treated with EA (500 mL), saturated sodium potassium tartrate (200 mL), and brine (50 mL) and the mixture stirred at room temperature overnight. The mixture was filtered through a celite pad and the filtrate dried over MgS04. The crude product was obtained by concentration to dryness gave 46 g (61%) of (5)-N-((5)-4-bromo-2,3-dihydro-lH-inden-l -yl)-2-methylpropane-2- sulfinamide IND ΓΝΤ-1 1 as an off-white solid which was used in the next experiment without purification. LCMS-ESI (m/z) calculated C13H16BrNOS: 313.0; found 314.0 [M+H]+, tR = 3.84 min.
[0376] (/?)-N-((/?)-4-bromo-2,3-dmydro-lH-inden-l-yl)-2-methylpropane-2-sulfinamide IND ΓΝΤ-12 was prepared in an analogous fashion using (R,E)-N-(4-bromo-2,3-dihydro-lH- inden-l-ylidene)-2-methylpropane-2-sulfinamide IND INT- 10.
[0377] (S)-4-bromo-2,3-dihydro-lH-inden-l -amine hydrochloride (IND ΓΝΤ-13)
Figure imgf000102_0002
[0378] To a stirred suspension of crude (5)-N-((5)-4-bromo-2,3-dihydro-lH-inden-l-yl)- 2-methylpropane-2-sulfinamide END INT- 11 (46 g, 145 mol) in MeOH (100 mL) was added 4N HC1 in dioxane (109 mL) and the yellow suspension was stirred at room temperature for 3 h. The crude reaction was diluted with MeOH (100 mL) and filtered. The filtrate was concentrated and the solid obtained was dispersed into acetonitrile (600 mL) and refluxed for 90 min. The suspension was cooled to 0 °C and the solid filtered to produce 25 g of (69%) (5)-4-bromo-2,3-dihydro-lH-inden-l -amine hydrochloride IND ΓΝΤ-13 which was used in the next step without purification. LCMS-ESI (m/z) calculated for C H10BrN: 211.09; found 197.0 [M-NH2]+, ¾ = 1.76 min. 1H NMR (400 MHz, DMSO) δ 8.76 (s, 2H), 7.71 (d, J= 7.5 Hz, 1H), 7.57 (d, /= 7.9 Hz, 1H), 7.26 (t, J= 7.7 Hz, 1H), 4.80 (s, 1H), 3.06 (ddd, J = 16.9, 8.9, 5.2 Hz, 1H), 2.93 - 2.76 (m, 1H), 2.57 - 2.39 (m, 1H), 2.11 - 1.92 (m, 1H); I3C NMR (101 MHz, DMSO) δ 144.12, 141.60, 131.71, 129.02, 124.54, 119.29, 55.30, 31.52, 29.10.
[0379] (i?)-4-bromo-2,3-dihydro-lH-inden-l -amine hydrochloride IND INT- 14 was prepared in an analogous fashion using (/?)-N-((S)-4-bromo-2,3-d ydro-lH-inden-l-yl)-2- methylpropane-2-sulfinamide IND INT- 12.
[0380] (S)-tert-butyl 4-bromo-2, 3-dihydro-lH-inden-l-ylcarbamate (IND INT- 15)
Figure imgf000103_0001
[0381] To crude (5 -4-bromo-2,3-dmydro-lH-inden-l-amine hydrochloride IND ΓΝΤ-13 (16.6 g, 66 mmol) in DCM (140 mL) at 0 °C was added triethylamine (14.8 g, 146 mmol) and di-tert-butyl dicarbonate (16.0 g, 73 mmol). The reaction was stirred at room temperature overnight. The reaction was diluted with DCM (50 mL) and washed with water and brine. The organic layers were dried over MgS04 and the product purified by crystallization from 10 % EA / hexanes to afford 14 g of (70%) (S)-tert-butyl 4-bromo-2,3-dihydro-lH-inden-l- ylcarbamate IND INT- 15 as an off-white solid. LCMS-ESI (m/z) calculated for Ci4H18BrN02: 312.2; found 197.0 [M-NH2Boc]+, tR = 3.94 min. 1H NMR (400 MHz, CDC13) δ 7.38 (d, J = 7.9 Hz, 1H), 7.25 (d, J = 7.8 Hz, 1H), 7.08 (t, J = 7.7 Hz, 1H), 5.25 (dd, J = 15.9, 7.9 Hz, 1H), 4.78 (d, J= 7.6 Hz, 1H), 2.99 (ddd, J= 16.5, 9.0, 3.4 Hz, 1H), 2.81 (dt, J= 16.5, 8.2 Hz, 1H), 2.70 - 2.36 (m, 1H), 1.94 - 1.71 (m, 1H), 1.47 (d, J = 5.2 Hz, 9H). 13C NMR (101 MHz, CDC13) δ 155.99, 146.13, 143.83, 131.35, 129.02, 123.41, 120.64, 80.10, 57.21, 33.71, 31.82, 28.86; Chiral HPLC: (5)-tert-butyl 4-bromo-2,3-dihydro-lH-inden-l- ylcarbamate was eluted using 2 % IPA in hexanes: >99.9% ee, ¾ = 11.08 min. [0382] (R)-tert-buty\ 4-bromo-2,3-dihydro-lH-inden-l-ylcarbamate IND INT- 16 was prepared in an analogous fashion from (/?)-4-bromo-2,3-dihydro-lH-inden-l -amine hydrochloride IND INT-14: >99.9% ee tR for (R)-enantiomer = 9.98 min.
[0383] (S)-tert-butyl 4-(4, 4, 5, 5-tetramethyl-l,3, 2-dioxaborolan-2-yl)-2, 3-dihydro-lH- inden-l-ylcarbamate (IND INT- 17)
Figure imgf000104_0001
[0384] A solution of (S)-tert-bu y\ 4-bromo-2,3-dihydro-lH-inden-l-ylcarbamate IND INT- 15 (13.1 g, 42 mmol), 4,4,4',4,,5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (11.7 g, 46 mmol), and potassium acetate (12.3 mg, 125 mmol) in anhydrous 1,4-dioxane (100 mL) was degassed by passing N2 through the solution for 30 min before the addition of PdCl2(dppf).CH2Cl2 (6.8 g, 8.3 mmol). The reaction mixture was heated at 85 °C for 8 h. The solvent was removed under vacuum and the residue was dissolved in EA (500 mL) and filtered through celite. The filtrate was washed with water and brine, dried over MgS04, and purified by chromatography (EA / hexanes) to afford 13 g (87%) of (Sj-tert-butyl 4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3-dihydro-lH-inden-l-ylcarbamate IND INT-17 as white solid. LCMS-ESI (m/z) calculated for C20H30BNO4: 359.2; found 382.2 [M+Na]+, tR = 4.26 min. lH NMR (400 MHz, CDC13) δ 7.66 (d, J= 7.3 Hz, 1H), 7.39 (d, J = 7.5 Hz, 1H), 7.19 (t, J= 7.4 Hz, 1H), 5.14 (dd, J= 15.8, 7.8 Hz, 1H), 4.69 (d, J= 8.7 Hz, 1H), 3.23 (ddd, J = 17.0, 8.8, 3.5 Hz, 1H), 2.94 (dt, J= 16.6, 8.2 Hz, 1H), 2.53 (ddd, J= 11.4, 8.0, 3.9 Hz, 1H), 1.73 (ddd, J = 16.4, 12.8, 8.6 Hz, 1H), 1.46 (s, 9H), 1.36 - 1.25 (m, 12H). 13C NMR (101 MHz, CDC13) δ 156.21, 150.64, 143.43, 135.37, 127.25, 126.43, 83.95, 79.78, 56.19, 34.60, 31.57, 28.88, 25.37, 25.34.
[0385] (R)-tert-buty\ 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3-dihydro-lH- inden-l-yl carbamate IND INT- 18 was prepared in an analogous fashion using
Figure imgf000104_0002
4-bromo-2,3-dihydro-lH-inden-l-ylcarbamate IND ΓΝΤ-16.
[0386] General Procedure 4: Coupling of Heterocyclic Bromides to Indane Amine
[0387] A reaction pressure flask was charged sequentially with the iheterocyclic bromide (1 eq), (R)- or (5)-Boc-protected indane amine (1 eq), DME:H20 (3:1, 0.07 M) and potassium carbonate (3 eq). The mixture was degassed by bubbling N2 gas through the stirring solution for 20 min. Then Pd(PPh3)4 (0.07 eq) was added and the mixture was degassed for additional 5 min. The reaction flask was capped tightly and the mixture was heated at 85°C for 12-24 h. The reaction was cooled to room temperature, diluted with water (2x volume), and stirred for 30 min. The resulting solid was filtered, washed with hexanes, and dried under high vacuum. The crude product was purified by silica gel column chromatography (EA / hexanes) or used in the next experiment without purification.
[0388] (S)-tert-butyl (4-(5-(3-cyano-4-fluorophenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3-dihydro- lH-inden-l-yl)carbamate
Figure imgf000105_0001
[0389] Prepared using General Procedure 4. A suspension of 5-(5-bromo-l,3,4- thiadiazol-2-yl)-2-fluorobenzonitrile TDZ ΓΝΤ-3 (1.5 g, 5.3 mmol), (S)-tert-bxxty\ (4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3-dihydro-lH-inden-l-yl)carbamate IND INT-17 (1.9 g, 5.3 mmol) and potassium carbonate (2.2 g, 16 mmol) in DME:H20 (3:1, 70 mL) was degassed with N2 for 20 min before the addition of Pd(PPh3)4 (0.43 g, 0.3 mmol). The mixture was degassed with N2 for an additional 5 min and the suspension was heated under N2 at 85°C for 12 h. Upon cooling, the reaction mixture was diluted with water (150 mL) and the mixture stirred for 30 min. The resulting solid was filtered, washed with water, and dried under high vacuum to afford 2.3 g (100%) of crude (S)-tert-butyl (4-(5-(3-cyano-4- fluorophenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH-inden-l-yl)carbamate as light brown solid which was used in the next experiment without purification. LCMS-ESI (m/z) calculated for C23H21FN402S: 436.1; found 459.1 [M+Na]+, ¾ = 4.19 min.
[0390] (R)-tert-b tyl (4-(5-(3-cyano-4-fluorophenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro- lH-inden-l-yl)carbamate was prepared in an analogous fashion using (i?)-tert-butyl (4- (4,4,5,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)-2,3-dihydro- 1 H-inden- 1 -yl)carbamate IND INT-18.
[0391] (S)-tert-butyl (4-(2-(3-cyano-4-fluorophenyl)thiazol-5-yl)-2,3-dihydro-lH-inden- l-yl)carbamate
Figure imgf000105_0002
[0392] Prepared using General Procedure 4. A solution of 5-(5-bromothiazol-2-yl)-2- fluorobenzonitrile THZ ΓΝΤ-2 (2.0 g, 7.0 mmol), (5)-tert-butyl (4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-2,3-dihydro-lH-inden-l-yl)carbamate IND INT- 17 (2.5 g, 7.0 mmol), potassium carbonate (2.9 g, 21 mmol) and a 3:1 mixture of DME/H20 (30 mL) was degassed with N2 for 10 min before the addition of Pd(PPh3)4 (0.57 g, 0.005 mmol). The mixture was degassed with N2 for an additional 2 min and the suspension was heated under nitrogen at 80°C for 12 h. Upon cooling, the reaction mixture was diluted with EA (20 mL) and washed with brine (20 mL). The combined organic layers were dried over MgS04> filtered, and concentrated. The crude product was purified by silica gel flash chromatography (30% EA / hexanes) to produce 3.0 g (83 %) of («S)-tert-butyl (4-(2-(3-cyano-4-fluorophenyl)thiazol-5- yl)-2,3-dihydro-lH-inden-l-yl)carbamate as a white solid. LCMS-ESI (m/z) calculated for C24H22FN302S: 435.5; found 436.1 [M+H]+, tR = 4.14 min. Ή NMR (400 MHz, CDC13) δ 8.20 (m, 2H), 7.93 (s, 1H), 7.44 (d, J = 7.5 Hz, 1H), 7.32 (m, 3H), 5.26 (m, 1H), 4.76 (d, J - 8.4 Hz, 1H), 3.09 (m, 2H), 2.65 (ddd, J = 12.5, 8.3, 4.6 Hz, 1H), 1.84 (dq, J = 12.9, 8.5 Hz, 1H), 1.48 (s, 9H).
[0393] (S)-tert-butyl ( 4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3- dihydro- 1 H-inden-l-y I) carbamate
Figure imgf000106_0001
[0394] Prepared using General Procedure 2. To a solution of (5 -tert-butyl (4-(5-(3- cyano-4-fluorophenyl)-l,3,4-lMadiazol-2-yl)-2,3-dihydro-lH-inden-l-yl)carbamate (2.5 g , 5.7 mmol) in EPA (30 mL) was added sodium isopropoxide (0.61 g, 7.4 mmol). The reaction mixture was heated at 60°C for 4 h. Upon cooling, the mixture was concentrated to 50% volume and the suspension was cooled to 0°C. The resulting solid was filtered and dried under high vacuum to afford 1.14 g (42%) of (5)-tert-butyl (4-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 -yl)carbamate as off-white solid. LCMS-ESI (m/z) calculated for C26H28N403S: 476.2; found 477.2 (M+H). tR = 4.12 min. Ή NMR (400 MHz, CDC13) δ 8.26 - 8.04 (m, 2H), 7.82 (d, J = 7.7 Hz, 1H), 7.49 (d, J = 7.5 Hz, 1H), 7.38 (d, J = 7.6 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 5.38 - 5.08 (m, 1H), 4.94 - 4.62 (m, 1H), 3.54 - 3.32 (m, 1H), 3.21 (s, 1H), 2.80 - 2.59 (m, 1H), 1.97 - 1.74 (m, 1H), 1.52 - 1.35 (m, 15H). 13C NMR (101 MHz, CDC13) 8 166.93, 165.54, 161.59, 155.65, 145.84, 142.05, 133.28, 128.71, 127.59, 126.64, 126.33, 122.72, 115.54, 113.86, 103.69, 79.59, 72.50, 60.35, 55.73, 33.78, 31.27, 28.39, 21.74. [0395] (i?)-tert-butyl (4-(5-(3-cyano-4-isopropoxyphenyl)-l ,3,4-thiadiazol-2-yl)-2,3- dihydro-lH-inden-l-yl) carbamate was prepared in an analogous fashion using (R)-tert-buty\ (4-(5-(3-cyano-4-fluorophenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)carbamate.
[0396] (S)-tert-butyl (4-(5-(3-cyano-4-isopropoxyphenyl)thiazol-2-yl)-2, 3-dihydro-lH- inden- -yl) carbamate
Figure imgf000107_0001
[0397] Prepared using General Procedure 2. To a solution of (S)-tert-butyl (4-(2-(3- cyano-4-fluorophenyl)thiazol-5-yl)-2,3-dihydro-l H-inden- l-yl)carbamate (2.5 g, 5.7 mmol) in IPA (50 mL) was added sodium isopropoxide (0.61 g, 7.4 mmol). The reaction mixture was heated at 60°C for 4 h. Upon cooling, the mixture was concentrated to 50% volume and the suspension was cooled to 0°C. The resulting solid was filtered and dried under high vacuum to afford 2.66 g (98%) of (S)-tert-buty\ (4-(5-(3-cyano-4-isopropoxyphenyl) tbiazol- 2-yl)-2,3-dihydro-l H-inden- l-yl)carbamate. LCMS-ESI (m/z) calculated for C27H29N303S: 475.1; found 476.2 (M+H). tR = 4.30 min. 1H NMR (400 MHz, CDC13) δ 8.19 - 8.05 (m, 2H), 7.89 (s, 1H), 7.43 (s, 1H), 7.39 - 7.28 (m, 2H), 7.04 (d, J= 8.9 Hz, 1H), 5.39 - 5.15 (m, 1H), 4.73 (s, 2H), 3.20 - 2.94 (m, 2H), 2.69 - 2.57 (m, 1H), 1.94 - 1.77 (m, 1H), 1.49 (s, 9H), 1.44 (d, J = 6.1 Hz, 6H).
[0398] General Procedure 5: Preparation of Heterocyclic Indane Amines
[0399] To a stirred suspension of the (R)- or (5)-Boc protected indane amine (1 eq) in 1,4-dioxane (0.2 M) was added 4N HC1 in 1,4-dioxane (10 eq) and the mixture was heated at 55°C until completion of the reaction (3-5 h). The reaction was cooled to room temperature and diluted with diethyl ether. The resulting solid was filtered and dried under vacuum to obtain the pure product as the hydrochloride salt.
[0400] Compounds 4 - 6 and 71 - 72 were prepared by the sequential use of General Procedures 4, 2, and 5.
[0401] (S)-5-(5-(l-amino-2,3-dihydro-lH-inden-4-yl)-l,3,4-thiadiazol-2-yl)-2- is
Figure imgf000107_0002
[0402] Prepared using General Procedure 5. To a stirred solution of («S)-tert-butyl (4-(5- (3 -cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 -yl)carbamate (1.1 g, 2.3 mmol) in 1,4-dioxane (10 mL) was added 4N HCl solution of in 1,4-dioxane (10 mL). The reaction mixture was stirred at 55°C for 2.5 h. Upon cooling to 0°C, the reaction mixture was diluted with diethyl ether (100 mL) and the resulting solid was filtered and dried to afford 980 mg (96%) of (5)-5-(5-(l-amino-2,3-dihydro-lH-inden-4-yl)-l,3,4-thiadiazol-2- yl)-2-isopropoxybenzonitrile hydrochloride 4 as off-white solid. LCMS-ESI (m/z) calculated for C21H2oN4OS, 376.1; found 377.1 (M+H). fR = 2.35 min. Ή NMR (400 MHz, DMSO) 5 8.64 - 8.51 (m, 3H), 8.41 (d, J= 2.3 Hz, 1H), 8.32 (dd, J= 8.9, 2.4 Hz, 1H), 7.99 (d, J= 7.4 Hz, 1H), 7.84 (d, J= 7.6 Hz, 1H), 7.59 - 7.49 (m, 2H), 4.95 (dt, J = 12.2, 6.1 Hz, 1H), 4.84 (s, 1H), 3.54 - 3.32 (m, 1H), 3.30 - 3.15 (m, 1H), 2.65 - 2.53 (m, 1H), 2.12 (ddd, J = 13.9, 5.6, 3.0 Hz, 1H), 1.37 (dd, J = 10.4, 6.1 Hz, 6H). 13C NMR (101 MHz, CDC13) 6 166.61, 166.11, 161.5, 143.05, 141.73, 134.16, 133.45, 129.74, 128.26, 127.84, 126.47, 122.33, 115.79, 115.2, 102.53, 72.43, 54.75, 31.48, 30.12, 21.74. Chiral HPLC: (S 5-(5-(l-amino- 2,3-dihydro-lH-inden-4-yl)-l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 4 was eluted using 30% EtOH in hexanes plus 0.1% DEA: 99.0% ee, tR = 34.2 min.
[0403] (i?)-5-(5-(l-amino-2,3-dihydro-lH-inden-4-yl) -l,3,4-thiadiazol-2-yl) -2 isopropoxybenzonitrile hydrochloride 5 was prepared in an analogous fashion using (R)-tert- butyl (4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH-inden-l- yl)carbamate: >99.9% ee, tR = 28.8 min.
[0404] (S)-5-(2-(l-amino-2,3-dihydro-lH-inden-4-yl)thiazol-5-yl)-2- isopropoxybenzonitrile hydrochloride (Compound 71)
Figure imgf000108_0001
[0405] Prepared using General Procedure 5. To a stirred solution of (5)-tert-butyl (4-(5- (3-cyano-4-isopropoxyphenyl)thiazol-2-yl)-2,3-dihydro-l H-inden- l-yl)carbamate (1.0 g, 2.1 mmol) in 1,4-dioxane (5 mL) was added 4N HCl solution in 1,4-dioxane (5 mL). The reaction mixture was stirred at 55°C for 2.5 h. Upon cooling to 0°C, the reaction mixture was diluted with diethyl ether (50 mL) and the resulting solid was filtered, washed with ether (20 mL), and dried to afford 0.86 g (100%) of (5)-5-(2-(l-arnino-2,3-dihydro-lH-inden-4- yl)thiazol-5-yl)-2 -isopropoxybenzonitrile hydrochloride 71. LCMS-ESI (m/z) calculated for C22H21N3OS: 375.1; found 376.2 (M+H). tR = 2.45 min. Ή NMR (400 MHz, DMSO) 5 8.64 (d, J= 3.6 Hz, 2H), 8.30 (d, J= 2.3 Hz, 1H), 8.23 (dd, J= 8.9, 2.4 Hz, 1H), 8.21 (s, 1H), 7.70 (dd, J= 7.6, 2.6 Hz, 2H), 7.45 (dd, J = 8.4, 5.4 Hz, 2H), 4.91 (dt, J= 12.2, 6.1 Hz, 1H), 4.85 - 4.58 (m, 1H), 3.36 - 3.21 (m, 1H), 3.21 - 3.04 (m, 1H), 2.63 - 2.51 (m, 1H), 2.09 (td, J = 8.3, 2.8 Hz, 1H), 1.43 - 1.28 (m, 6H). 13C NMR (101 MHz, DMSO) δ 166.61, 166.11, 161.50, 143.05, 141.73, 134.16, 133.45, 129.74, 128.26, 127.84, 126.47, 122.33, 115.79, 115.20, 102.53, 72.43, 54.75, 31.48, 30.12, 21.74. Chiral HPLC: (5)-5-(2-(l-amino-2,3- dihy<ko-lH-indm-4-yl)thiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride was eluted in 8% EtOH / hexanes: >99.9% ee, tR = 67.15 min (Chiral Method 1).
[0406] (R)-5-(2-( 1 -amino-2,3-dihydro- 1 H-inden-4-yl)thiazol-5-yl)-2- isopropoxybenzonitrile hydrochloride 72 was prepared in an analogous fashion using (R)- tert-butyl (4-(5-(3-cyano-4-isopropoxyphenyl)thiazol-2-yl)-2,3-dihydro-lH-inden-l- yl)carbamate: 99.0% ee, tRfor (i?)-enantiomer = 62.18 min.
[0407] General Procedure 6. Preparation of Indane Amides via Acid Coupling
[0408] To the appropriate acid (1 eq) in DMF (0.05 M) was added HOBt (1.3 eq), and EDC (1.3 eq). The reaction was stirred at room temperature for 0.5 h or until the acid was fully activated. The (R)- or (5)-indane amine (1 eq) was added in one portion and the reaction was stirred at room temperature for 12 h. The crude reaction mixture was subjected to preparative HPLC purification. Products that contain Boc protected amine side chains were further treated with 4N HCl in 1,4-dioxane and heated 55°C for 2 h. The reaction mixture was diluted with diethyl ether and filtered to afford the desired products as the hydrochloride salts.
[0409] Compounds 7 - 13, 49, 73, 74, 77 - 86 were prepared using General Procedure 6.
[0410] (S)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inde -l-yl)-2-hydroxyacetamide (Compound 7)
Figure imgf000109_0001
[0411] Prepared using General Procedure 6. A solution of 2-hydroxyacetic acid (4 mg, 0.05 mmol), HOBt (8.8 mg, 0.06 mmol), EDC (12.5 mg, 0.06 mmol) and DIEA (15 mg, 0.11 mmol) in DMF (1 mL) was stirred for 30 min before the addition of (5)-5-(5-(l-amino-2,3- dmydro-lH-inden-4-yl)-l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 4 in DMF (0.5 mL). The reaction mixture was stirred at room temperature overnight. The crude reaction mixture was preparative HPLC to produce 10 mg (50%) of (5)-N-(4-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 -yl)-2-hydroxyacetamide 7 as white solid. LCMS-ESI (m/z) calculated for: C23H22N403S: 434.1; found 435.1 [M+H]+, tR = 3.11 min. Ή NMR (400 MHz, CDC13) δ 8.19 (dd, J= 8.9, 2.3 Hz, 1H), 8.12 (d, J= 2.2 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.42 (d, J = 7.5 Hz, 1H), 7.34 (t, J= 7.6 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 6.85 (d, J = 8.5 Hz, 1H), 5.71 - 5.46 (m, 1H), 4.76 (dt, / = 12.2, 6.1 Hz, 1H), 4.21 (s, 2H), 3.46 (ddd, J= 17.0, 8.7, 3.6 Hz, 1H), 3.30 - 3.09 (m, 1H), 2.69 (ddd, J= 16.6, 8.3, 4.0 Hz, 1H), 2.08 - 1.80 (m, 2H), 1.46 (d, J= 6.1 Hz, 6H). 13C NMR (101 MHz, CDC13) δ 167.44, 166.23, 162.20, 145.53, 142.82, 133.89, 133.79, 129.53, 128.28, 127.20, 126.93, 123.12, 116.03, 1 14.38, 104.22, 73.06, 62.72, 54.46, 33.91, 31.98, 22.24.
[0412] (7?)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-tWadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl)-2-hydroxyacetamide 8 was prepared in an analogous fashion using (i?)-5-(5-(l- amino-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 5.
[0413] (S)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l- yl)- -hydroxyacetamide (Compound 73)
Figure imgf000110_0001
[0414] Prepared using General Procedure 6. A solution of 2-hydroxyacetic acid (2 mg, 0.02 mmol), HOBt (4.8 mg, 0.06 mmol), EDC (7.0 mg, 0.06 mmol) and DIEA (7.7 mg, 0.06 mmol) in DMF (1 mL) was stirred for 30 min before the addition of ( )-5-(2-(l-amino-2,3- dihydro-lH-inden-4-yl)tWazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 71 in DMF (0.5 mL). The reaction mixture was stirred at room temperature overnight. The crude reaction mixture was subjected to preparative HPLC to produce 5 mg (58%) of ((5)-N-(4-(2-(3-cyano- 4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l-yl)-2-hydroxyacetamide 73 as white solid. LCMS-ESI (m/z) calculated for: C24H23N303S: 433.2; found 434.1 [M+H]+, tR = 3.11 min.
[0415] General Procedure 7. Preparation of Indane Amides via Acid Chlorides
[0416] To a stirred solution of (R)- or (5)-indane amine hydrochloride (1 eq) in anhydrous DCM (0.03 M) was added triethylamine (3 eq) followed by the appropriate acid chloride (1.5 eq). The reaction mixture was stirred overnight at room temperature. The solvent was evaporated and the product was purified by preparative HPLC.
[0417] Compounds 14, 15, 75, 76, 87, and 88 were prepared using General Procedure 7. [0418] (S)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydt inden- -yl)acetamide (Compound 14)
Figure imgf000111_0001
[0419] Prepared using General Procedure 7: To a stirred solution of (5)-5-(5-(l-arnino- 2,3-dihydro-lH-inden-4-yl)-l ,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 4 (15 mg, 0.03 mmol) in anhydrous DCM (1 mL) was added triethylamine (11 mg, 0.1 mmol) followed by acetyl chloride (4.2 mg, 0.05 mmol) and the reaction mixture was stirred at room temperature overnight. The solvent was evaporated and the crude mixture purified by preparative HPLC to afford (5)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2- yl)-2,3-dihydro-lH-inden-l-yl)acetamide 14. LCMS-ESI (m/z) calculated for: C23H22N402S: 418.2; found 419.3 [M+H]+, tR = 3.34 min. 1H NMR (400 MHz, CDC13) δ 8.20 (dd, J = 8.9, 2.3 Hz, 1H), 8.13 (d, J= 2.2 Hz, 1H), 7.81 (d, J= 7.6 Hz, 1H), 7.45 (d, J= 7.5 Hz, 1H), 7.36 (t, J = 7.6 Hz, 1H), 7.08 (d, J = 9.0 Hz, 1H), 5.83 (d, J = 8.4 Hz, 1H), 5.57 (q, J = 7.9 Hz, 1H), 4.76 (dt, J = 12.2, 6.1 Hz, 1H), 3.46 (ddd, J = 17.1, 8.8, 3.8 Hz, 1H), 3.28 - 3.15 (m, 1H), 2.75 - 2.62 (m, 1H), 2.07 (s, 3H), 1.98 - 1.80 (m, 1H), 1.46 (d, J = 6.1 Hz, 6H). I3C NMR (101 MHz, CDC13) δ 170.36, 167.43, 166.14, 162.16, 145.92, 142.83, 133.89, 133.77, 129.44, 128.23, 127.18, 126.95, 123.21, 116.05, 114.36, 104.23, 73.03, 55.00, 34.03, 31.95, 23.92, 22.24.
[0420] (i?)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl)acetamide 15 was prepared in an analogous fashion using (i?)-5-(5-(l-arnino-2,3- dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 5.
[0421] (S)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l- yl)- -methoxyacetamide (Compound 76)
Figure imgf000111_0002
[0422] Prepared using General Procedure 7: To a stirred solution of (5)-5-(2-(l-amino- 2,3-dihydro-lH-inden-4-yl)thiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 71 (15 mg, 0.03 mmol) in anhydrous DCM (1 mL) was added triethylamine (11 mg, 0.1 mmol) followed by 2-methoxyacetyl chloride (11.8 mg, 0.1 mmol) and the reaction mixture was stirred at room temperature overnight. The solvent was evaporated and the crude mixture was purified by preparative HPLC to afford (5 -N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3- dihydro-lH-inden-l-yl)-2-methoxyacetamide 76. LCMS-ESI (m/z) calculated for: C25H25N303S: 447.2; found 448.1 [M+H]+, tR = 3.70 min.
[0423] General Procedure 8. Preparation of Indane Carbamates
[0424] To a stirred solution of (R)- or (5 -indane amine (1 eq) in DCM (0.03M) was added TEA (3 eq) and the appropriate carbonochloridate (1.5 eq) at room temperature. The reaction was stirred at room temperature for 4 h. The solvent was evaporated and the pure product isolated by precipitation with water or preparative HPLC.
[0425] Compounds 16, 68, 89, and 90 were prepared using General Procedure 8.
[0426] (S)-methyl (4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3- dihyd -lH-inden-l-yl)carbamate (Compound 16)
Figure imgf000112_0001
[0427] Prepared using General Procedure 8. To a stirred solution of (S)-5-(5-(l-amino- 2,3-dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 4 (15 mg, 0.03 mmol) and TEA (11 mg, 0.1 mmol) in DCM (1 mL) was added methyl chloroformate (10 mg, 0.1). The reaction mixture was stirred at room temperature for 16 h. The solvent was evaporated and water (2 mL) was added. The resulting solid was filtered, washed with water, and dried under high vacuum to afford 12 mg (92%) of (5)-methyl (4-(5- (3-cyano-4-isopropoxyphenyl)- 1 ,3,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 -yl)carbamate 16 as white solid. LCMS-ESI (m/z) calculated for C23H22N403S: 434.1; found 435.3 [M+H]+, tR = 3.69 min.
[0428] (S)-methyl (4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH- ind -l-y I) carbamate (Compound 90)
Figure imgf000112_0002
[0429] Prepared using General Procedure 8. To a stirred solution of (5)-5-(2-(l-amino- 2,3-dihydro-lH-inden-4-yl)thiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 71 (15 mg, 0.03 mmol) and TEA (11 mg, 0.1 mmol) in DCM (1 mL) was added methyl chloroformate (10 mg, 0.1). The reaction mixture was stirred at room temperature for 16 h. The solvent was evaporated and water (2 mL) was added. The resulting solid was filtered, washed with water, and dried under high vacuum to afford 6 mg (51%) of (5)-methyl (4-(2-(3-cyano-4- isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l-yl)carbamate 90 as white solid. LCMS-ESI (m/z) calculated for C24H23N303S: 433.2; found 434.1 [M+H]+, tR = 3.86 min.
[0430] General Procedure 9. Alkylation of Indane Amines
[0431] To a solution of the (R)- or (5)-indane amine in CH3CN (0.2 M) was added K2C03 (3 eq) and the appropriate alkyl halide (1.2 eq). In some cases TEA (3 eq) and DMF (0.1 M) was used. The mixture was heated at 80-95°C until the starting material was consumed or di- alkylation of the amine becomes prevalent. If necessary, additional alkyl halide is added to drive the reaction. The reaction mixture was filtered to remove inorganic solids and concentrated, re-suspended in EA and washed with water. The organic layer is dried and concentrated, then purified by chromatography (MeOH/DCM) or preparative HPLC to provide the desired product. TBS-protected alcohols were deprotected using 4N HC1.
[0432] Compounds 17 - 20 and 91 - 95 were prepared using General Procedure 9.
[0433] (R)-5-(5-(l-((2-((tert-butyldimethylsilyl)oxy)ethyl)amino)-2, 3-dihydro-lH-inden- 4-yl)- -thiadiazol-2-yl)-2-isopropoxybenzonitrile
Figure imgf000113_0001
[0434] Prepared using General Procedure 9. To a suspension of (i?)-5-(5-(l-amino-2,3- dihydro-lH-inden-4-yl)-l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 5 (50 mg, 0.12 mmol) in anhydrous DMF (5 mL) was added TEA (36.7 mg, 0.36 mmol) and (2- bromoethoxy)(tert-butyl)dimethylsilane (34.6 mg, 0.14 mmol). The solution was stirred at 95°C. After 16 h more (2-bromoethoxy)(tert-butyl)dimethylsilane (34.6 mg, 0.14 mmol) was added and heating continued for 12 h. Water (5 mL) was added and the reaction mixture was extracted with EA (2 x 5 mL). The organic layers were washed with brine, dried, and purified by column chromatography (EA / hexanes) to afford 10 mg (15 %) of (#)-5-(5-(l-((2-((tert- butyldimethylsilyl)oxy)ethyl)amino)-2,3 -dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl) -2- isopropoxybenzonitrile. LCMS-ESI (m/z) calculated for: C29H38N402SSi: 534.3; found 535.3 [M+H]+, tR = 3.08 min.
[0435] (5)-5-(5-( 1 -((2-((tert-butyldimethylsilyl)oxy)ethyl)amino)-2,3-dihydro- 1 H-inden- 4-yl) -l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile was prepared in an analogous fashion using (5)-5-(5-(l-an mo-2,3-dmydro-lH-inden-4-yl)-l,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 4. [0436] (R)-5-(5-(l-((2-hydroxyethyl)amino)-2 -dihydro-lH-inden-4-yl)-^ 2- -2-isopropoxybenzonitrile (Compound 17)
Figure imgf000114_0001
[0437] To (Λ)-5-(5-(1 -((2-((tert-butyldimethylsilyl)oxy)ethyl)amino)-2,3-dihydro-lH- inden-4-yl)-l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile (10 mg, 0.018 mmol), in 1,4- dioxane (1.5 mL) was added 4N HCl in dioxanes (0.5 mL). The mixture was stirred at room temperature for 3 h and solvent was evaporated. The crude material was purified by a preparative HPLC to afford 7 mg (90%) of (i?)-5-(5-(l-((2-hydroxyethyl)amino)-2,3-dihydro- lH-inden-4-yl)-l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile 17. LCMS-ESI (m/z) calculated for: C23H24N402S: 420.2; found 421.2 [M+H]+, tR = 2.38 min. 1H NMR (400 MHz, CDC13) δ 8.19 (dd, J= 8.9, 2.3 Hz, 1H), 8.15 (d, J = 2.2 Hz, 1H), 7.97 (d, J = 7.6 Hz, 1H), 7.88 (d, J = 7.7 Hz, 1H), 7.48 (t, J = 7.7 Hz, 1H), 7.10 (d, J = 9.0 Hz, 1H), 4.94 (d, J = 4.2 Hz, 1H), 4.76 (dt, J= 12.2, 6.1 Hz, 1H), 3.89 (d, J = 16.3 Hz, 2H), 3.68 - 3.20 (m, 2H), 3.20 - 2.89 (m, 2H), 2.72 - 2.53 (m, 2H), 2.65 - 2.53 (m, 1H), 2.49 - 2.27 (m, 1H), 1.44 (d, J = 6.1 Hz, 6H).
[0438] (5)-5-(5-(l-((2-hydroxyethyl)amino)-2,3-dihydro-lH-inden-4-yl)-l,3,4-thiadiazol- 2-yl) -2- isopropoxy-benzonitrile 18 was prepared in an analogous fashion using (5)-5-(5-(l- ((2-((te^butyldimethylsilyl)oxy)ethyl)amino)-2,3 -dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol- 2-yl) -2-isopropoxybenzonitrile.
[0439] (S)-5-(5-(l-((2-((tert-butyldimethylsilyl)oxy)ethyl)amino)-2,3-dity
4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile
Figure imgf000114_0002
[0440] Prepared using General Procedure 9. To a suspension of (5)-5-(2-(l-amino-2,3- dmydro-lH-inden-4-yl)thiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 71 (25 mg, 0.06 mmol) in anhydrous DMF (2 mL) was added TEA (7.3 mg, 0.36 mmol) and (2- bromoethoxy)(tert-butyl)dimethylsilane (6.9 mg, 0.14 mmol). The solution was stirred at 100°C for 48 hours. The reaction was diluted with EA (10 mL), washed with water and brine and dried. Concentration and purification by column chromatography (EA / hexanes) gave 29 mg (90 %) of (5)-5-(5-(l-((2-((tert-butyldimemylsilyl)oxy)ethyl)annno)-2,3-dihydro-lH- inden-4-yl)thiazol-2-yl) -2-isopropoxybenzonitrile as a dark gray solid. LCMS-ESI (m/z) calculated for: C30H39N3O2SSi: 533.3; found 534.3 [M+H]+, tR = 3.22 min.
[0441] (i?)-5-(5-(l-((2-((te^butyldime lsilyl)oxy)e l)amino)-2,3-dihydro-lH-inden-
4-yl) -thiazol-2-yl) -2-isopropoxybenzonitrile was prepared in an analogous fashion using
(i?)-5-(2-(l-amino-2,3-dihydro-lH-inden-4-yl)tWazol-5-yl)-2-isopropoxyberizonitrile hydrochloride 72.
[0442] (S)-5-(5-(l-((2-hydroxyethyl)amino)-2 -dihydro-lH-inde
isopropoxybenzonitrile (Compound 92)
Figure imgf000115_0001
[0443] To a solution of (-S)-5-(5-(l-((2-((te^butyldimethylsilyl)oxy)ethyl)amino)-2,3- dihydro-lH-inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile (10 mg, 0.018 mmol) in ether (1 mL) was added 2N HC1 in ether (0.1 mL). The mixture was stirred at room temperature for 12 h and solvent was evaporated. The crude material was purified by a preparative HPLC to afford 6 mg (80%) of (5)-5-(5-(l-((2-hydroxyethyl)amino)-2,3-dihydro-lH-inden-4- yl)thiazol-2-yl)-2-isopropoxybenzonitrile 92. LCMS-ESI (m/z) calculated for: C24H25N302S: 419.2; found 420.2 [M+H]+, tR = 2.43 min.
[0444] (R)-5-(5-( 1 -((2-hydroxyethyl)amino)-2,3-dihydro- 1 H-inden-4-yl)thiazol-2-yl) -2- isopropoxybenzonitrile 91 was prepared in an analogous fashion using (i?)-5-(5-(l-((2-((tert- butyldimemylsilyl)oxy)ethyl)amino)-2,3 -dihydro- 1 H-inden-4-yl)thiazol-2-yl) -2- isopropoxybenzonitrile.
[0445] (S)-methyl 2-((4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3- dihydro-lH-inden-l-yl)amino)acetate
Figure imgf000115_0002
[0446] Prepared using General Procedure 9. To a suspension of (S)-5-(5-(l-amino-2,3- dihydro-lH-inden-4-yl)-l, 3, 4-thiadiazol-2-yl)-2 -isopropoxybenzonitrile hydrochloride 4 (150 mg, 0.36 mmol) in CH3CN (5 mL) was added K2C03 (150.9 mg, 1.09 mmol) and methyl 2- bromoacetate (67 mg, 0.43 mmol). The suspension was stirred at 80°C. After 6 h more methyl 2-bromoacetate (6.7 mg, 0.043 mmol) was added and heating continued for 12 h. The reaction mixture was filtered and concentrated. The residue was re-suspended in EA (15 mL), washed with water and brine, dried and concentrated. The product was purified by a silica gel column chromatography (MeOH/DCM) to afford 146 mg (90%) of (5)-methyl 2-((4-(5-(3- cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 -yl)amino)acetate as white solid. LCMS-ESI (m/z) calculated for C24H24N403S: 448.2; found 449.1 [M+H]+, tR = 2.48 min.
[0447] ( i)-methyl 2-((4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3- dihydro-1 H-inden- l-yl)amino)acetate was prepared in an analogous fashion using (Z?)-5-(5- ( 1 -amino-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 5.
[0448] (S)-methyl 2-((tert-butoxycarbonyl)(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4- thiadiazol-2-yl)-2,3-dihydro-lH-inden-l-yl)amino)acetate
Figure imgf000116_0001
[0449] To a solution of (5)-methyl 2-((4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4- thiadiazol-2-yl)-2,3-dihydro-l H-inden- l-yl)amino)acetate (146 mg, 0.35 mmol) in DCM (2 mL) was added di-tert-butyl dicarbonate (85.3 mg, 0.39 mmol) and reaction was stirred at room temperature for 16 h. Reaction was diluted with DCM (10 mL) and washed with NaHC03, water, and brine. The product was purified by a silica gel column chromatography (EA / hexanes) to afford 118 mg (66%) of (5)-methyl 2-((tert-butoxycarbonyl)(4-(5-(3- cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)amino)acetate as white solid. LCMS-ESI (m/z) calculated for C29H32N405S: 548.2; found no M+, tR = 4.19 min.
[0450] (i?)-methyl 2-((tert-butoxycarbonyl)(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4- thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- l-yl)amino)acetate was prepared in an analogous fashion using (R)-methyl 2-((4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3- dihydro- 1 H-inden- 1 -yl)amino)acetate.
[0451] (S)-2-((tert-butoxycarbonyl)(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol- 2-y -2, 3-dihydro-lH-inden-l-yl)amino)acetic acid
Figure imgf000116_0002
[0452] To a stirred solution of (5 -methyl 2-((tert-butoxycarbonyl)(4-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)amino)acetate ( 120 mg, 0.21 mmol) in MeOH (2 mL) was added 6N solution of sodium hydroxide (180 μί) and the mixture was stirred at room temperature for overnight. The solvent was evaporated and the residue was dissolved in water (5 mL) and acidified with IN HCl. The mixture was extracted with EA (3 x 5 mL) and the organic layers washed with brine, dried over MgS0 , and concentrated to afford 108 mg (92%) of (5 2-((/ert-butoxycarbonyl)(4-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 yl)amino)acetic acid as white solid which was used in the next experiments without purification. LCMS-ESI (m/z) calculated for C28H3oN4C>5S: 534.2; found no M+, tR = 3.81 min.
[0453] (i? -2-((tert-butoxycarbonyl)(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol- 2-yl) -2,3-dihydro-l H-inden- lyl)amino)acetic acid was prepared in an analogous fashion using (i?)-methyl 2-((tert-butoxycarbonyl)(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4- thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)amino)acetate.
[0454] (S)-methyl 2-((4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2, 3-dihydro-lH- inden- -yl)amino)acetate (Compound 99)
Figure imgf000117_0001
[0455] Prepared using General Procedure 9. To a suspension of (5 -5-(2-(l-amino-2,3- dihydro-lH-inden-4-yl)thiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 71 (150 mg, 0.36 mmol) in CH3CN (5 mL) was added K2C03 (150.9 mg, 1.09 mmol) and methyl 2- bromoacetate (66 mg, 0.43 mmol). The suspension was stirred at 80°C for 16 h. The reaction mixture was filtered and concentrated. The residue was re-suspended in EA (15 mL), washed with water and brine, dried and concentrated. The product was purified by a silica gel column chromatography (EA / hexanes) to afford 76 mg (47%) of (5)-methyl 2-((4-(2-(3- cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)amino)acetate as white solid. LCMS-ESI (m/z) calculated for C25H25N303S: 447.2; found 448.2 [M+H]+, tR = 2.57 min. 1H NMR (400 MHz, CDC13) δ 8.23 - 8.01 (m, 2H), 7.90 (s, 1H), 7.41 (dd, J= 21.0, 7.4 Hz, 1H), 7.29 (dd, J = 9.8, 5.2 Hz, 1H), 7.04 (d, J = 8.9 Hz, 1H), 7.04 (d, J - 8.9 Hz, 1H), 4.74 (dt, J= 12.1, 6.1 Hz, 1H), 4.33 (t, J= 6.1 Hz, 1H), 3.76 (d, J= 4.8 Hz, 3H), 3.55 (s, 2H), 3.24 (ddd, J = 15.8, 8.2, 5.6 Hz, 1H), 3.05 - 2.86 (m, 1H), 2.47 - 2.25 (m, 2H), 2.02 - 1.84 (m, 1H), 1.53 - 1.36 (m, 6H). 13C NMR (101 MHz, CDC13) δ 173.14, 164.87, 161.08, 146.06, 141.45, 141.28, 138.20, 132.13, 131.96, 128.08, 127.98, 127.56, 126.77, 124.66,
116.17, 114.01, 103.76, 72.55, 62.94, 52.19, 48.53, 32.99, 31.34, 22.04.
[0456] ( )-methyl 2-((4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-diliydro-lH- inden-l-yl)amino)acetate was prepared in an analogous fashion using (7?)-5-(2-(l-amino-2,3- dihydro- 1 H-inden-4-yl)thiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 72.
[0457] (S)-methyl 2-((tert-butoxycarbonyl)(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5- yl)- -dihydro- lH-inden- 1 -yl)amino )acetate
Figure imgf000118_0001
[0458] To a solution of (^-methyl 2-((4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)- 2,3-dihydro-lH-inden-l-yl)amino)acetate (76 mg, 0.17 mmol) in DCM (1 mL) was added di- tert-butyl dicarbonate (44.5 mg, 0.20 mmol) and reaction was stirred at room temperature for 16 h. Reaction was diluted with DCM (10 mL) and washed with NaHC03, water, brine and then dried. Concentration of the filtrate gave 90 mg (96%) of (5)-methyl 2-((tert- butoxycarbonyl)(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l- yl)amino)acetate as white solid. LCMS-ESI (m/z) calculated for C3oH33N305S: 547.21; found no M+, ¾ = 4.42 min.
[0459] (i?)-methyl 2-((tert-butoxycarbonyl)(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5- yl)-2,3-dihydro-lH-inden-l-yl)amino)acetate was prepared in an analogous fashion using (i?)-methyl 2-((4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl) amino) acetate.
[0460] (S)-2-((tert-butoxycarbonyl)(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2, 3- dihydro-lH-inden-1 -yl)amino)acetic acid
Figure imgf000118_0002
[0461] To a stirred solution of (5)-methyl 2-((tert-butoxycarbonyl)(4-(2-(3-cyano-4- isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l-yl)amino)acetate (120 mg, 0.22 mmol) in MeOH (2 mL) was added 6N sodium hydroxide (180 μί) and the mixture was stirred at room temperature for overnight. The solvent was evaporated and the residue was dissolved in water (5 mL) and acidified with IN HC1. The mixture was extracted with EA (3 x 5 mL) and the organic layers washed with brine, dried over MgS04, and concentrated to afford 110 mg (94%) of (-S)-2-((tert-butoxycarbonyl)(4-(2-(3-cyano-4- isopropoxyphenyl)tWazol-5-yl)-2,3-dihydro-lH-inden-l-yl)arnino)acetic acid as white solid which was used in the next experiments without purification. LCMS-ESI (m/z) calculated for C29H31N305S: 533.2; found 534.2 [M+H]+, tR = 3.92 min.
[0462] (/?)-2-((tert-butoxycarbonyl)(4-(2-(3-cyano-4-isopropoxyphenyl) thiazol-5-yl) - 2,3-dihydro-lH-inden-l-yl)amino)acetic acid was prepared in an analogous fashion using (i?)-methyl 2-((tert-butoxycarbonyl)(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3- dihydro- 1 H-inden- 1 -yl)amino)acetate.
[0463] General Procedure 10. Preparation of Indane Amino Amides
[0464] To the Boc-protected (R)- or (5)-indane aminoacid (1 equivalent) in DMF (2 M) was added HOBt (1.35 eq) and EDC (1.35 eq) and the reaction was stirred at room temperature for 60 min. The appropriate amine (1.1 eq) was added and the reaction was stirred at room temperature for 2 h. The Boc-protected amino amide was precipitated out of water or extracted with EA and dried over MgS04. The product was purified by recrystallization or preparative HPLC. The resulting solid was heated in 4M HCl / dioxane at 50°C until the reaction was complete. The product was precipitated as the hydrochloride salt by the addition of diethyl ether.
[0465] Compounds 21 - 25, 39, and 98, 100 - 108 were prepared using General Procedure 10.
[0466] (S)-2-((4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3-dihydro-lH- ind -l-yl)amino)-N,N-dimethylacetamide hydrochloride (Compound 21)
Figure imgf000119_0001
[0467] Prepared using General Procedure 10. To (5 2-((tert-butoxycarbonyl)(4-(5-(3- cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 yl)amino)acetic (25 mg, 0.04 mmol) was added HOBt (9.4 mg, 0.07 mmol) and EDC (13.3 mg, 0.07 mmol) in anhydrous DMF (1 mL) and the reaction mixture stirred at room temperature for 60 min. Dimethyl amine (2.3 mg, 0.05 mmol) was added and the mixture was stirred at room temperature for 12 h. The crude reaction was purified by preparative HPLC purification to provide the Boc-product amido amide as white solid. This material was treated with 4N HCl in dioxane at 50°C for 2 h. The reaction mixture was diluted with diethyl ether (5 mL), and the resulting solid collected to afford 10 mg (46% over two steps) of (5)-2-((4-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)amino)-N,N- dimethylacetamide hydrochloride 21. LCMS-ESI (m/z) calculated for C25H27N502S: 461.2; found 462.1 [M+H]+, tR - 3.90 min.
[0468] (i?)-2-((4-(5-(3-cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yt)-2,3-dihydro- 1H- inden-l-yl)amino) -N,N-dimethylacetamide hydrochloride 22 was prepared in an analogous fashion using (i?)-2-((tert-butoxycarbonyl)(4-(5-(3-cyano-4~isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 yl)amino)acetic.
[0469] 5-(5-((S)-l-((2-((S)-3-hydroxypyrrolidin-l-yl)-2-oxoethyl)am
inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile (Compound 104)
Figure imgf000120_0001
[0470] Prepared using General Procedure 10. To (-S)-2-((tert-butoxycarbonyl)(4-(2-(3- cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)amino)acetic acid (12 mg, 0.02 mmol) and HOBt (4.5 mg, 0.03 mmol) was added EDC (6.4 mg, 0.03 mmol) in anhydrous DMF (1 mL) and the reaction mixture stirred at room temperature for 60 min. (S)- pyrrolidin-3-ol (2.3 mg, 0.02 mmol) was added and the mixture was stirred at room temperature for 12 h. The crude reaction was purified by preparative HPLC purification to provide the Boc-product amido amide as white solid. This material was treated with 4N HCl in dioxane at 50°C for 2 h. The reaction mixture was diluted with diethyl ether (5 mL), and the resulting solid collected to afford 5 mg (50% over two steps) of 5-(5-((5)-l-((2-((5)-3- hydroxypyrrolidin- 1 -yl)-2-oxoethyl)amino)-2,3-dihydro- 1 H-inden-4-yl) thiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 104. LCMS-ESI (m/z) calculated for C28H30N4O3S: 502.2; found 503.2 [M+H]+, tR = 3.77 min.
[0471] 5-(5-((/?)-l-((2-((5)-3-hydroxypyrrolidm-l-yl)-2-oxoethyl)an ino)-2,3-dihydro- lH-inden-4-yl) thiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 102 was prepared in an analogous fashion using (i?)-2-((tert-butoxycarbonyl)(4-(2-(3-cyano-4- isopropoxyphenyl)thiazol-5-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)amino)acetic acid.
[0472] (R)-2-((4-(5-(3-cyano-4 sopropoxyphenyl)-l,3 -t iadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl)amino)acetic acid (Compound 27)
Figure imgf000120_0002
[0473] To a stirred solution of (7?)-2-((tert-butoxycarbonyl)(4-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)amino)acetic acid (20 mg, 0.03) mmol in 1,4-dioxane (0.5 mL) was added 4N HCl in 1,4-dioxane (0.2 mL). The mixture was stirred at 50°C for 2 h before it was concentrated and triturated with ether to afford 13 mg of (i?)-2-((4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3- dihydro-1 H-inden- l-yl)amino)acetic acid as yellow-green solid. LCMS-ESI (m/z) calculated for C23H22N403S: 434.14, found 435.2 [M+H]+, tR - 2.51 min.
[0474] (5 -2-((4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl)amino)acetic acid 26 was in prepared in similar fashion using (_S)-2-((tert- butoxycarbonyl)(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl)amino) acetic acid.
[0475] (R)-2-((4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2 -dihydro-lH-inden-l- yl)amino)acetic acid (Compound 96)
Figure imgf000121_0001
[0476] To (i?)-methyl 2-((4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- l-yl)amino)acetate (100 mg, 0.22 mmol) in ethanol was added 2N NaOH (1.1 mL) and the mixture stirred at room temperature for 12 h. The solvent was evaporated and the residue dissolved in water and acidified with IN HCl. The resulting solid was filtered and dried to give 60 mg (63%) of (i?)-2-((4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3- dihydro-1 H-inden- l-yl)amino)acetic acid 96 as yellow-green solid. LCMS-ESI (m/z) calculated for C24H23N303S: 433.1; found 434.2 [M+l] f, tR = 2.61 min.
[0477] (1S)-2-((4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H- inden- 1 - yl)amino)acetic acid 97 was prepared in an analogous fashion using (S)-methyl 2-((4-(2-(3- cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)amino)acetate.
[0478] General Procedure 11. Reductive Animations of Indane Amines.
[0479] To a solution of the primary or optionally substituted secondary (R)- or (5)-indane amine (1 eq) in MeOH (0.01 M) was added acetic acid (0.01 eq) and the appropriate aldehyde (1.1 eq). The reaction was stirred at 25-50°C until imine formation was complete (2-18 h). Sodium borohydride or sodium triacetoxyborohydride (10 eq) was added and the reaction was stirred at room temperature until reduction was complete (2-8 h). The solvent was evaporated and the residue partitioned between NaHC03 and EA. The organic layer was collected, dried and purified by preparative HPLC.
[0480] Compounds 28 - 30, 109 and 110 were prepared using General Procedure 11.
[0481] (S)-5-(5-(l-(((lH-imidazol-2-yl)methyl)ami^
thiadiazol-2-yl)-2-isopropoxybenzonitrile (Compound
Figure imgf000122_0001
[0482] Prepared using General Procedure 11. To (5)-5-(5-(l-amino-2,3-dihydro-lH- inden-4-yl)-l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 4 (25 mg, 0.06 mmol) and lH-imidazole-2-carbaldehyde (6.4 mg, 0.06 mmol) in anhydrous MeOH (1 mL) was added acetic acid (1 drop). The solution was stirred at 55°C for 3 h before cooling to room temperature and the addition of NaBH4 (4.6 mg, 0.12 mmol). The mixture was stirred at room temperature for 12 h. The reaction mixture was quenched with water (0.5 mL) and partitioned between EA (5 mL) and water (5 mL). The organic layers washed with water and brine, and the product purified by preparative HPLC to afford 22 mg (81%) of (5)-5-(5-(l- ((( 1 H-imidazol-2-yl)methyl)amino)-2,3 -dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile 28 as half- white solid. LCMS-ESI (m/z) calculated for C25H24N6OS: 456.2; found 457.2 [M+H]+, tR = 2.38 min.
[0483] (i?)-5-(5-(l-(((lH-imidazol-2-yl)methyl)amino)-2,3-dihydro-lH-inden-4-yl)- l,3,4-thiadiazol-2-yl) -2-isopropoxybenzonitrile 29 was prepared in an analogous fashion using ( ?)-5-(5-(l-amino-2,3-dihydro-lH-inden-4-yl)-l,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 5.
[0484] (R)-5-(5-(l-(((lH-imidazol-2-yl)methyl)a ino)-2 -dihydro
yl)thiazol-2- l)-2-isopropoxybenzonitrile (Compound
Figure imgf000122_0002
[0485] Prepared using General Procedure 11. To (i?)-5-(5-(l-amino-2,3-dihydro-lH- inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 72 (20 mg, 0.05 mmol) and lH-imidazole-2-carbaldehyde (7 mg, 0.07 mmol) in anhydrous MeOH (0.5 mL) was added acetic acid (1 drop). The solution was stirred at 55°C for 3 h before cooling to room temperature and the addition of NaBH4 (37.8 mg, 0.1 mmol). The mixture was stirred at room temperature for 12 h. The reaction mixture was quenched with water (0.5 mL) and partitioned between EA (5 mL) and water (5 mL). The organic layers washed with water and brine, and the product purified by preparative HPLC to afford 17 mg (77%) of (i?)-5-(5-(l- (((lH-imidazol-2-yl)methyl)amino)-2,3-dihydro
isopropoxybenzonitrile 109. LCMS-ESI (m/z) calculated for C^H^NsOS: 455.2; found 456.2 [M+H]+, tR = 2.53 min.
[0486] (5)-5-(5-( 1 -((( 1 H-imidazol-2-yl)methyl)amino)-2,3-dihydro- 1 H-inden-4- yl)thiazol-2-yl)-2 -isopropoxybenzonitrile 110 was prepared in an analogous fashion using (5)-5-(5-( 1 -amino-2,3-dihydro- 1 H-inden-4-yl)thiazol-2-yl)-2 -isopropoxybenzonitrile hydrochloride 71.
[0487] (S)-2 sopropoxy-5-(5-(l-((2-(methylsulfonyl)ethyl)amino)-2,3-dihy
4-y -l,3,4-thiadiazol-2-yl)benzonitrile (Compound 31)
Figure imgf000123_0001
[0488] To a stirred solution of (5)-5-(5-(l-amino-2,3-dihydro-lH-inden-4-yl)-l,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 4 (25 mg, 0.06 mmol) and DIEA (32 mg, 0.24 mmol) in DMA (1 mL) was added (methylsulfonyl)ethene (20 mg, 0.18 mmol). The reaction mixture was heated at 90°C for 24 h. The solvent was evaporated and the product purified by preparative HPLC to afford 9 mg (31%) of (5)-2-isopropoxy-5-(5-(l-((2- (methylsulfonyl)emyl)ammo)-2,3-dihydro-lH-inden-4-yl)-l,3,4-tWadiazol-2-yl)benzonitri 31 as half- white solid. LCMS-ESI (m/z) calculated for C24H26N403S2: 482.1; found 483.1 [M+H]+, tR = 2.49 min.
[0489] (i?)-2-isopropoxy-5-(5-(l-((2-(memylsulfonyl)emyl)amino)-2,3-dihydro-lH- inden-4-yl)-l,3,4-thiadiazol-2-yl)benzonitrile 32 was prepared in an analogous fashion using (i?)-5-(5-(l -amino-2,3-dihydro- 1 H-inden-4-yl)-l ,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 5.
[0490] (S)-2Hsopropoxy-5-(5-(l-((2-(methylsulfonyl)ethyl)amino)-2,3-dihydro-lH-ind 4-yl)thiazol-2-yl)benzonitrile (Compound 221)
Figure imgf000123_0002
[0491] To a stirred solution of (5 -5-(5-(l-ammo-2,3-dihydro-lH-inden-4-yl)thiazol-2- yl)-2-isopropoxybenzonitrile hydrochloride 71 (60 mg, 0.15 mmol) and DIEA (32 mg, 0.24 mmol) in 1,4-dioxane (0.5 mL) was added (methylsulfonyl)ethene (92 mg, 0.88 mmol). The reaction mixture was heated at 90°C for 24 h. The reaction was diluted with DCM (5 mL) and washed with saturated aqueous ammonium chloride (2 x 5 mL) and saturated aqueous sodium bicarbonate (2 x 5 mL) and then dried. The crude reaction was purified by a silica gel column (MeOH/DCM) to yield 44 mg (61%) of (5)-2-isopropoxy-5-(5-(l-((2- (methylsulfonyl)emyl)amino)-2,3-dihydro-lH-inden-4-yl)tWazol-2-yl)benzonitrile 221 as brown liquid. LCMS-ESI (m/z) calculated for C25H27N303S2: 481.1; found 482.1 [M+H]+, tR = 2.49 min. lH NMR (400 MHz, CDC13) δ 8.19 - 8.08 (m, 2H), 7.92 (s, 1H), 7.46 (dd, J = 7.4, 0.9 Hz, 1H), 7.33 (dt, J= 14.9, 7.3 Hz, 2H), 7.06 (d, J= 8.9 Hz, 1H), 5.31 (s, 1H), 4.75 (dt, J = 12.2, 6.1 Hz, 1H), 4.35 (t, J= 6.6 Hz, 1H), 3.41 - 3.15 (m, 5H), 3.10 - 2.96 (m, 4H), 2.57 - 2.45 (m, 1H), 1.93 (ddd, J= 12.8, 6.2, 1.7 Hz, 1H), 1.46 (d, J= 6.1 Hz, 6H).
[0492] (i?)-2-isopropoxy-5-(5-(l -((2-(methylsulfonyl)ethyl)amino)-2,3-dihydro- 1 H- inden-4-yl)thiazol-2-yl)benzonitrile 220 was prepared in an analogous fashion using (i?)-5-(5- ( 1 -amino-2,3 -dihydro- 1 H-inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 72.
[0493] General Procedure 12. Preparation of Indane Sulfonamides via Sulfonyl Chlorides
[0494] To a stirred solution of (R)- or (5)-indane amine (1 eq) in DCM (0.08M) was added TEA (3 eq) and the appropriate sulfonyl chloride (1.5 eq.) at room temperature. The reaction was stirred at room temperature for 18 h. The solvent was evaporated and the pure product isolated after preparative HPLC purification.
[0495] Compounds 33 - 36 and 111 - 120 were prepared using General Procedure 12.
[0496] (S)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden- -yl)methanesulfonamide (Compound 33)
Figure imgf000124_0001
[0497] Prepared using General Procedure 12. To a stirred solution of (5)-5-(5-(l- amino-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 4 (20 mg, 0.04 mmol) and TEA (14.7 mg, 0.14 mmol) in DCM (2 mL) was added methane sulfonylchloride (8.3 mg, 0.07 mmol) and the mixture was stirred at room temperature for 16 h. The solvent was evaporated and the residue purified by preparative HPLC to afford 12 mg (55%) of (,S N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol- 2-yl)-2,3-dihydro-lH-inden-l-yl)methanesulfonamide 33 as white solid. LCMS-ESI (m/z) calculated for C22H22N403S2: 454.1; found 455.1 [M+H]+, tR = 3.48 min.
[0498] (i? N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl)methanesulfonamide 34 was prepared in an analogous fashion using (R)-5- (5-(l- amino-2,3 -dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 5.
[0499] (R)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH nden-l - yl)methanesulfonamide (Compound 111)
Figure imgf000125_0001
[0500] Prepared using General Procedure 12. (i?)-5-(5-(l-amino-2,3-dihydro-lH-inden- 4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 72 (60 mg, 0.15 mmol) and TEA (0.06 mL, 0.4 mmol) in DCM (0.5 mL) was added methane sulfonylchloride (8.3 mg, 0.07 mmol) at 0°C and the reaction mixture was stirred at room temperature for 16 h. The mixture was diluted with DCM (5 mL) and washed with aqueous ammonium chloride, and brine. The crude material was purified by silica gel column chromatography (MeOH / DCM) to afford 39 mg (58%) of (i?)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl) -2,3-dihydro-lH- inden-l-yl) methane sulfonamide 111 as white solid. LCMS-ESI (m/z) calculated for C23H23N303S2: 453.1; found 454.1 [M+H]+, tR = 3.64 min.
[0501] (-S)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H- inden- 1 - yl)methanesulfonamide 112 was prepared in an analogous fashion using (5)-5-(5-(l-amino- 2,3-dmydro-lH-inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 71.
[0502] (S)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3-dihydro-lH- ind -1 - l)ethenesulfonamide
Figure imgf000125_0002
[0503] To a stirred solution of (5)-5-(5-(l-amino-2,3-dihydro-lH-inden-4-yl)-l,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 4 (40 mg, 0.5 mmol) and TEA (49 mg, 0.48 mmol) in DCM (2 mL) was added 2-chloroethanesulfonyl chloride (79 mg, 0.48 mmol) at 0°C. The reaction was warmed to room temperature and stirred for 2 h. The reaction was quenched by the addition of NaHC03. The product was purified by chromatography (EA/hexane) to provide 30 mg (66%) of (5 -N-(4-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)ethenesulfonamide as yellow solid. LCMS-ESI (m/z) calculated for C2 H22N403S2: 466.1; found 467.1 [M+H]+, tR = 3.63 min.
[0504] 7?)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l ,3,4-thiadiazol-2-yl)-2,3-dihydro-l H- inden-l-yl)ethenesulfonamide was prepared in an analogous fashion using (i?)-5-(5-(l-amino- 2,3-dihydro-lH-inden-4-yl)-l ,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 5.
[0505] (R)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2 -dihydro-lH-inden-l^ yl)ethenesulfonamide
Figure imgf000126_0001
[0506] To (Λ)-5-(5-(1 -amino-2,3 -dihydro- 1 H-inden-4-yl)thiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 72 (0.5 g, 1.3 mmol) in DCM (10 mL) was added TEA (0.88 mL, 6.3 mmol) followed by 2-chloroethanesulfonyl chloride (0.4 mL, 163 mmol) at 0°C and the reaction was stirred at room temperature overnight. During this time additional reagents TEA (0.2 mL) and 2-chloroethanesulfonyl chloride (0.15 mL) were added to drive the reaction to completion. The reaction mixture was concentrated and the crude residue was purified by a silica gel column (EA / hexanes) to afford 378 mg of (R)-N-(4-(2-(3-cyano-4- isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l-yl)ethenesulfonamide as fine yellow powder. LCMS-ESI (m/z) calculated for C24H23N303S2: 465.12; found 466.1 [M+H]+, tR = 3.82 min. 1H NMR (400 MHz, CDC13) δ 8.10 (s, 2H), 7.85 (s, 1H), 7.48 - 7.26 (m, 3H), 7.01 (d, J= 7.3 Hz, 1H), 6.64 (dd, J= 16.5, 9.8 Hz, 1H), 6.33 (d, J= 16.5 Hz, 1H), 5.97 (d, J= 9.8 Hz, 1H), 4.90 (d, J = 7.3 Hz, 1H), 4.77 - 4.46 (m, 2H), 3.32 - 2.83 (m, 2H), 2.64 (s, 1H), 2.02 - 1.84 (m, 1H), 1.40 (t, J= 5.8 Hz, 6H).
[0507] (5)-N-(4-(2-(3-cyano-4-isoprorx)xyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l- yl) ethane sulfonamide was prepared in an analogous fashion using (5 -5-(5-(l-amino-2,3- dihydro-lH-indm-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 71.
[0508] General Procedure 13. Preparation of Indane Sulfonamides via Michael Addition [0509] To a stirred solution of the (/?)- or (5)-indane vinyl sulfonamide (1 eq) in DMF (0.1M) was added the appropriate amine (10 eq) The reaction was stirred at 80°C for 18 h. The product was purified by preparative HPLC.
[0510] Compounds 37 - 38 and 121 - 153 were prepared using General Procedure 13.
[0511] N-((S)-4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-1 -yl)-2-((R)-3-hydroxypyrrolidin-l -yl)ethanesulfonamide (Compound 37)
Figure imgf000127_0001
[0512] Prepared using General Procedure 13. To a solution of 5)-N-(4-(5-(3-cyano-
4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 -yl)ethenesulfonamide (40 mg, 0.5 mmol) in DMF (0.5 mL) was added (i?)-pyrrolidin-3-ol (18.7 mg, 0.21 mmol) and the reaction was heated to 80°C for 18 h. The product was purified by preparative HPLC to give 30 mg (56%) of N-((5)-4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3- dihydro-1 H-inden- l-yl)-2-((/?)-3-hydroxypyrrolidin-l-yl)ethanesulfonamide 37 as an off- white solid. LCMS-ESI (m/z) calculated for C27H31N504S2: 553.2; found 554.2 [M+H]+, fR = 2.52 min.
[0513] N-((/-)-4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH -inden-1 -yl)-2- ((i?)-3-hydroxypyrrolidin-l-yl)ethanesulfonamide 38 was prepared in an analogous fashion using (7?)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)- 2,3 -dihydro- 1 H-inden- 1 -yl)ethenesulfonamide..
[0514] N-((R)-4-(2-(3-cyano-44sopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l- -2-((R)-3-hydroxypiperidin-l-yl)ethanesulfonamide (Compound 143)
Figure imgf000127_0002
[0515] Prepared using General Procedure 13. To a solution of (i?)-N-(4-(2-(3-cyano-4- isopropoxyphenyl)tWazol-5-yl)-2,3-dmydro-lH-inden-l-yl)emenesulfonarnide (10 mg, 0.02 mmol) in DMF (0.5 mL) was added (i?)-piperidin-3-ol hydrochloride (20.6 mg, 0.15 mmol) and the reaction was heated to 80°C for 18 h. The product was purified by preparative HPLC to give 10 mg (80%) of N-((R)-4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- l-yl)-2-((i?)-3-hydroxypiperidin-l-yl)ethanesulfonamide 143. LCMS-ESI (m/z) calculated for C29H34N404S2: 566.2; found 567.2 [M+H]+, tR = 2.62 min. [0516] N-((5)-4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- 1 - yl)-2- ((i?)-3-hydroxypiperidin-l-yl)ethanesulfonarnide 141 was prepared in an analogous fashion using (1S)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- 1 -yl)ethenesulfonamide.
[0517] General Procedure 14. Preparation of Indane Sulfonamide Esters
[0518] To a stirred solution of (R)- or (5)-indane amine (1 eq) in DCM (0.2 M) was added the sulfonyl chloride (1.5 eq) at room temperature. For less reactive or hindered sulfonyl chloride esters DIEA (2-3 eq) was added. The reaction was stirred at room temperature for 18 h. The crude reaction was partitioned between DCM and NaHC03. The organic layer was dried over MgS04, concentrated, and purified by column chromatography.
[0519] Compounds 154 - 157 were prepared using General Procedure 14.
[0520] (S)-ethyl 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3- dih dro-lH-inden-l-yl)sulfamoyl)acetate
Figure imgf000128_0001
[0521] Prepared using General Procedure 14: To a stirred solution of (.S -5-(5-(l- ammo-2,3-dihydro-lH-inden-4-yl)-l,3,4-tWadiazol-2-yl)-2-isopropoxybenzoni le 4 (177 g, 0.47 mmol) and DIEA (182 mg, 1.4 mmol) in DCM (8 mL) was added freshly prepared ethyl-2-(chlorosulfonyl)acetate (131 mg, 0.7 mmol). After 45 min, the crude reaction was partitioned between DCM and NaHC03. The organic layer was dried over MgS04, concentrated, and purified by column chromatography (EA/hexanes) to provide 75 mg (30%) of («S)-ethyl 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl) -l,3,4-thiadiazol-2-yl)-2,3- dihydro- 1 H-inden- l-yl)sulfamoyl)acetate as light yellow solid. LCMS-ESI (m/z) calculated for C25H26N405S2: 526.1; found 527.1 [M+H]+, tK = 3.71 min. Ή NMR (400 MHz, CDC13) δ 8.16 (dd, J = 8.9, 2.3 Hz, 1H), 8.09 (d, J = 2.2 Hz, 1H), 7.76 (d, J = 7.7 Hz, 1H), 7.57 (d, J = 7.6 Hz, 1H), 7.35 (t, J = 7.6 Hz, 1H), 7.03 (d, J = 9.0 Hz, 1H), 5.46 (t, J = 7.9 Hz, 1H), 4.70 (dt, J = 12.2, 6.1 Hz, 1H), 4.26 - 4.17 (m, 2H), 4.00 (d, J = 8.2 Hz, 2H), 3.49 (ddd, J = 17.4, 9.5, 3.9 Hz, 1H), 3.26 - 3.05 (m, 1H), 2.56 (ddd, J = 12.9, 9.0, 4.4 Hz, 1H), 2.23 - 2.08 (m, 1H), 1.41 - 1.37 (m, 6H), 1.28 (dd, J= 1 1.7, 4.6 Hz, 3H).
[0522] (7?)-ethyl 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3- dihydro-1 H-inden- l-yl)sulfamoyl)acetate was prepared in an analogous fashion using (R)-5- (5-( 1 -amino-2,3-dihydro- 1 H-inden-4-yl)- 1 ,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile 5. [0523] (S)-methyl 2-(N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2, 3-dihydro-lH- inden- -yl)sulfamoyl)acetate (Compound 155)
Figure imgf000129_0001
[0524] Prepared using General Procedure 14: To a solution of (5)-5-(5-(l-amino-2,3- dmydro-lH-inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile 71 (20 mg, 0.04 mmol) in DCM (1 mL) was added methyl-2-(chlorosulfonyl)acetate (10 mg, 0.04 mmol). The reaction mixture was stirred at room temperature overnight and then diluted with DCM (5 mL), washed with saturated aq. NaHC03, and brine. The organic layers were dried over MgS04, and the crude product purified by silica gel column chromatography to afford 11.2 mg (41%) of (5)-methyl 2-(N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- l-yl)sulfamoyl)acetate 155 as orange-brown oil. LCMS-ESI (m/z) calculated for C25H25N305S2: 511.1; found 512.2 [M+H]+, tR = 3.71 min.
[0525] (i?)-methyl 2-(N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- lH-inden-l-yl)sulfamoyl)acetate 154 was prepared in an analogous fashion using (R)-5-(5- (1 -amino-2,3-dihydro-l H-inden-4-yl)thiazol-2-yl)-2-isopropoxyberizonitrile 72.
[0526] General Procedure 15. Preparation of Indane Sulfonamide Acids
[0527] To a solution of (R)- or (S)-indane sulfonamide ester (1 eq) in 2: 1 EtOH/THF (0.2 M) was added 6N NaOH (5 eq) at room temperature. The reaction was stirred at room temperature for 24 h. The crude reaction was concentrated then partitioned between DCM/IPA and IN HC1. The organic layer was dried over MgS04, concentrated, and isolated after preparative HPLC purification.
[0528] Compounds 40 - 41 and 158 - 161 were prepared using General Procedure 15.
[0529] (S)-2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3-dihydro- -inden-l-yl)sulfamoyl)acetic acid (Compound 40)
Figure imgf000129_0002
[0530] Prepared using General Procedure 15: To a stirred solution of (.S)-ethyl 2-(N-(4- (5-(3-cyano-4-isopropoxyphenyl)- 1 ,3,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 - yl)sulfamoyl) acetate (75 mg, 0.8 mmol) in MeOH (4 mL) was added 6N NaOH (0.12 mL). After 3 h, the crude reaction was concentrated then partitioned between DCM/IPA and IN HC1. The organic layer was dried over MgS04 and concentrated to give 43 mg (60%) of (5)- 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)- 1 ,3,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 - yl)sulfamoyl)acetic acid 40 as light yellow solid. LCMS-ESI (m/z) calculated for C23H22N405S2: 498.1; found 499.1 [M+H]+, tR = 3.34 min.
[0531] (^)-2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- l-yl)sulfamoyl)acetic acid 41 was prepared in an analogous fashion using (i?)-ethyl 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l ,3,4-thiadiazol-2-yl)-2,3-dihydro- 1 H- inden- 1 -yl) sulfamoyl)acetate.
[0532] (S)-2-(N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2 -dihydro-lH-inden- l-yl)sulfamoyl)acetic acid (Compound 159)
Figure imgf000130_0001
[0533] Prepared using General Procedure 15: To a stirred solution containing (5)-methyl 2-(N-(4-(2-(3-cyano-4-isopropoxyphenyl)tWazol-5-yl)-2,3-dihydro-lH-inden-l-yl)sulfamoyl )acetate (11.2 mg, 0.02 mmol) in MeOH (1 mL) was added 6N NaOH (100 uL). After 1 h, the crude reaction was concentrated and the product purified by preparative HPLC to give 5 mg (45%) of (5)-2-(N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1H- inden-l-yl)sulfamoyl)acetic acid 159 as light yellow solid. LCMS-ESI (m/z) calculated for C24H23N305S2: 497.1; found 498.1 [M+H]+, fR = 3.44 min.
[0534] (i?)-2-(N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H- inden-l-yl)sulfamoyl)acetic acid 158 was prepared in an analogous fashion using (i?)-methyl 2-(N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- 1 - yl)sulfamoyl)acetate.
[0535] General Procedure 16. Preparation of Indane Sulfonamide Amides
[0536] To a stirred solution of (R)- or (5)-indane sulfonamide acid (1 eq) in DCM (0.25 M) was added HATU (3 eq) and DIEA (2 eq). After 30 min, the amine was added and the reaction mixture stirred 18 h at room temperature. The reaction was quenched with water and purified by preparative HPLC.
[0537] Compounds 42 - 44, 162, and 163 were prepared using General Procedure 16.
[0538] (R)-2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3-dihydro- 1 H-inden- l-yl)sulfamoyl)-N,N-dimethylacetamide (Compound 43)
Figure imgf000131_0001
[0539] Prepared using General Procedure 16: To (R)-2-(N-(4-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)sulfamoyl)acetic acid (20 mg, 0.04 mmol) in DCM (0.4 mL) was added HATU (45 mg, 0.12 mmol) and DIEA (10.3 mg, 0.08 mmol). After 30 min, dimethylamine (2M solution in THF, 200 μί, 0.4 mmol) was added and the reaction stirred for 18 h at room temperature. The reaction was quenched with water (100 μί) and the solvent evaporated. The crude material was purified by preparative HPLC to afford 14 mg (66%) of (R)-2-(N-(4-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)sulfamoyl)-N,N- dimethylacetamide 43 as white solid. LCMS-ESI (m/z) calculated for C25H27N504S2: 525.2; found 526.2 [M+H]+, ¾ = 3.42 min. Ή NMR (400 MHz, CDC13) 6 8.20 (dd, J= 8.9, 2.3 Hz, 1H), 8.14 (d, J = 2.3 Hz, 1H), 7.82 (d, J= 7.7 Hz, 1H), 7.73 (d, J = 7.6 Hz, 1H), 7.40 (t, J = 7.7 Hz, 1H), 7.09 (d, J= 9.0 Hz, 1H), 5.50 (d, J= 8.2 Hz, 1H), 5.07 (q, J= 7.7 Hz, 1H), 4.76 (hept, J = 6.1 Hz, 1H), 4.28 (d, J = 14.6 Hz, 1H), 4.09 (d, J = 14.6 Hz, 1H), 3.50 (ddd, J = 17.0, 8.8, 3.4 Hz, 1H), 3.20 (dt, J= 9.7, 7.1 Hz, 1H), 3.15 (s, 3H), 3.02 (s, 3H), 2.72 (dtd, J = 11.4, 8.0, 3.5 Hz, 1H), 2.20 (dq, J = 13.1, 8.4 Hz, 1H), 1.46 (d, J = 6.1 Hz, 6H). 13C NMR (101 MHz, CDC13) 6 166.99, 165.73, 163.22, 161.71, 144.17, 141.92, 133.44, 133.34, 129.31, 127.92, 127.33, 126.37, 122.70, 115.57, 113.91, 103.71, 72.56, 59.23, 54.92, 38.30, 35.99, 31.36, 21.74. Chiral HPLC: (i?)-2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2- yl)-2,3-dihydro-lH-inden-l-yl)sulfamoyl)-N,N-dimethylacetamide was eluted in 40% IPA in hexanes, 100% ee, ¾ = 22.87 min.
[0540] (5)-2-(N-(4-(5-(3 -cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl) -2,3 -dihydro- 1 H-inden- 1-yl) sulfamoyl)-N,N-dimethylacetamide 42 was prepared in an analogous fashion using (5)-2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl) sulfamoyl) acetic acid. Chiral HPLC: 97.8 % ee, ¾ for 5-enantiomer = 29.06 min.
[0541] (R)-N-(4-(2-(3-cyano-4 sopropoxyphenyl)thiazol-5-yl)-2 -dihydro-lH-inden-l- -2-morpholino-2-oxoethanesulfonamide (Compound 162)
Figure imgf000131_0002
[0542] Prepared using General Procedure 16: To (R)-2-(N-(4-(2-(3-cyano-4- isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l-yl)sulfamoyl)acetic acid 158 (15 mg, 0.03 mmol) in DCM (0.4 mL) was added HATU (26 mg, 0.07 mmol) and DIEA (7.8 mg, 0.06 mmol). After 30 min, morpholine (52 mg, 0.6 mmol) was added and the reaction stirred for 18 h at room temperature. The reaction was quenched with water (100 μϋ) and the solvent evaporated. The crude material was purified by preparative HPLC to afford 8 mg (47%) of (i?)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l- yl)- 2-morpholino-2-oxoethanesulfonamide 162. LCMS-ESI (m/z) calculated for C28H3oN405S2: 566.1; found 567.2 [M+H]+, tR = 3.77 min.
[0543] General Procedure 17. Preparation of Indane Sulfonamide Alcohols
[0544] To a stirred solution of (R)- or (5)-indane sulfonamide ester (1 eq) in THF (0.06 M) was added sodium borohydride (4 eq) at room temperature. The reaction was heated to 75°C and methanol (10 eq) was added dropwise. After 1 h, the reaction was cooled and concentrated. The pure product was obtained by preparative HPLC purification.
[0545] Compounds 45, 46, 164, and 165 were prepared using General Procedure 17.
[0546] (i?)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl)-2-hydroxyethanesulfonamide (Compound 46)
Figure imgf000132_0001
[0547] Prepared using General Procedure 17: To a stirred solution of (7?)-methyl 2-(N- (4-(5-(3 -cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- 1 - yl)sulfamoyl) acetate (13 mg, 0.02 mmol) in THF (0.5 mL) was added sodium borohydride (2.3 mg, 0.06 mmol) at room temperature. The reaction was heated to 75°C and methanol (0.03 mL, 0.7 mmol) was added dropwise. After 1 h, the reaction was cooled and concentrated. Purification of the crude material by preparative HPLC gave 6 mg (60%) of (i?)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH-inden-l- yl)-2-hydroxyethanesulfonamide 46. LCMS-ESI (m/z) calculated for C23H24N404S2: 484.1; found 485.1 [M+H]+, tR = 3.26 min. 1H NMR (400 MHz, CDC13) δ 8.23 (dd, J= 8.9, 2.3 Hz, 1H), 8.16 (d, J - 2.3 Hz, 1H), 7.83 (d, J= 7.7 Hz, 1H), 7.64 (d, J = 7.6 Hz, 1H), 7.43 (t, J = 7.6 Hz, 1H), 7.11 (d, J= 9.0 Hz, 1H), 5.19 - 4.96 (m, 1H), 4.87 - 4.63 (m, 3H), 4.17 (dd, J- 8.2, 4.4 Hz, 2H), 3.53 (ddd, J= 17.2, 8.8, 3.5 Hz, 1H), 3.46 - 3.34 (m, 2H), 3.32 - 3.11 (m, 1H), 2.86 - 2.59 (m, 1H), 2.19 - 1.97 (m, 1H), 1.48 (d, J= 6.1 Hz, 6H). 13C NMR (101 MHz, CDC13) 5 166.83, 165.72, 161.71, 144.11, 141.85, 133.47, 133.26, 129.53, 127.99, 126.92, 126.58, 122.64, 115.51, 113.87, 103.79, 72.54, 58.86, 57.43, 55.67, 34.69, 31.27, 21.73. Chiral HPLC: (i?)-N-(4-(5-(3-cyano-4-isoprorx)xyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro- lH-inden-l-yl)-2-hydroxyethanesulfonamide was eluted in MeOH, 96.2% ee, tR = 12.58 min (Chiral Method 2).
[0548] (5)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl)-2-hydroxyethanesulfonamide 45 was prepared in an analogous fashion using (5)- methyl 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl) -2,3-dihydro-lH- inden-l-yl) sulfamoyl) acetate. Chiral HPLC: 97.6% ee, tR for 5-enantiomer = 10.99 min (Chiral Method 2).
[0549] (S)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thia∑ol-5-yl)-2,3-dihydro-lH-inden-l- -2-hydroxyethanesulfonamide (Compound 165)
Figure imgf000133_0001
[0550] Prepared using General Procedure 17: To a stirred solution of (5)-methyl 2- (N- (4-(2-(3 -cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3 -dihydro- 1 H-inden- 1 - yl)sulfamoyl)acetate (20 mg, 0.04 mmol) in THF (0.5 mL) was added sodium borohydride (3.6 mg, 0.09 mmol) at room temperature. The reaction was heated to 75°C and methanol (0.06 mL, 1.4 mmol) was added dropwise. After 1 h, the reaction was cooled and concentrated. Purification of the crude material by preparative HPLC gave 12.2 mg (64%) of (S)-N-(4-(2-(3-cyano-4-isopropoxyphenyl) thiazol-5-yl)-2,3 -dihydro- 1 H-inden- l-yl)-2- hydroxyethanesulfonamide 165. LCMS-ESI (m/z) calculated for C24H25N304S2: 483.1; found 484.2 [M+H]+, tR = 3.45 min.
[0551] (i?)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)miazol-5-yl)-2,3-dihydro-lH-inden-l- yl)-2 -hydroxyethanesulfonamide 164 was prepared in an analogous fashion using (i?)-methyl 2-(N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3 -dihydro- 1 H-inden- 1 - yl)sulfamoyl)acetate.
[0552] General Procedure 18. Preparation oflndane Sulfamides
[0553] To a stirred solution of (R)- or (<S)-indane amine (1 eq) in 1,4-dioxane (0.06M) was added sulfamide (5 eq) and the reaction was stirred at 90°C for 16 h. The solvent was evaporated and the reaction mixture was purified by preparative HPLC.
[0554] Compounds 47, 48, 166, and 167 were prepared using General Procedure 18. [0555] (S)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3-dihydro- ind -1 -yl)sulfamide (Compound 47)
Figure imgf000134_0001
[0556] Prepared using General Procedure 18: To (-S)-5-(5-(l-amino-2,3-dihydro-lH- inden-4-yl)-l,3,4-tWadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 4 (25 mg, 0.06 mmol) in dioxane (1 mL) was added sulfamide (30 mg, 0.3 mmol) and the mixture was heated to 90°C. After 16 h, the solvent was evaporated and the residue was purified by column chromatography. Additional purification by recrystallization from MeOH provided 15.9 mg (26%) of (5)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3- dihydro-lH-inden-l-yl) sulfamide 47. LCMS-ESI (m/z) calculated for C21H21N503S2: 455.1; found 456.1 [M+H]+, tR = 3.33 min. Ή NMR (400 MHz, DMSO) δ 8.40 (d, J= 2.3 Hz, 1H), 8.32 (dd, J = 8.9, 2.4 Hz, 1H), 7.88 (d, J = 7.7 Hz, 1H), 7.66 (d, J= 7.6 Hz, 1H), 7.56 - 7.38 (m, 2H), 7.23 (d, J= 9.0 Hz, 1H), 6.75 (s, 2H), 4.95 (dt, J = 12.2, 6.1 Hz, 1H), 4.87 (dd, J = 16.6, 8.2 Hz, 1H), 3.42 - 3.26 (m, 1H), 3.07 (dt, J = 16.4, 8.3 Hz, 1H), 2.61 (dtd, J = 11.0, 7.9, 3.0 Hz, 1H), 2.00 (dq, J = 12.7, 8.8 Hz, 1H), 1.38 (d, J = 6.0 Hz, 6H). 13C NMR (101 MHz, DMSO) 6 166.64, 165.62, 161.19, 146.08, 141.36, 133.89, 133.15, 127.97, 127.51, 127.27, 125.78, 122.22, 115.57, 114.95, 102.29, 72.17, 57.67, 33.41, 30.73, 21.52. Chiral HPLC: (5)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l ,3,4-thiadiazol-2-yl)-2,3-dihydro- 1 H- inden-l-yl) sulfamide was eluted in MeOH: 98.6% ee, ¾ = 7.63 min (Chiral Method 2).
[0557] (7?)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro-lH- inden-l-yl) sulfamide 48 was prepared in an analogous fashion using (i?)-5-(5-(l-arnino-2,3- dihydro- 1 H-inden-4-yl)- 1 ,3 ,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride. Chiral HPLC: 98% ee, tR for tf-enantiomer - 9.10 min (Chiral Method 2).
[0558] (R)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-l H-inden- 1 - yl)sulfamide (Compound 166)
Figure imgf000134_0002
[0559] Prepared using General Procedure 18: To (i?)-5-(5-(l-amino-2,3-dihydro-lH- inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 72 (100 mg, 0.02 mmol) in dioxane (1 mL) was added DIEA (58 mg, 0.32 mmol) and sulfamide (115 mg, 1.2 mmol) and the reaction was heated to 90°C for 4 h. The solvent was evaporated and the residue was diluted with EA (10 mL) and washed with successively with NH4CI and brine. The product was purified by column chromatography (MeOH/DCM) to yield 80 mg (73%) of (i?)-N-(4- (2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- 1 -yl)sulfamide 166. LCMS-ESI (m/z) calculated for C22H22N403S2: 454.1; found 455.4 [M+H]+, tR = 3.46 min. Ή NMR (400 MHz, DMSO) δ 8.29 (d, J - 2.3 Hz, 1H), 8.23 (dd, J = 8.9, 2.4 Hz, 1H), 8.16 (s, 1H), 7.55 (d, J = 7.6 Hz, 1H), 7.47 (dd, J - 18.4, 8.3 Hz, 2H), 7.36 (t, J = 7.6 Hz, 1H), 7.17 (d, J = 9.0 Hz, 1H), 6.73 (s, 2H), 4.98 - 4.75 (m, 2H), 3.19 - 3.05 (m, 1H), 3.00 (dd, J = 16.3, 8.0 Hz, 1H), 2.61 - 2.54 (m, 1H), 2.04 - 1.89 (m, 1H), 1.38 (t, J = 5.5 Hz, 6H). 13C NMR (101 MHz, CDC13) δ 165.43, 161.42, 144.43, 141.31, 140.70, 138.02, 132.43, 132.20, 128.52, 128.18, 128.08, 126.65, 124.98, 116.30, 114.25, 103.78, 72.82, 59.25, 34.62, 31.13, 22.14. Chiral HPLC: (JR)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- 1 H-inden- l-yl)sulfamide was eluted in 50% ethanol in hexanes, 99.0% ee, ¾ = 40.47 min.
[0560] (5)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl) -2,3-dihydro-lH -inden-1- yl) sulfamide 167 was prepared in an analogous fashion using (iS)-5-(5-(l-amino-2,3-dihydro- lH-inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile 71. Chiral HPLC: 99.1% ee, tR for S- enantiomer = 27.67 min.
[0561] General Procedure 19. Preparation oflndane Ureas
[0562] To a stirred solution of CDI (1.7 eq) in DCM (0.16M) was added the stirred suspension of (R)- or (5)-indane amine (1 eq) and Et N (3 eq) in DCM (0.16M) and the mixture was stirred for 2 h or until all the indane amine consumed. If necessary, additional CDI was added. This solution was added to the appropriate amine and the reaction mixture stirred at room temperature for 16 h. The solvent was evaporated and the pure product isolated after preparative HPLC.
[0563] Compounds 50 - 67 and 168 - 205 were prepared using General Procedure 19.
[0564] (R)-N-((R)-4-(5-(3-cyano-4-isopropoxyphenyl)-l, 3, 4-thiadiazol-2-yl)-2, 3-dihydro- -inden-l-yl)-3-(dimethylamino)pyrrolidine-l-carboxamide (Compound 56)
Figure imgf000135_0001
[0565] Prepared using General Procedure 19: To a CDI ((13.4 mg, 0.08 mmol) in DCM (0.5 mL) was added a suspension of (R)-5-(5-(l-amino-2,3-dihydro-lH-inden-4-yl)-l,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 5 (20.0 mg, 0.04 mmol) and Et3N (14.7 mg, 0.14 mmol) in DCM (0.5 mL) and the mixture stirred for 2 h at room temperature. The resulting solution was added to the preparative solution of azetidin-3-ol hydrochloride (15.9 mg, 0.14 mmol)) at room temperature. The reaction was stirred at room temperature for 16 h. The solvent was evaporated and the crude material was purified by preparative HPLC to afford 15 mg of (62%) of (i?)-N-((i?)-4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2- yl)-2,3 -dihydro- 1 H-inden- 1 -yl)-3-(dimethylamino)pyrrolidine- 1 -carboxamide 56. LCMS- ESI (m/z) calculated for C28H32N6(¾S: 516.2; found 517.2 [M+H]+, tR = 2.43min. 1H NMR (400 MHz, DMSO) δ 8.40 (d, J= 2.4 Hz, 1H), 8.32 (dd, J= 9.0, 2.4 Hz, 1H), 7.99 - 7.76 (m, 1H), 7.51 (d, J= 9.2 Hz, 1H), 7.49 - 7.34 (m, 2H), 6.73 (d, J = 8.4 Hz, 1H), 5.32 (d, J= 8.2 Hz, 1H), 5.09 - 4.80 (m, 1H), 3.86 (dd, J= 14.3, 7.0 Hz, 1H), 3.75 (dd, J= 11.0, 7.5 Hz, 1H), 3.63 - 3.48 (m, 1H), 3.45 - 3.22 (m, 3H), 3.10 (dt, J= 16.5, 8.3 Hz, 1H), 2.82 (t, J- 5.1 Hz, 6H), 2.56 - 2.40 (m, 1H), 2.32 (dd, J = 9.8, 2.5 Hz, 1H), 2.15 - 2.02 (m, 1H), 2.00 - 1.81 (m, 1H), 1.38 (d, J= 6.0 Hz, 6H). 13C NMR (101 MHz, CDC13) δ 167.10, 165.84, 161.80, 145.94, 142.30, 133.54, 133.36, 128.86, 127.82, 126.94, 126.41, 122.72, 115.78, 114.01, 103.72, 72.71, 64.71, 55.95, 46.76, 44.20, 42.04, 34.06, 31.45, 27.30, 21.90, 21.89.
[0566] (5 N-((i?)-4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- l-yl)-3-(dimethylamino)pyrrolidine-l -carboxamide 57 was prepared in an analogous fashion using (5)-5-(5-(l-amino-2,3-dihydro-lH-inden-4-yl) -l,3,4-thiadiazol-2- yl)-2 -isopropoxybenzonitrile hydrochloride.
[0567] (R)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-2,3-dihydro- 1 H-inden- l-yl)morpholine-4-carboxamide (compound 58)
Figure imgf000136_0001
[0568] Prepared using General Procedure 19. LCMS-ESI (m/z) calculated for C26H27N503S: 489.2; found 490.2 [M+H]+, tR = 3.54 min. Ή NMR (400 MHz, CDC13) δ 8.20 (dd, J= 8.9, 2.3 Hz, 1H), 8.13 (d, J = 2.2 Hz, 1H), 7.82 (d, J= 7.7 Hz, 1H), 7.49 (d, J= 7.5 Hz, 1H), 7.36 (t, J - 7.6 Hz, 1H), 7.08 (d, J = 9.0 Hz, 1H), 5.51 (d, J = 7.6 Hz, 1H), 4.83 - 4.56 (m, 2H), 3.71 (dd, J= 10.0, 5.0 Hz, 4H), 3.54 - 3.33 (m, 5H), 3.29 - 3.05 (m, 1H), 2.81 - 2.56 (m, 1H), 1.91 (ddd, J = 16.4, 13.1, 7.9 Hz, 1H), 1.46 (d, J = 6.1 Hz, 6H). 13C NMR (101 MHz, CDC13) δ 167.45, 166.17, 162.16, 157.98, 146.58, 142.87, 133.90, 133.77, 129.34, 128.20, 127.29, 126.95, 123.21, 116.06, 114.36, 104.22, 73.03, 66.92, 56.41, 44.54, 34.72, 31.85, 22.25. [0569] (R)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l- yl)pyrrolidine-l-carboxamide (Compound 172)
Figure imgf000137_0001
[05701 Prepared using General Procedure 19: To CDI (117 mg, 0.72 mmol) in DCM (1 mL) was added a suspension of ( ?)-5-(5-(l-amino-2,3-dmydro-lH-inden-4-yl)thiazol-2-yl)- 2-isopropoxybenzonitrile hydrochloride 72 (150 mg, 0.36 mmol), Et3N (145 mg, 1.44 mmol) and DCM (1 mL) and the mixture stirred for 2 h at room temperature. The resulting solution was added to the preparative solution of pyrrolidine (77 mg, 1.08 mmol)) at room temperature. The reaction was stirred at room temperature for 16 h. The solvent was evaporated and the crude material was purified by preparative HPLC to afford 110 mg of (78%) of (i?)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l- yl)pyrrolidine-l-carboxamide 172. LCMS-ESI (m/z) calculated for C27H28N402S: 472.1; found 473.2 [M+H]+, tR = 3.79 min. Ή NMR (400 MHz, DMSO) δ 8.28 (d, J = 2.3 Hz, 1H), 8.22 (dd, J= 8.9, 2.4 Hz, 1H), 8.15 (s, 1H), 7.53 (d, J= 7.3 Hz, 1H), 7.44 (d, J= 9.1 Hz, 1H), 7.36 - 7.24 (m, 2H), 6.42 (d, J= 8.6 Hz, 1H), 5.29 (q, J= 8.4 Hz, 1H), 4.91 (hept, J= 5.9 Hz, 1H), 3.31 - 3.20 (m, 4H), 3.17 - 2.95 (m, 2H), 2.43 (ddd, J = 10.7, 6.2, 2.8 Hz, 1H), 2.00 - 1.87 (m, 1H), 1.87 - 1.72 (m, 4H), 1.37 (d, J = 6.0 Hz, 6H); 13C NMR (101 MHz, CDC13) δ 164.79, 160.90, 156.46, 146.16, 141.10, 140.75, 137.75, 131.89, 131.80, 127.75, 127.71, 127.63, 126.53, 124.27, 115.92, 1 13.78, 103.60, 72.34, 55.78, 45.61, 34.87, 30.80, 25.57, 21.81.
[0571] (5)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro- lH-inden-1- yl)pyrrolidine-l-carboxamide 173 was prepared in an analogous fashion using (5)-5-(5-(l- ammo-2,3-dmydro-lH-inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 71.
[0572] (R)-N-(4-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-2,3-dihydro-lH-inden-l- yl)morpholine-4-carboxamide (Compound 186)
Figure imgf000137_0002
[0573] Prepared using General Procedure 19. LCMS-ESI (m/z) calculated for
C27H2gN403S: 488.2; found 489.2 [M+H]+, tR = 3.54 min. 1H NMR (400 MHz, DMSO) δ 8.28 (d, J= 2.3 Hz, 1H), 8.22 (dd, J = 8.9, 2.4 Hz, 1H), 8.16 (s, 1H), 7.54 (d, J= 7.5 Hz, 1H), 7.44 (d, /= 9.1 Hz, 1H), 7.36 - 7.24 (m, 2H), 6.89 (d, J - 8.3 Hz, 1H), 5.30 (d, J = 8.2 Hz, 1H), 4.99 - 4.83 (m, 1H), 3.61 - 3.50 (m, 4H), 3.42 - 3.24 (m, 4H), 3.23 - 2.91 (m, 2H), 2.48 - 2.40 (m, 1H), 2.00 - 1.82 (m, 1H), 1.37 (d, J= 6.0 Hz, 6H). 13C NMR (101 MHz, CDC13) δ 165.23, 161.35, 157.93, 146.06, 141.53, 141.16, 138.11, 132.35, 132.20, 128.26, 128.14, 126.90, 124.68, 116.38, 114.20, 103.97, 72.80, 66.91, 56.53, 44.50, 34.89, 31.31, 22.26.
[0574] General Procedure 20. Preparation of Indane Amines from Indanols
[0575] To a flask containing the indanol (1 eq) in DCM (0.14M) at 0°C was added SOCl2 (2 eq). After stirring for 30 min, the reaction mixture was concentrated in vacuo and placed under high vacuum for 2 h. The resulting crude chloride was dissolved in DMA (0.02M). The amine (3 eq), DIEA (3 eq), and in some cases NaBr (3 eq) were added and the resulting reactions were stirred at 55-60°C overnight and purified either by preparative HPLC or column chromatography.
[0576] Compounds 206 - 219 and were prepared using General Procedure 20.
[0577] 5-(5-(l-(3-hydroxyazetidin-l-yl)-2,3-dihydro-lH-inden-4-yl)thiazol-2-yl)-2- isopropoxybenzonitrile (Compound 207)
Figure imgf000138_0001
[0578] Prepared using General Procedure 20: To a stirred solution of 5-(5-(l-hydroxy- 2,3-dmydro-lH-inden-4-yl)thiazol-2-yl)-2-isopropoxyberizonitrile (20 mg, 0.05 mmol) in DCM (1 mL) was added thionyl chloride (12.6 mg, 0.106 mmol ) at 0°C. The reaction was stirred at room temperature for 3 h. The solvent was evaporated and the crude chloride re- dissolved in dimethyl acetamide (1 mL). Diisopropyl ethylamine (20.5 mg, 0.16 mmol) and ethanolamine (9.7 mg, 0.16 mmol) were added and the reaction mixture was stirred at 70°C overnight. The reaction mixture was quenched with water (200 uL) and purified by preparative HPLC to afford 11 mg (46%) of 5-(5-(l-(3-hydroxyazetidin-l-yl)-2,3-dihydro- lH-inden-4-yl)thiazol-2-yl)-2-isopropoxybenzonitrile 208. LCMS-ESI (m/z) calculated for C25H25N302S: 431.1; found 432.1 [M+H]+, rR = 6.48 min (Method 2).
[0579] 2-fluoro-5- thiazol-5-yl)benzonitrile (THZ ΓΝΤ-3)
Figure imgf000138_0002
[0580] To 5-(tributylstannyl)thiazole (1.00 g, 2.7 mmol) in THF (10 mL) was added 2- fluoro-5-iodobenzonitrile (0.791 g, 3.2 mmol). The solution was degassed with N2 and bis(triphenylphosphine)palladium(II) chloride Pd(Ph)2Cl2 (0.187 g, 0.27 mmol) was added. The solution was further degassed for five minutes before heating to 85°C for 2 h. Upon cooling, the reaction mixture was diluted with saturated NaHC03 and washed with EA (3 x 50 mL). The combined organic layers were dried over MgSC^, filtered, and concentrated. The crude product was purified by chromatography (10% EA/Hexanes) to afford 0.450 g (82%) of 2-fluoro-5-(trnazol-5-yl)benzonitrile THZ ΓΝΤ-3 as a tan solid. LCMS-ESI (m/z) calculated for Ci0H5FN2S: 204.2; found 205.0 [M+H]+, ¾ = 3.00 min.
[05811 5-(2-bromothia∑ol-5- l -2- uoroben∑onitrile THZ INT-4
Figure imgf000139_0001
[0582] To a stirring solution of 2-fluoro-5-(thiazol-5-yl)benzonitrile THZ INT-3 (0.429 g , 2.1 mmol) in acetic acid (10.5 mL) was added potassium acetate (0.412 g, 4.2 mmol). Bromine (0.647 mL, 12.6 mmol) was added dropwise over 10 minutes and the reaction mixture stirred at room temperature for 48 h. The reaction mixture was basified with IN NaOH and washed with EA and brine. The combined organic layers were dried over MgSC^ filtered, and concentrated. The crude product was purified by chromatography (20% EA/Hexanes) to produce 0.10 g (30%) of 5-(2-bromothiazol-5-yl)-2-fluorobenzonitrile THZ INT-4. LCMS-ESI (m z) calculated for C10H4BrFN2S: 283.1; found 284.9 [M+H]+, tR = 3.33 min.
[0583] 5-(2-(l-(tert-butyldimethylsilyloxy)-2, 3-dihydro-lH-inden-4-yl)thiazol-5-yl)-2- fluorobenzonitrile
Figure imgf000139_0002
[0584] Prepared using General Procedure 1. To 5-(2-bromothiazol-5-yl)-2- fluorobenzonitrile THZ INT-4 (0.100 g, 0.35 mmol), tert-butyldimethyl(4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3-dihydro-lH-inden -1-yloxy) silane IND ΓΝΤ-8 (0.143 g, 0.38 mmol) and sodium carbonate (0.1 12 g, 1.1 mmol) in dioxane (1.8 mL) and H20 (0.2 mL) was added tetrakis(triphenylphosphine)palladium (0.041 g, 0.035 mmol). The solution was degassed with N2 and the reaction mixture heated at 85°C for 6 h. Upon cooling, the reaction mixture was diluted with brine and washed with DCM (3 x 100 mL). The combined organic layers were dried over MgS04i filtered, and concentrated. The crude product was purified by chromatography (30% EA/Hexanes) to produce 0.05 g (32%) of 5- (2-(l-(te^butyldimemylsilyloxy)-2,3-dihydro-lH-inden-4-yl)thiazol-5-yl)-2-fluorobenzo- nitrile as a white solid. LCMS-ESI (m/z) calculated for C25H27FN2OSSi: 450.6; found 451.1 [M+H]+, tR = 4.84 min (Method 3).
[0585] 5-(2-(l-(tert-butyldimethylsilyloxy)-2,3-dihydro-lH nden-4-yl)thiazo
isopropoxy-benzonitrile
Figure imgf000140_0001
[0586J Prepared using General Procedure 2. To a solution of 5-(2-(\-(tert- butyldimethylsilyloxy)-2,3-dihydro-lH-inden-4-yl)thiazol-5-yl)-2-fluoroberizonitrile (0.043 g, 0.095 mmol) in isopropanol (2 mL) was added sodium isopropoxide (0.07 g, 0.090 mmol). The reaction mixture was heated at 60°C for 12 h. Upon cooling, the solvent was removed under a stream of N2 and the crude reaction mixture was carried onto the next step without further purification. LCMS-ESI (m/z) calculated for C2gH34N202SSi: 490.7, t = 5.06 min (Method 3).
[0587] 5-(2-(l-hydroxy-2,3-dihydro-lH-inden-4-yl)thiazol-5-yl)-2-isopropoxy- benzonitrile (Compound 222)
Figure imgf000140_0002
[0588] Prepared using General Procedure 3. To crude 5-(2-(l-(tert- butyldimethylsilyloxy)-2,3-dmydro-lH-mden-4-yl)tWazol-5-yl)-2-isopropoxyberizom
(0.043 g, 0.095 mmol) was added 4N HCl in dioxane (1.0 mL). The reaction mixture was stirred at room temperature for 2 h. The solvent was concentrated under a stream of N2 and the mixture dissolved in MeOH (1.0 mL). The crude product was purified by preparative HPLC to yield 0.02 g (43%) of 5-(2-(l-hydroxy-2,3-dihydro-lH-inden-4-yl)thiazol-5-yl)-2- isopropoxybenzonitrile 222. LCMS-ESI (m/z): calcd for: C22H20N2O2S: 376.5; found 377.1 [M+H]+, ¾ = 3.31 min.
[0589] 2-isopropoxy-5-(thiophen-2-yl)benzonitrile (THIO INT-1)
Figure imgf000141_0001
[0590] A microwave vial was charged with 5-bromo-2-isopropoxybenzonitrile (200 mg, 0.83 mmol), thiophen-2-ylboronic acid (106.5 mg, 0.83 mmol), potassium carbonate (345.3 mg, 2.49 mmol) and 3:1 mixture of dimethylethylene glycol / H20 (2 mL). The reaction mixture was degassed by bubbling N2 gas through the stirred solution for 10 min. Pd(PPh3)4 (20.4 mg, 0.02 mmol) was added and the solution degassed for additional 2 min. The vial was subjected to microwave irradiation at 100°C for 30 min. The solvent was removed and the residue dissolved in EA (10 mL), washed with brine, and dried over MgS04. The product was purified chromatography (EA / hexanes) to afford 165 mg (82%) of 2-isopropoxy-5- (thiophen-2-yl)benzonitrile THIO INT-1 as colorless oil. LCMS-ESI (m/z) calculated for C14H13NOS: 243.1; found 266.0 [M+Na]+, tR = 3.90 min. Ή NMR (400 MHz, CDC13) 6 7.74 (d, J = 2.3 Hz, 1H), 7.69 (dd, J= 8.8, 2.4 Hz, 1H), 7.28 - 7.23 (m, 1H), 7.19 (dd, J = 3.6, 1.1 Hz, 1H), 7.05 (dd, J = 5.1, 3.6 Hz, 1H), 6.95 (d, J = 8.8 Hz, 1H), 4.65 (dt, J = 12.2, 6.1 Hz, 1H), 1.43 - 1.37 (m, 6H).
[0591] -(5-bromothiophen-2-yl)-2-isopropoxybenzonitrile (THIO ΓΝΤ-2)
Figure imgf000141_0002
[0592] To a solution of 2-isopropoxy-5-(thiophen-2-yl)benzonitrile THIO INT-1 (160 mg, 0.66 mmol) in anhydrous DMF (5 mL) was added freshly crystallized N- bromosuccinimide (118 mg, 0.66 mmol) at 0°C. The reaction mixture was stirred at room temperature for 3h (longer reaction times and use of excess NBS caused dibromination). The reaction mixture was diluted with EA (10 mL), washed with water (2 x 10 mL) and brine, and dried over MgS04. The crude product was purified by silica gel column (EA / hexanes) to provide 126 mg (60 %) of 5-(5-bromothiophen-2-yl)-2-isopropoxybenzonitrile THIO ΓΝΤ-2 as white solid. LCMS-ESI (m/z) calculated for Ci4Hi2BrNOS: 320.9; no M+, tR = 4.26 min. Ή NMR (400 MHz, CDC13) δ 7.60 (d, J = 2.3 Hz, 1H), 7.53 (dd, J = 8.8, 2.4 Hz, 1H), 6.95 (d, J = 3.9 Hz, 1H), 6.89 (t, J = 6.2 Hz, 2H), 4.60 (dt, J = 12.1, 6.1 Hz, 1H), 1.35 (d, J = 6.1 Hz, 6H).
[0593] 5-(5-(l-(tert-butyldimethylsilyloxy)-2, 3-dihydro-lH-inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile
Figure imgf000142_0001
[0594] Prepared from 5-(5-bromothiophen-2-yl)-2-isopropoxybenzonitrile ΤΉΙΟ INT-2 and (±)-((4-bromo-2,3-dihydro-lH-inden-l-yl)oxy)(te^butyl)dimethylsilane IND INT 8 using General Procedure 1. LCMS-ESI (m/z) calculated for C29H35N02SSi: 489.2; no M+ found, tK = 8.10 min (Method 1). Ή NMR (400 MHz, CDC13) δ 7.77 (d, J = 2.3 Hz, 1H), 7.71 (dd, 7 = 8.8, 2.4 Hz, 1H), 7.44 (t, 7 = 4.4 Hz, 1H), 7.27 (d, J= 4.5 Hz, 2H), 7.17 (dd, 7 = 12.5, 3.8 Hz, 2H), 6.96 (d, J= 8.9 Hz, 1H), 5.29 (t, = 7.1 Hz, 1H), 4.78 - 4.55 (m, 1H), 3.21 (ddd, 7 = 15.9, 8.8, 2.8 Hz, 1H), 2.95 (dt, 7 = 16.2, 8.1 Hz, 1H), 2.56 - 2.36 (m, 1H), 1.94 (dd, J= 12.6, 7.3 Hz, 1H), 1.41 (d, J = 6.1 Hz, 6H), 1.01 - 0.86 (m, 9H), 0.17 (d, J = 9.3 Hz, 6H).
[0595] 5-(5-(l-hydroxy-2,3-dihydro-lH-inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile (Compound 223)
Figure imgf000142_0002
[0596] To a stirred solution of 5-(5-(l-(tert-butyldimethylsilyloxy)-2,3-dihydro-lH- inden-4-yl)thiophen-2-yl)-2-isopropoxybenzonitrile (80 mg, 0.16 mmol) in 1,4-dioxane (1 mL) was added 4N HCl solution in 1,4-dioxane (1 mL). The reaction mixture was stirred at room temperature for 2 h. Solvent was evaporated and the crude product was purified by chromatography (EA / hexanes) to afford 26 mg (40%) of 5-(5-(l-hydroxy-2,3-dihydro-lH- inden-4-yl)thiophen-2-yl)-2-isopropoxybenzonitrile 223 as a white solid. LCMS-ESI (m/z) calculated for C23H21N02S: 375.1; found 398.1 [M+Na]+, tR = 3.96 min. 1H NMR (400 MHz, CDC13) 6 7.77 (d, 7 = 2.3 Hz, 1H), 7.71 (dd, 7 = 8.8, 2.4 Hz, 1H), 7.53 - 7.45 (m, 1H), 7.38 (d, 7 = 7.4 Hz, 1H), 7.30 (t, 7 = 7.5 Hz, 1H), 7.19 (q, 7 = 3.8 Hz, 2H), 6.97 (d, 7 = 8.9 Hz, 1H), 5.28 (t, 7= 6.1 Hz, 1H), 4.66 (dt, 7= 12.2, 6.1 Hz, 1H), 3.28 (ddd, 7= 16.2, 8.5, 4.7 Hz, 1H), 3.11 - 2.91 (m, 1H), 2.53 (dddd, 7 = 13.1, 8.2, 6.9, 4.7 Hz, 1H), 2.08 - 1.92 (m, 1H), 1.57 (s, 1H), 1.41 (d, 7= 6.1 Hz, 6H). 13C NMR (101 MHz, CDC13) δ 159.37, 146.65, 142.74, 141.52, 140.36, 131.55, 131.08, 130.96, 130.79, 127.79, 127.54, 126.67, 123.82, 116.57, 114.40, 103.82, 76.59, 72.43, 36.07, 30.88, 22.08.
[0597] 5-(5-(l-(2-hydroxyethyla ino)-2,3-dihydro-lH-inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile (Compound 224)
Figure imgf000143_0001
[0598] Prepared using General Procedure 20 from 5-(5-(l-hydroxy-2,3-dihydro-lH- inden-4-yl)thiophen-2-yl)-2-isopropoxybenzonitrile and ethanolamine. LCMS-ESI (m/z) calculated for C25H26N202S: 418.2; found 419.1 [M+H]+, tR = 2.73 min.
[0599] (R)-tert-butyl 4-(5-(3-cyano-4-isopropoxyphenyl)thiophen-2-yl)-2,3-dihydro-lH- inden- -ylcarbamate
Figure imgf000143_0002
[0600] A 20 mL microwave vial was charged with (i?)-tert-butyl 4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-2,3-dihydro-lH-inden-l-ylcarbamate IND Γ Τ-18 (44 mg, 0.12 mmol), 5-(5-bromothiophen-2-yl)-2-isopropoxybenzonitrile THIO ΓΝΤ-2 (40 mg, 0.12 mmol), potassium carbonate (51 mg, 0.37 mmol) and a 3:1 mixture of dimethylethylene glycol / H20 (2 mL). The reaction mixture was degassed by bubbling N2 gas through the stirred solution for 10 min. Pd(PPh3)4 (10.1 mg, 0.008 mmol) was added and the solution degassed for an additional 2 min. The vial was subjected to microwave irradiation at 100°C for 30 min. The solvent was removed and the residue dissolved in EA (10 mL), washed with brine, and dried over MgS04. The product was purified by chromatography (EA / hexanes) to afford 15 mg (51%) of (i?)-/er/-butyl 4-(5-(3-cyano-4-isopropoxyphenyl)thiophen-2-yl)-2,3- dihydro-lH-inden-l-ylcarbamate as an off-white solid. LCMS-ESI (m/z) calculated for C28H30N2O3S: 474.2; no M+ found, tR = 4.43 min. Ή NMR (400 MHz, CDC13) δ 7.76 (d, J = 2.3 Hz, 1H), 7.70 (dd, J= 8.8, 2.4 Hz, 1H), 7.45 (dd, J= 6.3, 2.2 Hz, 1H), 7.27 (d, J= 6.6 Hz, 2H), 7.17 (dd, J= 1 1.9, 3.8 Hz, 2H), 6.97 (d, J= 8.9 Hz, 1H), 5.30 - 5.11 (m, 1H), 4.78 (d, J = 8.6 Hz, 1H), 4.66 (dt, J= 12.2, 6.1 Hz, 1H), 3.18 (ddd, J= 16.1, 8.7, 3.4 Hz, 1H), 3.02 (dt, J - 16.1, 8.1 Hz, 1H), 2.68 - 2.51 (m, 1H), 1.90 - 1.73 (m, 1H), 1.47 (d, J = 8.2 Hz, 9H), 1.41 (d, J = 6.1 Hz, 6H).
[0601] (R)-5-(5-(l-amino-2 -dihydro-lH-inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile (Compound 225)
Figure imgf000143_0003
[0602] Prepared using General Procedure 5. To a stirred solution of (K)-tert-butyl 4-(5- (3-cyano-4-isopropoxyphenyl)thiophen-2-yl)-2,3 -dihydro- 1 H-inden- 1 -ylcarbamate ( 15 mg, 0.03 mmol) in 1,4-dioxane (1 mL) was added 4N HCl solution in 1,4-dioxane (0.5 mL). The reaction mixture was stirred at room temperature for 16 h. Solvent was evaporated the resulting solid was dissolved 1 :1 DMSO:MeOH (1 mL) and purified by preparative HPLC to afford 10 mg (90%) of (JR)-5-(5-(l-amino-2,3-dihydro-lH-inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile 225 as a white solid. LCMS-ESI (m/z) calculated for C23H22N2OS: 374.2; found 358.1. [M-NH2]+, tR - 2.69 min.
[0603] 5-(4-bromothiophen-2-yl)-2-isopropoxybenzonitrile (THIO ΓΝΤ-3)
Figure imgf000144_0001
[0604] A 2 mL microwave vial was charged with 2,4-dibromothiophene (20 mg, 0.08 mmol), (3-cyano-4-isopropoxyphenyl)boronic acid (17 mg, 0.08 mmol), potassium carbonate (35 mg, 0.25 mmol) and 3:1 mixture of DME/H20 (4 mL). The reaction mixture was degassed by bubbling N2 through the stirred solution for 10 min. Pd(PPh3)4 (7 mg, 0.006 mmol) was added and the solution degassed for an additional 2 min. The vial was subjected to microwave irradiation at 70°C for 30 min or until starting material consumed. 5-(4- bromothiophen-2-yl)-2-isopropoxybenzonitrile THIO ΓΝΤ-3 was used in the next experiment without purification. LCMS-ESI (m/z) calculated for C14H12BrNOS: 320.9; no M+ observed, ¾ = 4.15 min.
[0605] 5-(4-(l-((tert-butyldimethylsilyl)oxy)-2,3-dihydro-lH-inden-4-yl)thiophe isopropoxybenzonitrile
Figure imgf000144_0002
[0606] Prepared from 5-(4-bromothiophen-2-yl)-2-isopropoxybenzonitrile THIO ΓΝΤ-3 (0.08 mmol) and tert-butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3- dihydro-1 H-inden- l-yl)oxy)silane IND INT-8 (31 mg, 0.08 mmol) using General Procedure 1, to afford 12 mg (30%, for two steps) of 5-(4-(l-((tert-butyldimethylsilyl)oxy)-2,3-dihydro- lH-inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile. LCMS-ESI (m/z) calculated for C29H35N02SSi: 489.2; no M+ found, tR = 6.66 min (Method 1). Ή NMR (400 MHz, CDC13) δ 7.76 (d, J - 2.3 Hz, 1H), 7.70 (dd, J = 8.8, 2.4 Hz, 1H), 7.36 - 7.31 (m, 2H), 7.29 - 7.25 (m, 2H), 7.25 - 7.19 (m, 1H), 6.96 (d, J = 8.9 Hz, 1H), 5.28 (t, J = 6.9 Hz, 1H), 4.73 - 4.50 (m, 1H), 3.09 (ddd, J= 15.8, 8.7, 2.9 Hz, 1H), 2.88 (dt, J= 16.0, 8.1 Hz, 1H), 2.47 - 2.30 (m, 1 H), 1.96 - 1.81 (m, 1 H), 1.40 (d, J = 6.1 Hz, 6H), 0.94 (s, 9H), 0.16 (d, J = 9.9 Hz, 6H).
[0607] 5-(4-(l -hydroxy-2, 3-dihydro-lH-inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile (Compound 226)
Figure imgf000145_0001
[0608] Prepared using General Procedure 3. To a solution of 5-(4-(l-((tert- butyldimethylsilyl)oxy)-2,3 -dihydro- 1 H-inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile (17 mg, 0.03 mmol) in THF (1 mL) was added 1M solution of TBAF in tetrahydrofuran (0.3 mL, 0.3 mmol) and the reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated and purified by preparative HPLC to yield 8 mg (46%) of 5-(4-(l-hydroxy-2,3-dihydro-lH-mden-4-yl)tWophen-2-yl)-2-isopropoxybenzonitrile 226 as white solid. LCMS-ESI (m/z) calculated for C23H21N02S: 375.1; found 398.1 [M+Na]+. tR = 3.84 min; Ή NMR (400 MHz, CDC13) δ 7.78 (d, J = 2.3 Hz, 1H), 7.72 (dd, J= 8.8, 2.4 Hz, 1H), 7.43 - 7.36 (m, 2H), 7.35 - 7.28 (m, 1H), 7.26 (d, J= 1.4 Hz, 1H), 7.24 (s, 1H), 7.06 - 6.89 (m, 1H), 5.29 (t, J = 6.1 Hz, 1H), 4.77 - 4.49 (m, 1H), 3.20 (ddd, J= 16.0, 8.4, 4.7 Hz, 1H), 3.01 - 2.86 (m, 1H), 2.50 (dddd, J = 13.0, 8.1, 6.8, 4.7 Hz, 1H), 2.11 - 1.88 (m, 1H), 1.58 (s, 1H), 1.41 (d, 7= 6.1 Hz, 6H); 13C NMR (101 MHz, CDC13) δ 159.51, 146.35, 142.55, 142.00, 140.88, 133.16, 131.79, 131.24, 128.14, 127.69, 127.56, 124.16, 123.54, 122.06, 116.54, 114.39, 103.84, 76.68, 72.45, 36.28, 30.50, 22.08.
[0609] ((4-(4-bromothiophen-2-yl)-2,3-dihydro-lH-inden-l-yl)oxy)(tert- butyl)dimethyls -4)
Figure imgf000145_0002
[0610] Prepared using General Procedure 1. A 2 mL microwave vial was charged with 2,4-dibromothiophene (15 mg, 0.06 mmol), tert-butyldimethyl((4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-2,3-dihydro-lH-inden-l-yl)oxy)silane IND INT-8 (23 mg, 0.06 mmol), potassium carbonate (26 mg, 0.18 mmol) and 3:1 mixture of DME/H20 (2 mL). The reaction mixture was degassed by bubbling N2 through the stirred solution for 10 min. Pd(PPh3)4 (5 mg, 0.004 mmol) was added and the solution degassed for an additional 2 min. The vial was subjected to microwave irradiation at 70°C for 30 min or until starting material consumed. The resulting ((4-(4-bromothiophen-2-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)oxy)(tert- butyl)dimethylsilane THIO INT-4 was carried onto the next experiment without workup and purification. LCMS-ESI (m/z) calculated for C19H25BrOSSi: 408.1; no M+ found, tR = 6.50 min (Method 1).
[0611] 2-isopropoxy-5-(4, 4, 5, 5-tetramethyl-l, 3,2-dioxaborolan-2-yl)benzonitrile
Figure imgf000146_0001
[0612] A suspension of 5-bromo-2-isopropoxybenzonitrile (200 mg, 0.83 mmol), 4,4,4',4,,5,5,5,,5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (233.7 mg, 920 mmol), and potassium acetate (246 mg, 2.5 mmol) in anhydrous 1,4-dioxane (100 mL) was degassed by passing N2 through the solution for 30 min. PdCl2(dppf).CH2Cl2 (136 mg, 0.16 mmol) was added and the reaction mixture was heated at 85°C for 6 h. The solvent was removed under vacuum and the residue was dissolved in EA (100 mL) and filtered through celite. The filtrate was washed with water and brine, dried over MgS04, and purified by chromatography (EA / hexanes) to afford 40 mg (13%) of 2-isopropoxy-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)benzonitrile as white solid. LCMS-ESI (m/z) calculated for C16H22BN03: 287.2; found 288.2 [M+H]+, tR = 4.07 min. 1H NMR (400 MHz, CDC13) δ 7.97 (d, 7= 1.5 Hz, 1H), 7.88 (dd, J= 8.5, 1.7 Hz, 1H), 6.91 (d, J= 8.5 Hz, 1H), 4.67 (dt, J = 12.2, 6.1 Hz, 1H), 1.38 (d, J= 6.1 Hz, 6H), 1.30 (s, 12H).
[0613] 5-(5-(l-((tert-butyldimethylsilyl)oxy)-2 -dihydro-lH nden-4-yl)M
isopropoxybenzonitrile (THIO INT-5)
Figure imgf000147_0001
[0614] To the crude reaction mixture containing ((4-(4-bromothiophen-2-yl)-2,3-dihydro- lH-inden-l-yl)oxy)(tert-butyl)dimethylsilane THIO INT-4 (0.12 mmol) in 3:1 mixture of DME/H20 (4 mL) was added 2-isopropoxy-5-(4,4,5,5-tetramethyl- 1,3,2 -dioxaborolan-2- yl)benzonitrile (17.9 mg, 0.06 mmol) and the solution was degassed for 2 min. Pd(PPh3)4 (7 mg, 0.006 mmol) was added and the reaction mixture degassed for an additional 2 min. The reaction mixture was heated under microwave condition at 100°C for 30 min. The reaction mixture was diluted with EA (10 mL), washed with water and brine, and dried over MgS04. The crude product was purified by silica gel column chromatography (EA / Hexanes) to afford 12 mg (40%, for two steps) of 5-(4-(l-((tert-butyldimethylsilyl)oxy)-2,3-dihydro-lH- inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile THIO ΓΝΤ-5. LCMS-ESI (m/z) calculated for C29H35N02SSi: 489.2; no M+ found, tR = 6.66 min (Method 1).
[0615] 5-(5-(l-hydroxy-2,3-dihydro-lH-inden-4-yl)thiophen-3-yl)-2- isopropoxybenzonitrile (Compound 227)
Figure imgf000147_0002
[0616] Prepared using General Procedure 3. To a solution of 5-(4-(l-((tert- butyldimethylsilyl)oxy)-2,3-dihydro- 1 H-inden-4-yl)thiophen-2-yl)-2- isopropoxybenzonitrile THIO ΓΝΤ-5 (12 mg, 0.02 mmol) in THF (1 mL) was added 1M solution of TBAF in tetrahydrofuran (0.2 mL, 0.2 mmol) and the reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated and purified by preparative HPLC to yield 3 mg (22%) of 5-(5-(l-hydroxy-2,3-dihydro-lH-inden-4-yl)thiophen-3-yl)-2- isopropoxybenzonitrile 227 as white solid. LCMS-ESI (m/z) calculated for C23H21N02S: 375.1; found 398.1 [M+Na]+. tR = 3.85 min; Ή NMR (400 MHz, CDC13) δ 7.77 (dd, J= 6.9, 2.2 Hz, 1H), 7.74 - 7.68 (m, 1H), 7.51 (d, J = 8.0 Hz, 1H), 7.45 - 7.36 (m, 1H), 7.36 - 7.28 (m, 1H), 7.27 - 7.22 (m, 2H), 7.10 - 6.74 (m, 1H), 5.37 - 5.15 (m, 1H), 4.67 (dt, J= 12.2, 6.1 Hz, 1H), 3.90 (ddd, J= 16.2, 8.5, 4.7 Hz, 1H), 3.14 - 2.98 (m, 1H), 2.66 - 2.40 (m, 1H), 2.09 - 1.87 (m, 1H), 1.57 (s, 1H), 1.41 (d, J= 6.1 Hz, 6H). PREPARATION OF COMPOUNDS WITH A TETRAHYDRONAPHTHALENE
MOIETY
General Methods
[0617] Ή NMR (400 MHz) and 13C NMR (100 MHz) were obtained in solution of deuteriochloroform (CDC13) or dimethyl sulfoxide - D6 (DMSO). NMR spectra were processed using Mestrec 5.3.0 and 6.0.1. C NMR peaks that are bracketed are two rotomers of the same carbon. Mass spectra (LCMS) were obtained using an Agilent 1100/6110 HPLC system equipped with a Thompson ODS-A, 100 A, 5 μ (50 X 4.6 mm) column using water with 0.1% formic acid as the mobile phase A, and acetonitrile with 0.1% formic acid as the mobile phase B. The gradient was 20-100% with mobile phase B over 2.5 min then held at 100% for 2.5 mins. The flow rate was 1 mL/min. Unless otherwise indicated, the LCMS data provided uses this method (Method /). Final compounds were checked for purity using Method 2; 5% for 1 min, 5-95% over 9 min, then hold at 95% for 5 min, with a flow rate of 1 mL/min. Enantiomeric excess was determined by integration of peaks that were separated on a Daicel Chiralpak AD-H, 250 x 4.6 mm column at a flow rate of 1 mL/min and an isocratic mobile phase. Pyridine, dichloromethane (DCM), tetrahydrofuran (THF), and toluene used in the procedures were from Aldrich Sure-Seal bottles kept under nitrogen (N2). All reactions were stirred magnetically and temperatures are external reaction temperatures. Chromatographies were carried out using a Combiflash Rf flash purification system (Teledyne Isco) equipped with Redisep (Teledyne Isco) silica gel (Si02) columns. Preparative HPLC purifications were done on Varian ProStar/PrepStar system using water containing 0.05% trifluoroacetic acid as mobile phase A, and acetonitrile with 0.05% trifluoroacetic acid as mobile phase B. The gradient was 10-80% mobile phase B over 12 min, hold at 80% for 2 min, and then return to 10% over 2 min with flow rate of 22 mL/min. Other methods similar to this may have been employed. Fractions were collected using a Varian Prostar fraction collector and were evaporated using a Savant SpeedVac Plus vacuum pump. Compounds with salt-able centers were presumed to be the trifluoroacetic acid (TFA) salt. Microwave heating was performed using a Biotage Initiator microwave reactor equipped with Biotage microwave vessels. The following abbreviations are used: ethyl acetate (EA), triethylamine (TEA), diethyl amine (DEA), jV-hydroxybenzotriazole (HOBt), l-ethyl-3-(3- dimethylaminopropyl) carbodiimide hydrochloride (EDC), isopropanol (ΓΡΑ), NJV- dimethylformamide (DMF), dimethyl acetamide (DMA), l,8-Diazabicyclo[5.4.0]undec-7-ene (DBU), Diphenylphosphoryl azide (DPP A), Di-tert-butyl dicarbonate (Boc20), 1,2- dimethoxyethane (DME), sodium isopropoxide (NaO'Pr), N,N-Diisopropylethylamine (DIEA) acetic acid (AcOH).
Experimental Procedures
[0618] (S)-5-bromo- -tetrahydronaphthalen-l- T-1)
Figure imgf000149_0001
To a stirring solution of bromotetralone (5.0 g, 22.2 mmol) in 5:1 HC02H/NEt3 (50 mL) was added chloro {[15',2-S))-(-)-2-amino-l,2-diphenylethyl](4-toluene sulfonyl) amidi} (p-cymene) ruthenium (II) (0.22g, 0.22 mmol). After stirring for 18 h at 30°C, the reaction mixture was diluted with H20, extracted with EA, and purified by chromatography (EA / hexanes) to provide 4.79g (95%) of (5)-5-bromo-l,2,3,4-tetrahydronaphthalen-l-ol THN INT-1. LCMS-ESI (m/z) calculated for C10HnBrO: 227.1 ; found 211.1 [M-OH]+, tR = 3.33 min. Ή NMR (400 MHz, CDC13) δ 7.48 (d, J - 7.9 Hz, 1H), 7.42 (d, J = 7.7 Hz, 1H), 7.09 (s, 1H), 4.78 (dd, J = 10.3, 5.7 Hz, 1H), 2.85 (dt, J = 17.9, 5.7 Hz, 1H), 2.67 (dt, J - 17.7, 7.0 Hz, 1H), 2.10 - 1.74 (m, 4H), 1.67 (d, J = 6.2 Hz, 1H).
[0619] (i?)-5-bromo-l,2,3,4-tetrahydronaphthalen-l-ol THN INT-2 was prepared in an analogous fashion from bromotetralone using chloro {[li?,2/?))-(-)-2-amino-l,2- diphenylethyl](4-toluene sulfonyl)amidi} (p-cymene) ruthenium (II).
[0620] (R)-l-azido-5-bromo-l, 2, 3, 4-tetrahydronaphthalene (THN INT-3)
Figure imgf000149_0002
To a stirring solution of (5)-5-bromo-l,2,3,4-tetrahydronaphthalen-l-ol THN INT-1 (4.79 g, 21.1 mmol) in anhydrous THF (55 mL) at 0°C was added DPP A (9.09 g, 42.2 mmol) followed by slow, dropwise addition of DBU (3.79 mL, 25.3 mmol). After stirring for 2.5 h at 0°C, the reaction mixture was slowly warmed to room temp over 4 h. The solvent was evaporated and the residue was left under high vacuum overnight, then purified by chromatography (EA / hexanes) to provide 4.7g (89%) of (i?)-l-azido-5-bromo-l,2,3,4- tetrahydronaphthalene THN INT-3. LCMS-ESI (m/z) calculated for C10H10BrN3: 252.1; no m/z observed, tR = 4.29 min. Ή NMR (400 MHz, CDC13) 7.59 (dd, J = 7.6, 1.2 Hz, 1H), 7.55 (dd, J = 7.8, 0.5 Hz, 1H), 7.33 (t, J = 7.7 Hz, 1H), 4.59 (t, J = 4.4 Hz, 1H), 3.06 (dd, J = 14.1, 8.9 Hz, 1H), 2.92 (dd, J = 14.5, 8.8 Hz, 1H), 2.14 - 1.96 (m, 3H), 1.97 - 1.82 (m, 1H).
[0621] (5)-l-azido-5-bromo-l,2,3,4-tetrahydronaphthalene THN INT-4 was prepared in an analogous fashion from (i?)-5-bromo-l,2,3,4-tetrahydronaphthalen-l-ol THN INT-2.
[0622] (R)-tert-butyl (5-bromo-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate (THN INT- 5)
Figure imgf000150_0001
To a stirring solution of (i-J-l-azido-S-bromo-l^^^-tetrahydronaphthalene THN INT-3 (4.7g, 18.6 mmol) in EA (50 mL) were added Boc20 (6.1g, 28 mmol) and Pt02 (0.42g, 1.9 mmol). After stirring for 5 h under an atmosphere of H2, the reaction mixture was filtered through celite, rinsing with EA. The resulting filtrate was concentrated and purified by chromatography (EA / hexanes) to provide 5.63 g (93%) of (jR)-tert-butyl (5-bromo- l,2,3,4-tetrahydronaphthalen-l-yl)carbamate THN INT-5. LCMS-ESI (m/z) calculated for Ci5H20BrNO2: 326.2; no m/z , tR = 4.14 min.Ή NMR (400 MHz, CDC13) 7.45 (d, J = 7.9 Hz, 1H), 7.33 (t, J = 7.3 Hz, 1H), 7.05 (q, J = 7.6 Hz, 1H), 4.82 (dt, J = 30.0, 8.4 Hz, 2H), 2.74 (qt, J = 17.8, 6.4 Hz, 2H), 2.07 - 1.67 (m, 4H), 1.50 (d, J = 20.3 Hz, 9H).
[0623J (5)-tert-butyl (5-bromo-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate THN INT-6 was prepared in an analogous fashion from (5 -l-azido-5-bromo-l,2,3,4- tetrahydronaphthalene THN INT-4.
[0624] (R)-tert-butyl (5 -(4, 4, 5, 5-tetramethyl-l, 3, 2-dioxaborolan-2-yl)-l, 2, 3, 4- tetrahydronaphthalen-l-yl)c -7)
Figure imgf000150_0002
To a stirring solution of (R)-tert-buty\ (5-bromo-l,2,3,4-tetrahydronaphthalen-l- yl)carbamate THN INT-5 (1.0 g, 3.1 mmol) in degassed dioxanes (5 mL) were added OAc (0.90g, 9.2 mmol) and pinacolborane (0.93g, 3.7 mmol). The reaction mixture was further degassed prior to addition of PdCl2(dppf)-CH2Cl2 (0.7 lg, 0.6 mmol). After stirring for 16 h at 80°C, the reaction mixture was diluted with EA, filtered through celite then washed with brine. The resulting organic layer was concentrated and purified by chromatography (EA / hexanes) to provide 0.95g (83%) of (R)-tert-buty\ (5-(4,4,5,5-teframethyl-l,3,2-dioxaborolan- 2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate THN INT-7. LCMS-ESI (m/z) calculated for C2iH32BN04: 373.29; no m/z, tR - 3.66 min. Ή NMR (400 MHz, CDC13) δ 7.67 (dd, J = 7.3, 1.1 Hz, 1H), 7.41 (t, J = 9.8 Hz, 1H), 7.17 (dd, J - 9.3, 5.7 Hz, 1H), 4.92 - 4.69 (m, 2H), 3.10 (dt, J = 17.7, 6.0 Hz, 1H), 2.96 (dt, J = 12.7, 6.2 Hz, 1H), 2.00 (dt, J = 10.9, 8.8 Hz, 1H), 1.91 - 1.67 (m, 3H), 1.49 (d, J - 15.6 Hz, 9H), 1.33 (d, J - 5.1 Hz, 12H).
[0625] (S)-tert-butyl (5- (4,4,5,5- tetramethyl- 1,3,2- dioxaborolan- 2- yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)carbamate THN INT-8 was prepared in an analogous fashion from (^-tert-butyl (5-bromo-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate THN INT-6.
[0626] (R)- tert-butyl (5-(5- (3-cyano-4-fluorophenyl)- 1,3,4- thiadiazol-2-yl)- 1,2,3,4- tetrahydronaphthalen- 1-yl) carbamate
Figure imgf000151_0001
To a stirring solution of {R)-tert-b ty\ (5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate THN INT-7 (0.85g, 2.3 mmol), 5-(5- bromo-l,3,4-tbiadiazol-2-yl)-2-fluorobenzonitrile TDZ INT-3 (0.65g, 2.3 mmol) and K2C03 (0.94 g, 6.8 mmol) in 3:1 DME/H20 (32 ml, degassed) in a sealable flash was added Pd(PPh3)4 (0.26g, 0.23 mmol). The reaction mixture was further degassed, sealed and heated to 90°C. After stirring for 18 h, the solvent was evaporated and the residue diluted with EA and washed with water to provide crude 1.5g of {R)-tert-bu y\ (5-(5-(3-cyano-4- fluorophenyl)- 1 ,3 ,4-thiadiazol-2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl)carbamate, which was used without further purification. LCMS-ESI (m/z) calculated for C24H23FN402S: 450.5; found 451.1 [M+H]+, tR = 3.96 min.
[0627] (S)-tert-butyl (5- (5 -(3- cyano- 4- fluorophenyl)- 1, 3, 4-thiadiazol-2-yl)- 1,2,3 ,4- tetrahydronaphthalen- l-yl)carbamate was prepared in an analogous fashion from (S)-tert- butyl (5-(4,4,5,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 - yl)carbamate THN INT-8.
[0628] (R)-tert-butyl (5 -(2- (3- cyano- 4-fluorophenyl)thiazol-5-yl)- 1,2,3,4- tetrahydronaphthalen- 1-yl) carbamate
Figure imgf000152_0001
To a stirring solution of (R)-fert-butyl (5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate THN INT-7 (79.4 mg, 0.28 mmol), 5-(5- bromothiazol-2-yl)-2-fluorobenzonitrile THZ INT-2 (60.0 mg, .28 mmol) and K2C03 (88.2 mg, 0.63 mmol) in 3:1 DME/H20 in a microwave flask was added Pd(PPli3)4 (17.1 mg, 0.01 mmol). The reaction mixture was heated for 30 min at 90°C in a microwave reactor. The mixture was diluted with EA and washed with water. The organic layer was concentrated and purified by chromagraphy (EA / hexanes) to provide 47 mg (50%) of (R)-tert-butyl (5-(2-(3- cyano-4-fluorophenyl)thiazol-5-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl) carbamate. LCMS- ESI (m/z) calculated for C25H24FN302S: 449.2; no m/z, tR = 4.37 min.
[0629] (S)-tert-buty\ (5-(2-(3-cyano-4-fluorophenyl)thiazol-5-yl)-l ,2,3,4- tetrahydronaphthalen-l-yl)carbamate can be prepared in an analogous fashion from (S)-tert- butyl (5-(4,4,5,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl) carbamate THN INT-8.
[0630] (R)-tert-butyl (5- (5-(3-cyano-4-isopropoxyphenyl)- l,3,4-thiadiazol-2-yl)- 1,2,3,4-tetrahydronaphthalen-l-yl) carbamate
Figure imgf000152_0002
To a stirring solution of crude (R)-tert-bxAy\ (5-(5-(3-cyano-4-fluorophenyl)-l,3,4- thiadiazol-2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate (1.03g 2.3 mmol) in IPA (25 mL) was added NaO'Pr (0.24g, 3.0 mmol). After stirring for 4 h at 60°C, the solvent was evaporated and the residue was purified by chromatography (MeOH/ DCM), then rechromatographed (EA / hexanes) to provide 0.69 g (61% over 2 steps)
Figure imgf000152_0003
(5-(5- (3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2- yl)- 1,2,3,4- tetrahydronaphthalen- 1- yl)carbamate. LCMS-ESI (m/z) calculated for C27H30N4O3S: 490.6; no m/z, ¾ = 4.10 min. Ή NMR (400 MHz, CDC13) 6 8.20 (dt, J = 10.3, 5.2 Hz, 1H), 8.15 (d, J = 2.2 Hz, 1H), 7.60 - 7.48 (m, 2H), 7.36 - 7.26 (m, 1H), 7.09 (d, J = 9.0 Hz, 1H), 4.94 (d, J = 6.2 Hz, 1H), 4.89 - 4.80 (m, 1H), 4.75 (dq, J = 12.1, 6.1 Hz, 1H), 3.08 (dd, J = 17.5, 6.0 Hz, 1H), 3.02 - 2.87 (m, 1H), 2.15 - 1.99 (m, 1H), 1.83 (dd, J = 19.2, 6.3 Hz, 3H), 1.49 (s, 9H), 1.46 (d, J = 6.1 Hz, 6H). 13C NMR (101 MHz, CDC13) δ 166.83, 166.24, 161.45, 155.36, 139.02, 136.57, 133.20, 131.32, 129.94, 128.61, 126.13, 122.54, 115.41, 1 13.76, 103.51, 79.32, 72.36, 48.80, 29.60, 28.27, 28.04, 21.61, 19.68.
[0631] (5)-tert-butyl (5- (5- (3 -cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol-2-yl)- l,2,3,4-tetrahydronaphthalen-l-yl)carbamate was prepared in an analogous fashion from (S)- tert-butyl (5-(5-(3-cyano-4-fluorophenyl)-l,3,4-thiadiazol-2-yl)-l,2,3,4- tetrahydronaphthalen- 1 -yl)carbamate.
[0632] (R)-tert-butyl (5-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-l, 2,3,4- tetrahydronaphthalen-l-yl)carbamate
Figure imgf000153_0001
To a stirring solution of (R)-tert-buty\ (5-(2-(3-cyano-4-fluorophenyl)thiazol-5-yl)- l,2,3,4-tetrahydronaphthalen-l-yl)carbamate (45 mg, 0.03 mmol) in IP A (2 mL) was added NaO'Pr (10.7 mg, , 0.13 mmol). After stirring for 2 h at 62°C, the solvent was evaporated and the residue was purified by chromatography (EA / hexanes) to provide 44 mg (90%) (R)- tert-butyl (5-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-l,2,3,4-tetrahydronaphthalen-l- yl)carbamate. LCMS-ESI (m/z) calculated for C28H31N303S: 489.6; no m/z, tR = 4.38 min. 1H NMR (400 MHz, CDC13) δ 8.15 - 8.11 (m, 1H), 8.12 - 8.04 (m,lH), 7.66 (d, J = 4.8 Hz, 1H), 7.48 - 7.40 (m, 1H), 7.31 - 7.20 (m, 2H), 7.03 (d, J = 8.9 Hz, 1H), 4.99 - 4.78 (m, 2H), 4.82 - 4.67 (m, 1H), 2.90 - 2.56 (m, 2H), 1.77 (d, J = 1 1.4, 5.9 Hz, 4H), 1.48 (s, 9H), 1.43 (t, J = 5.4 Hz, 6H).
[0633] (S)-tert-butyl (5- (2- (3- cyano- 4-isopropoxyphenyl)thiazol- 5-yl) -1,2,3,4- tetrahydronaphthalen-l-yl)carbamate can be prepared in an analogous fashion from (S)-tert- butyl (5- (2- (3- cyano- 4-fluorophenyl) thiazol-5-yl)- 1,2,3 ,4-tetrahydronaphthalen-l- yl)carbamate.
[0634] (R)-5-(5-(5-amino-5, 6, 7, 8-tetrahydronaphthalen-l-yl)thiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride (Compound 228)
Figure imgf000154_0001
To a stirring solution of (R)-tert-buty\ (5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4- trnadiazol-2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate (0.77 g, 1.6 mmol) in MeOH (15 mL) was added a solution of 4N HCl in dioxanes (1.2 mL, 4.7 mmol). After stirring for 3.5 h at 50°C, the solvent was evaporated to provide 0.76 g (1 13%) of (i?)-5-(5-(5-amino- 5,6,7,8- tetrahydronaphthalen-l-yl)- l,3,4-thiadiazol-2-yl)- 2-isopropoxybenzonitrile hydrochloride 228. A portion of 1 was further purified by preparative chromatography. LCMS-ESI (m/z) calculated for C22H22N4OS: 390.5; found 374.1 [M-NH3]+, ¾ = 2.37 min. Ή NMR (400 MHz, DMSO) 8.37 (t, J - 8.1 Hz, 1H), 8.37 - 8.22 (m, 1H), 7.84 (d, J = 7.6 Hz, 1H), 7.70 (dd, J = 7.7, 1.1 Hz, 1H), 7.49 (dd, J - 12.5, 5.2 Hz, 2H), 4.94 (hept, J = 5.9 Hz, 1H), 4.56 (d, J = 4.9 Hz, 1H), 3.00 (dt, J = 17.5, 5.5 Hz, 1H), 2.86 (dt, J = 14.0, 6.1 Hz, 1H), 2.18 - 1.85 (m, 3H), 1.86 - 1.69 (m, 1H), 1.37 (d, J = 6.0 Hz, 6H). 13C NMR (101 MHz, DMSO) δ 166.59, 166.24, 161.28, 136.84, 134.32, 133.99, 133.23, 131.79, 130.92, 129.07, 126.54, 122.12, 115.60, 1 14.98, 102.31, 72.21, 47.98, 27.39, 26.55, 21.55, 17.74. Chiral HPLC(AD-H): (/?)-5-(5-(5-amino-5,6,7,8-tetrahydronaphthalen-l-yl)- 1,3,4- thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride was eluted using 40% EtOH in hexanes with 0.3% DEA: 93.3% ee, tR = 18.36 min.
[0635] (S)-5-(5- (5-amino- 5,6,7,8-tetrahydronaphthalen-l-yl)- 1,3,4-thiadiazol- 2-yl)- 2- isopropoxybenzonitrile hydrochloride 229 was prepared in an analogous fashion from (S)- tert-butyl (5-(5-(3 -cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)- 1 ,2,3 ,4- tetrahydronaphthalen-l-yl)carbamate: 93.1% ee, tR for (S)- enantiomer= 22.78 min.
[0636] (R)- 5- (5- (5- amino- 5,6, 7,8- tetrahydronaphthalen- 1- yl)- 1,3,4- thiadiazol-2- yl)- 2-isopropoxybenzonitrile hydrochloride (Compound 230)
Figure imgf000154_0002
To a stirring solution of (R)-tert-butyl (5-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5- yl)-l,2,3,4-tetrahydronaphthalen-l-yl)carbamate (44 mg, 0.9 mmol) in dioxane (2 mL) was added a solution of 4N HCl in dioxanes (2.25 mL, 9 mmol). After stirring for 2 h at 55°C, the solvent was evaporated, the residue triturated with Et20, then filtered and dried to provide 0.23 mg (96%) (R)-5-(5-(5-amino-5,6,7,8-tetrahydronaphmalen-l-yl)thiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 230. LCMS-ESI (m/z) calculated for C23H23N3OS: 389.5; found 373.1 [M-NH3]+, tR = 2.43 min. Ή NMR (400 MHz, DMSO) 8.27 (d, J = 2.3 Hz, 1H), 8.22 (dd, J = 8.9, 2.4 Hz, 1H), 7.94 (s, 1H), 7.68 (t, J - 8.1 Hz, 1H), 7.46 (ddd, J = 25.6, 12.0, 4.5 Hz, 3H), 4.99 - 4.83 (m, 1H), 4.55 (d, J = 5.2 Hz, 1H), 2.88 - 2.72 (m, 2H), 2.18 - 2.03 (m, 1H), 2.05 - 1.84 (m, 2H), 1.85 - 1.68 (m, 1H), 1.37 (d, J = 6.0 Hz, 6H).
[0637] (5)- 5- (5- (5- amino- 5, 6, 7, 8- tetrahydronaphthalen- 1- yl) thiazol- 2- yl)- 2-iso- propoxybenzonitrile hydrochloride can be prepared in an analogous fashion from (S)-tert- butyl (5- (2- (3- cyano- 4-isopropoxyphenyl) thiazol-5-yl)-l,2,3,4- tetrahydronaphthalen- 1-yl) carbamate.
[0638] General Procedure 21. Preparation of Tetrahydronapthalene Amides via Acid Chlorides
To a stirring solution of either (R)- or (5)-tetrahydronapthalene amine HCl (1 eq) in DCM were added an acid chloride (2 eq) or carbonochloridate (2eq) and NEt3 (2 eq). The reaction mixture was stirred at room temperature for 1 h. The solvent was evaporated and the final compounds were purified by preparative HPLC.
[0639] Compounds 231-244 were prepared using General Procedure 21.
[0640] (R)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen- 1-yl) acetamide (Compound 231)
Figure imgf000155_0001
Prepared using General Procedure 21: To a stirring solution of (i?)-5-(5-(5-amino- 5,6,7,8- tetrahydronaphthalen- 1- yl)- 1, 3, 4- thiadiazol- 2- yl)- 2- isopropoxybenzonitrile hydrochloride 228 (20 mg, 0.05 mmol) in DCM (1 mL) were added acetyl chloride (4 μί, 0.05 mmol) and NEt3 (20 μί, 0.15 mmol). After stirring for 1 h, volatiles were evaporated and the residue was purified by preparative HPLC to provide 8.3 mg (41%) of (J?)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen- 1-yl) acetamide 231. LCMS-ESI (m/z) calculated for C24H24N402S: 432.5; found 433.1 [M+H]+, tR - 3.34 min. Ή NMR (400 MHz, CDC13) 6 8.20 (dd, J = 8.9, 2.3 Hz, 1H), 8.14 (d, J = 2.3 Hz, 1H), 7.53 (dd, J= 6.4, 5.4 Hz, 1H), 7.48 (d, J= 7.7 Hz, 1H), 7.38 - 7.25 (m, 1H), 7.09 (d, J = 9.0 Hz, 1H), 5.82 (d, J = 8.6 Hz, 1H), 5.37 - 5.21 (m, 1H), 4.76 (hept, J = 6.0 Hz, 1H), 3.11 (dt, J= 17.8, 6.0 Hz, 1H), 3.03 - 2.84 (m, 1H), 2.12 - 1.99 (m, 4H), 1.94 - 1.75 (m, 3H), 1.46 (d, J= 6.1 Hz, 6H).
[0641] (S)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)acetamide 232 was prepared in an analogous fashion from (S)-5- (5-(5-amino- 5,6,7,8- tetrahydronaphthalen-l-yl)- l,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 229.
[0642] General Procedure 22. Preparation of Tetrahydronapthalene Amides via Peptide Coupling
To a stirring solution of the appropriate acid (1.5-2 eq) in DMF were added HOBT (2 eq) and EDC (2 eq). After 10 min, either (R)- or (5)-tetrahydronapthalene amine HCl (1 eq) in DMF was added and the reaction mixture was stirred for 18 h at room temperature. The final products were purified by preparative HPLC. Any Boc protecting groups were subsequently removed by using General Procedure 29.
[0643] Compounds 245-260 were prepared using General Procedure 22.
[0644] (R)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen-1 -yl)-3-(dimethylamino)propanamide (Compound 257)
Figure imgf000156_0001
Prepared using General Procedure 2: To a stirring solution of 3- dimethylarninopropioriic acid (4.5 mg, 0.04 mmol) in DMF (0.25 mL) were added HOBT (5.6 mg, 0.05 mmol) and EDC (7.2 mg, 0.05 mmol) and the mixture was stirred for 1 h. A solution of (R)-5-(5-(5-arnino-5,6,7,8-tetrahydronaphthalen- 1 -yl)- 1 ,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 228 (15 mg, 0.04 mmol) in DMF (0.25 mL) was added and the resulting mixture was stirred for 20 h. The mixture was purified by preparative HPLC to provide 10.5 mg (62%) of (i?)-N-(5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4- thiadiazol-2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl)-3 -(dimethylamino)propanamide 257. LCMS-ESI (m/z) calculated for C27H31N502S: 489.6; found 490.2 [M+H]+, ¾ = 2.44 min. 1H NMR (400 MHz, CDC13) δ 8.19 (dt, J = 11.5, 5.8 Hz, 1H), 8.16 (t, J = 6.6 Hz, 1H), 7.53 (t, J = 5.7 Hz, 1H), 7.41 (d, J = 7.5 Hz, 1H), 7.34 - 7.26 (m, 1H), 7.09 (d, J = 9.1 Hz, 1H), 6.90 (d, J = 8.5 Hz, 1H), 5.30 - 5.17 (m, 1H), 4.84 - 4.66 (m, 1H), 3.86 (s, 2H), 3.47 (t, J - 6.7 Hz, 2H), 3.16 - 2.91 (m, 2H), 2.88 (d, J = 14.4 Hz, 7H), 2.05 (dd, J - 14.4, 9.4 Hz, 1H), 1.86 (dd, J = 7.6, 3.9 Hz, 2H), 1.46 (d, J - 6.1 Hz, 6H).
[0645] (5)-N-(5-(5- (3-cyano-4- isopropoxyphenyl)- l,3,4-thiadiazol-2-yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)-3-(dimethylaniino)propanamide 256 was prepared in an analogous fashion from (5 -5-(5-(5-amino-5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 229.
[0646J (S)~ N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)-2-hydroxyacetamide (Compound 246)
Figure imgf000157_0001
Prepared using General Procedure 2: To a stirring solution of glycolic acid (3.9 mg, 0.05 mmol) in DMF (1 mL) were added HOBT (8.6 mg, 0.06 mmol) and EDC (12.1 mg, 0.06 mmol) and the mixture was stirred for 1 h. A solution of (£)-5-(5-(5-amino-5,6,7,8- tetrahydronaphthalen-l-yl)- l,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 229 (20 mg, 0.05 mmol) was added and the resulting mixture was stirred for 20 h. The mixture was purified by preparative HPLC to provide 11.4 mg (54%) of (5)-N-(5-(5-(3- cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl) -2- hydroxyacetamide 246. LCMS-ESI (m/z) calculated for C24H24N403S: 448.5; found 449.1 [M+H]+, tR = 3.14 min. Ή NMR (400 MHz, CDC13) δ 8.21 (dd, J= 8.9, 2.3 Hz, 1H), 8.15 (d, J = 2.3 Hz, 1H), 7.54 (t, J = 8.3 Hz, 1H), 7.46 (t, J = 8.5 Hz, 1H), 7.30 (dd, J= 15.4, 7.7 Hz, 1H), 7.10 (d, J = 9.1 Hz, 1H), 6.78 (d, J = 8.8 Hz, 1H), 5.44 - 5.27 (m, 1H), 4.77 (dp, J = 12.3, 6.0 Hz, 2H), 4.28 - 4.16 (m, 2H), 3.23 - 3.04 (m, 1H), 2.98 (dt, J = 17.7, 6.0 Hz, 1H), 2.20 - 2.00 (m, 2H), 1.89 (dd, 7= 9.6, 6.6 Hz, 2H), 1.47 (d, J= 6.1 Hz, 6H).
[0647] (R)- N- (5- (5- (3- cyano-4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2-yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)-2-hydroxyacetamide 245 was prepared in an analogous fashion from (5 -5-(5-(5-amino-5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 228.
[0648] General Procedure 23. Preparation of Tetrahydronapthalene Alkyl Amines Via Reductive Amination To a stirring solution of the appropriate aldehyde (1.1 eq) in MeOH was added either (R)- or (5)-tetrahydronapthalene amine (free base, 1 eq) and the reaction mixture was stirred for 16 h at room temperature. To the mixture was then added AcOH (1 drop) and NaBFL* (1.5 eq). After 20 min, the reaction mixtures were concentrated and purified by preparative HPLC.
[0649] Compounds 261-273 were prepared using General Procedure 23.
[0650] (R)-2-isopropoxy-5-(5-(5-((pyridin-3-ylmethyl)amino)-5, 6, 7, 8- tetrahydronaphthalen-l-yl)-l, 3, 4-thiadiazol-2-yl)benzonitrile (Compound 263)
Figure imgf000158_0001
Prepared using General Procedure 23: To a stirring solution of 3 -pyridine carboxaldehyde (4.6 mg, 0.05 mmol) in MeOH (0.5 mL) was added (R)-5-(5-(5-amino- 5,6,7,8-tetrahydronaphthalen- 1 -yl)- 1 ,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile 228 (20 mg, 0.05 mmol). After stirring for 16 h, AcOH (1 drop) and NaBH* (3 mg, 0.08 mmol) were added. After an additional 20 minutes of stirring, the mixture was concentrated and purified by preparative HPLC to provide 17.8 mg (73%) of (i-)-2-isopropoxy-5-(5-(5-((pyridin-3- ylmethyl)amino)- 5,6,7,8- tetrahydronaphthalen- 1-yl)- 1,3,4- thiadiazol- 2-yl) benzonitrile 263. LCMS-ESI (m/z) calculated for C28H27N5OS: 481.6; found 374.1 [M-(pyridin-3- ylmethanamine)]+, iR - 2.37 min. 1H NMR (400 MHz, CDC13) δ 9.06 (s, 1H), 8.61 (s, 1H), 8.48 (s, 1H), 8.21 - 8.04 (m, 2H), 7.75 (s, 1H), 7.65 (d, J = 7.0 Hz, 1H), 7.58 (d, J = 7.5 Hz, 1H), 7.31 (dd, J = 15.7, 8.4 Hz, 1H), 7.09 (d, J = 8.9 Hz, 1H), 4.84 - 4.64 (m, 2H), 4.46 (s, 2H), 3.11 (d, J = 18.0 Hz, 1H), 3.04 - 2.86 (m, 1H), 2.32 (s, 1H), 2.20 (s, 1H), 2.06 (s, 1H), 1.84 (s, 1H), 1.51 - 1.39 (m, 6H).
[0651] (.S)-2-isopropoxy-5-(5-(5-((pyrid -3-ylmemyl)amino)- 5,6,7,8- tetrahydronaphthalen- l-yl)-l,3,4-thiadiazol-2-yl)benzonitrile 271 was prepared in an analogous fashion from (5)- 5- (5- (5- amino- 5,6,7,8- tetrahydronaphthalen- 1- yl)- 1,3,4- thiadiazol- 2- yl)- 2-isopropoxybenzonitrile 229.
[0652] (R)- 5- (5- (5- ((2- (dimethylamino) ethyl) amino)- 5,6, 7,8- tetrahydronaphthalen- l-yl)-l,3,4-thiadiazol- 2-yl)-2-isopropoxybenzonitrile (Compound 274)
Figure imgf000159_0001
To a stirring solution of (7?)-5-(5-(5-amino-5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 228 (20 mg, 0.05 mmol) in CH3CN (1 mL) were added K2C03 (19.4 mg, 0.14 mmol) and 2-dimethylaminoethyl bromide (7.0 μL, 0.05 mmol). After stirring for 48 h at 50°C, the mixture was concentrated and purified by preparative HPLC to provide 1.8 mg (8%) of (i?)-5-(5-(5-((2-(dimemylamino)ethyl)amino)- 5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4- thiadiazol-2-yl)- 2-isopropoxybenzonitrile 274. LCMS-ESI (m/z) calculated for C26H31N5OS: 461.6; found 462.2 [M+H]+, tR = 2.27 min.
[0653] (5)-5-(5-(5-((2- (dimemylamino)ethyl)amino)- 5,6,7,8-tetrahydronaphthalen-l-yl)- 1,3,4- thiadiazol-2-yl)- 2-isopropoxybenzonitrile can be prepared in a similar fashion from (5)-5-(5-(5-ammo-5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 229.
[0654] General Procedure 24. Preparation of Tetrahydronapthalene Sulfamides
To a solution of either (R)- or (5)-tetrahydronapthalene amine HC1 (1 eq) in dioxane were added sulfamide (5 eq) and DIE A (3 eq). The reaction mixture was stirred at 100°C for 18 h. The solvent was evaporated and mixture was purified by preparative HPLC.
[0655] Compounds 275-277 and 309-310 were prepared using General Procedure 24.
[0656] (R)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen- 1-yl) sulfamide (Compound 275)
Figure imgf000159_0002
Prepared using General Procedure 24: To a stirring solution (i?)-5-(5-(5-amino- 5,6,7,8-tetrahydronaphthalen-l-yl)- l,3,4-thiadiazol-2-yl)- 2- isopropoxybenzonitrile hydrochloride 228 (25 mg, 0.06 mmol) in dioxane (0.5 mL) were added sulfamide (28 mg, 0.29 mmol) and DIEA (23 μί, 0.18 mmol) and the mixture was heated to 100°C for 18 h. The solvent was evaporated and the residue was purified by preparative HPLC to provide 12.9 mg (46%) of (^)-N-(5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-l,2,3,4- tetrahydronaphthalen-l-yl)sulfamide 275. LCMS-ESI (m/z) calculated for C22H23N503S2: 469.6; found 470.1 [M+H]+, ¾ - 3.31 min. 1H NMR (400 MHz, CDC13) δ 8.20 (dd, J - 8.9, 2.3 Hz, 1H), 8.14 (d, J = 2.2 Hz, 1H), 7.72 (d, J = 7.8 Hz, 1H), 7.57 (d, J - 7.5 Hz, 1H), 7.34 (dd, J = 16.2, 8.5 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 4.85 - 4.69 (m, 2H), 4.65 (d, J - 17.7 Hz, 2H), 4.61 (d, J - 8.0 Hz, 1H), 3.10 (dt, J = 17.7, 5.8 Hz, 1H), 3.03 - 2.88 (m, 1H), 2.24 - 2.02 (m, 2H), 1.95 - 1.83 (m, 2H), 1.51 - 1.39 (m, 6H).
[0657] (5)- N- (5- (5- (3- cyano-4-isopropoxyphenyl)- 1,3,4- thiadiazol-2-yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)sulfamide 276 was prepared in an analogous fashion from (S)-5- (5- (5- amino- 5,6,7,8- tetrahydronaphthalen-l-yl)- 1,3,4- thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 229.
[0658] (R)- N- (5- (2- (3 cyano- 4- isopropoxyphenyl) thiazol- 5-yl) -1,2,3,4- tetrahydrona hthalen-l-yl)sulfamide (Compound 277)
Figure imgf000160_0001
Prepared using General Procedure 24: To a stirring ( iJ-S-iS-iS-amino-S^ ^- tetrahydronaphmalen-l-yl)thiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 230 (13 mg, 0.03 mmol) in dioxane (1 mL) were added sulfamide (14.7 mg, 0.15 mmol) and DIEA (12 μΐ^, 0.09 mmol) and the mixture was heated to 100°C for 18 h. The solvent was evaporated and the residue was purified by preparative HPLC to provide 8 mg (57%) (R)-N-(5-(2-(3- cyano-4-isopropoxyphenyl) thiazol-5-yl)- 1,2,3,4 -tetrahydronaphthalen- 1 -yl)sulfamide 277. LCMS-ESI (m/z) calculated for C23H24N403S2: 468.6; found 469.1 [M+H]+, tR = 3.49 min. 1H NMR (400 MHz, CDC13) δ 8.21 - 7.99 (m, 2H), 7.72 - 7.49 (m, 2H), 7.36 - 7.28 (m, 2H), 7.04 (d, J = 8.9 Hz, 1H), 4.73 (dd, J = 12.1, 6.0 Hz, 5H), 2.93 - 2.57 (m, 2H), 2.21 - 2.04 (m, 2H), 1.93 - 1.79 (m, 2H), 1.45 (d, J = 6.1 Hz, 6H). 13C NMR (101 MHz, CDC13) d 165.67, 160.98, 142.26, 136.89, 136.74, 136.38, 131.96, 131.87, 130.58, 130.55, 130.32, 126.54, 126.40, 115.94, 113.81, 103.60, 72.39, 52.55, 30.02, 28.15, 21.83, 19.16.
[0659] (5)-N-(5-(2-(3-cyano-4-isopropoxyphenyl)thiazol-5-yl)-l ,2,3,4- tetrahydronaphthalen- l-yl)sulfamide can be prepared in a similar fashion from (5)-5-(5-(5- amino-5,6,7,8-tetrahydronaphmalen-l-yl)thiazol-2-yl)-2-isopropoxybenzonitrile
hydrochloride. [0660] General Procedure 25. Preparation of Tetrahydronaphthalene Sulfonamides via Sulfonyl Chlorides
To a solution of either (R)- or (5)-tetrahydronapthalene amine HC1 (1 eq) in DCM (0.05M) were added TEA (3 eq) and the appropriate sulfonyl chloride (1-2 eq.) at room temperature. The reaction mixture was stirred at room temperature for 18 h. The solvent was evaporated and the product isolated after preparative HPLC purification.
[0661] Compounds 278-286, 313 and TDZ THN INT-1 were prepared using General Procedure 25.
[0662] (R)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)methanesulfonamide (Compound 278)
Figure imgf000161_0001
Prepared using General Procedure 25: To a stirring solution of (i?)-5-(5-(5-amino- 5,6,7,8- tetrahydronaphthalen-l-yl)- 1,3,4- thiadiazol-2 -yl)- 2- isopropoxybenzonitrile hydrochloride 228 (20 mg, 0.05 mmol) in DCM (1 mL) at 0°C were added TEA (20 μί, 0.14 mmol) and methanesulfonyl chloride (4.5 μί, 0.06 mmol). The mixture was allowed to warm to room temperature over 2 h. The solvent was evaporated and crude mixture was purified by preparative HPLC to afford 12.5 mg (53%) of (#)-N-(5-(5-(3-cyano-4- isopropoxyphenyl)- 1,3,4- thiadiazol-2-yl)- 1,2,3,4- tetrahydronaphthalen-1- yl)methanesulfonamide 278. LCMS-ESI (m/z) calculated for C23H24N403S2: 468.6; found 469.1 [M+H]+, ¾ = 3.51 min. Ή NMR (400 MHz, CDC13) δ 8.25 - 8.16 (m, 1H), 8.15 (t, J = 6.6 Hz, 1H), 7.68 (d, J = 7.7 Hz, 1H), 7.55 (dt, J = 14.0, 7.0 Hz, 1H), 7.36 (t, J - 7.7 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 4.76 (hept, J = 6.0 Hz, 2H), 4.60 (d, J = 8.5 Hz, 1H), 3.19 - 3.04 (m, 4H), 2.97 (dt, J = 18.0, 6.7 Hz, 1H), 2.16 (ddd, J = 17.4, 8.8, 4.4 Hz, 1H), 2.06 - 1.94 (m, 1H), 1.91 (ddd, J = 22.2, 11.3, 6.2 Hz, 2H), 1.46 (t, J = 5.3 Hz, 6H).
[0663] (S)- N- (5- (5- (3 cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)methanesulfonamide 279 was prepared in an analogous fashion from (5)-5-(5-(5-amino-5,6,7,8-tetrahydronaphthalen- 1 -yl)- 1 ,3, 4- thiadiazol-2 -yl)-2- isopropoxybenzonitrile hydrochloride 229.
[0664] (R)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2-yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)-2-methoxyethanesulfonamide (Compound 280)
Figure imgf000162_0001
Prepared via General Procedure 25. To a stirring solution of (i?)-5-(5-(5-amino- 5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4-tMadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 228 (30 mg, 0.07 mmol) in DCM (1 mL) were added TEA (49 μί, 0.35 mmol) and 2-methoxyethanesulfonyl chloride (33.4 μί, 0.21 mmol). After stirring for 0.5 h, the reaction mixture was concentrated and purified by preparative chromatography to provide 8.5 mg (24%) of (i?)-N-(5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-l,2,3,4- tetrahydronaphthalen-l-yl)-2-methoxyethanesulfonamide 280 LCMS-ESI (m/z) calculated for C25H28N404S2: 512.6; found 513.1 [M+H]+, tR = 3.62 min. Ή NMR (400 MHz, CDC13) δ 8.20 (dt, J = 16.2, 8.1 Hz, 1H), 8.15 (d, J = 2.3 Hz, 1H), 7.75 (d, J = 7.7 Hz, 1H), 7.55 (dt, J = 13.5, 6.8 Hz, 1H), 7.34 (dd, J = 16.5, 8.8 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 4.86 - 4.70 (m, 2H), 4.66 (d, J = 8.2 Hz, 1H), 3.97 - 3.75 (m, 2H), 3.48 - 3.32 (m, 5H), 3.11 (dt, J = 17.8, 6.0 Hz, 1H), 2.97 (dt, J = 17.8, 6.7 Hz, 1H), 2.24 - 2.07 (m, 1H), 2.11 - 1.96 (m, 1H), 1.98 - 1.78 (m, 2H), 1.46 (d, J = 6.1 Hz, 6H).
[0665] (S)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)-2-methoxyethanesulfonamide 281 was prepared in an analogous fashion from (5)-5-(5-(5-amino-5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 229.
[0666] (R)- methyl 2- (N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- -tetrahydronaphthalen-l-yl)sulfamoyl)acetate (TDZ THN INT-1)
Figure imgf000162_0002
Prepared using General Procedure 25: To a stirring solution of (R)-5-(5-(5-amino- 5,6,7,8- tetrahydronaphthalen-l-yl)- 1,3,4- thiadiazol-2-yl)- 2- isopropoxybenzonitrile hydrochloride 228 (0.27 g, 0.63 mmol) in DCM (5 mL) were added TEA (0.19 mL, 1.4 mmol) and methyl-2-(chlorosulfonyl)acetate (0.13 g, 0.76 mmol). Additional TEA and methyl-2-(chlorosulfonyl)acetate were added to drive the reaction to completion over 24 h. The crude reaction mixture was partitioned between DCM and saturated NaHC03. The organic layer was dried over Na2S04, concentrated, and purified by chromatography (EA/hexanes) to give 0.22 g (65%) of (/?)-methyl 2-(N-(5-(5-(3-cyano-4-isopropoxyphenyl)- 1,3,4- thiadiazol- 2-yl)- 1,2,3,4- tetrahydronaphthalen- l-yl)sulfamoyl)acetate TDZ THN EVr-1. LCMS-ESI (m/z) calculated for C25H26N405S2: 526.6; found 527.1 [M+H]+, tR = 3.59 min. 1H NMR (400 MHz, CDC13) δ 8.23 - 8.14 (m, 1H), 8.14 (t, J= 3.3 Hz, 1H), 7.72 (d, J = 7.7 Hz, 1H), 7.54 (dt, J = 9.0, 4.5 Hz, 1H), 7.34 (t, J = 7.7 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 5.22 (t, J = 7.3 Hz, 1H), 4.77 (dp, J = 18.2, 6.2 Hz, 2H), 4.24 - 3.99 (m, 2H), 3.81 (s, 3H), 3.10 (dt, " = 17.7, 6.0 Hz, 1H), 2.96 (dt, J = 17.7, 6.5 Hz, 1H), 2.23 - 1.98 (m, 2H), 1.99 - 1.77 (m, 2H), 1.45 (d, J = 6.1 Hz, 6H). l3C NMR (101 MHz, CDC13) δ 166.74, 166.55, 164.38, 161.69, 137.15, 136.96, 133.45, 133.35, 132.04, 130.80, 129.01, 126.56, 122.62, 115.54, 113.87, 103.74, 72.53, 57.16, 53.17, 52.98, 30.12, 27.79, 21.74, 19.14.
[0667] (5)-methyl 2- (N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol-2-yl)- l,2,3,4-tetrahydronaphthalen-l-yl)sulfamoyl)acetate 284 was prepared in an analogous fashion from (5 -5-(5-(5-amino-5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4-thiadiazol-2-yl)-2- isopropoxybenzonitrile hydrochloride 229.
[0668] General Procedure 26. Preparation of Tetrahydronapthalene Sulfonamide Acids
To a stirring solution of either (R)- or (5)-indane sulfonamide ester (1 eq) in MeOH (0.2 M) was added 6N NaOH (2 eq) at room temperature. The reaction mixture was stirred at room temperature or 50°C for 6 h. The crude reaction mixture was diluted with water, acidified with IN HCl and extracted with DCM and EA. The organic layer was dried over Na2S04, concentrated, and isolated after preparative HPLC purification.
[0669] Compounds 287-288 were prepared using General Procedure 26.
[0670] (R)-2- (N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl) - l, -tetrahydronaphthalen-l-yl)sulfamoyl)acetic acid (Compound 287)
Figure imgf000163_0001
Prepared using General Procedure 26: To a stirring solution of (i?)-methyl 2-(N-(5- (5-(3-cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen-
1- yl) sulfamoyl) acetate TDZ THN INT-1 (0.10 g, 0.19 mmol) in MeOH (3 mL) was added 6N NaOH (5 drops). The reaction mixture was stirred at 50°C for 6 h. The crude reaction mixture was diluted with water, acidified with IN HCl and extracted with DCM and EA. The organic layer was dried over Na2S04 and concentrated, to give 0.094 g (97%) of (i?)-2-(N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen-l-yl) sulfamoyl)acetic acid 287. LCMS-ESI (m/z) calculated for C24H24N405S2: 512.6; found 513.1 [M+H]+, tR = 3.32 min.
[0671] (S)- 2- (N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- l,2,3,4-tetrahydronaphthalen-l-yl)sulfamoyl) acetic acid 288 was prepared in an analogous fashion from (S)- 5- (5- (5-amino- 5,6,7,8- tetrahydronaphthalen-l-yl) -l,3,4-thiadiazol-2-yl)-
2- isopropoxybenzonitrile hydrochloride 284.
[0672] General Procedure 27. Preparation of Tetrahydronapthalene Sulfonamide Alcohols
To a stirring solution of either (R)- or (.S -tetrahydronapthalene sulfonamide ester (1 eq) in THF (0.06 M) was added NaBFL; (2.5eq) at room temperature. The reaction mixture was heated to 70°C and MeOH (1 eq) was added dropwise. After 1 h, the reaction mixture was cooled, concentrated, and purified by preparative HPLC or chromatography.
[0673] Compounds 289-291 and 314 were prepared using General Procedure 27.
[0674] (R)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)-2-hydroxyethanesulfonamide (Compound 289)
Figure imgf000164_0001
Prepared using General Procedure 27: To a stirring solution of (i?)-methyl 2-(N-(5- (5-(3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2-yl)- 1,2,3,4- tetrahydronaphthalen-1- yl)sulfamoyl)acetate TDZ THN INT-1 (0.04 g, 0.08 mmol) in THF (1 mL) was added NaBH* (0.07 g, 0.19 mmol) at room temperature. The reaction mixture was heated to 70°C and MeOH (0.02 mL, 0.05 mmol) was added. After 1 h, the reaction mixture was cooled, concentrated, and purified by chromatography (MeOH / DCM) to give 11.4 mg (22%) of (R)- N-(5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)- 1,2,3,4- tetrahydronaphthalen- l-yl)-2-hydroxyethanesulfonamide 289. LCMS-ESI (m/z) calculated for C24H26N404S2: 498.6; found 499.1 [M+H]+, tR = 3.29 min. Ή NMR (400 MHz, CDC13) 6 8.19 (dt, J = 14.6, 7.3 Hz, 1H), 8.13 (d, J = 2.3 Hz, 1H), 7.70 (d, J = 7.7 Hz, 1H), 7.54 (dd, J = 10.8, 4.1 Hz, 1H), 7.34 (t, J = 7.7 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 4.92 (d, J - 8.5 Hz, 1H), 4.85 - 4.68 (m, 2H), 4.13 (t, J = 5.2 Hz, 2H), 3.45 - 3.29 (m, 2H), 3.09 (dt, J = 17.6, 5.9 Hz, 1H), 2.94 (dt, J = 13.7, 6.6 Hz, 1H), 2.22 - 2.07 (m, 1H), 2.08 - 1.94 (m, 1H), 1.92 (ddt, J = 13.5, 10.6, 5.0 Hz, 2H), 1.46 (d, J = 6.1 Hz, 6H).
[0675] (5)- N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2-yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)-2-hydroxyethanesulfonamide 290 was prepared in an analogous fashion from (5)-methyl 2-(N-(5-(5-(3-cyano-4-isopropoxyphenyl)- l,3,4-thiadiazol-2-yl)- l,2,3,4-tetrahydronaphthalen-l-yl)sulfamoyl)acetate 284. Yield: 7.2 mg (38.3%). LCMS-ESI (m/z) calculated for C24H26N404S2: 498.6; found 499.1 [M+H]+, tR = 3.28 min. 1H NMR (400 MHz, CDC13) 6 8.19 (dd, J= 8.9, 2.3 Hz, 1H), 8.13 (d, J= 2.2 Hz, 1H), 7.70 (d, J = 7.9 Hz, 1H), 7.54 (d, J= 6.7 Hz, 1H), 7.34 (t, J= 7.7 Hz, 1H), 7.09 (d, J= 9.1 Hz, 1H), 5.07 (d, J = 8.4 Hz, 1H), 4.87 - 4.60 (m, 2H), 4.22 - 4.00 (m, 2H), 3.37 (td, J- 4.8, 2.2 Hz, 2H), 3.17 - 2.84 (m, 2H), 2.24 - 2.07 (m, 1H), 2.07 - 1.74 (m, 3H), 1.46 (d, J= 6.1 Hz, 6H).
[0676] General Procedure 28. Preparation of Tetrahydronapthalene Sulfonamide Amides
To a stirring solution of either (R)- or (5)-tetrahydronapthalene sulfonamide acid (1 eq) in DMF (0.25 M) were added EDC and HOBT. After 5 min, the amine was added and the reaction mixture was stirred 18 h at room temperature. The crude reaction mixture was diluted with NaHC03 and extracted with EA. The combined organic layers were dried over Na2S04, and purified by preparative HPLC.
[0677] Compounds 292-295 were prepared using General Procedure 28.
[0678] (R)- 2- (N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1, 3, 4- thiadiazol- 2-yl)- 1, 2, 3, -tetrahydronaphthalen-l-yl)sulfamoyl)-N,N-dimethylacetamide (Compound 292)
Figure imgf000165_0001
Prepared using General Procedure 28: To a stirring solution of (i?)-2-(N-(5-(5-(3- cyano-4-isopropoxyphenyl)-l ,3,4- thiadiazol-2-yl)- 1 ,2,3,4- tetrahydronaphthalen- 1- yl) sulfamoyl) acetic acid 287 (30 mg, 0.06 mmol) in DMF (1 mL) were added HOBT (12 mg, 0.09 mmol) and EDC (17 mg, 0.09 mmol). After 5 min, dimethylamine (40 wt % solution in H20, 66 iL, 0.6 mmol) was added and the reaction mixture was stirred 18 h at room temperature. Additional dimethylamine (40 wt % solution in H20, 100 μί, 0.9 mmol) was added and the reaction mixture was further stirred for 7h. The crude reaction mixture was diluted with saturated NaHC03 and extracted with EA. The combined organic layers were dried over Na2S04 and purified by preparative HPLC to give 8.66 mg (27%) of (R)-2-(N-(5- (5-(3-cyano-4- isopropoxyphenyl)- l ,3,4-thiadiazol-2-yl)- 1,2,3,4-tetrahydronaphthalen-l- yl)sulfamoyl)-N,N-dimethylacetamide 292. LCMS-ESI (m/z) calculated for C26H29N504S2: 539.7; found 540.2 [M+H]+, *R = 3.40 min. 1H NMR (400 MHz, CDC13) δ 8.21 (dd, J = 8.9, 2.3 Hz, 1H), 8.15 (d, J = 2.3 Hz, 1H), 7.82 (d, J = 7.7 Hz, 1H), 7.55 (t, J = 10.5 Hz, 1H), 7.36 (t, J = 7.7 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 5.41 (d, J - 8.1 Hz, 1H), 4.92 - 4.60 (m, 2H), 4.29 (d, J = 14.6 Hz, 1H), 4.07 (d, J = 14.6 Hz, 1H), 3.13 (d, J = 10.5 Hz, 3H), 3.07 (t, J = 6.0 Hz, 1H), 3.00 (d, J = 1 1.6 Hz, 3H), 2.97 (dd, J - 12.4, 5.5 Hz, 1H), 2.23 - 2.02 (m, 2H), 2.06 - 1.78 (m, 2H), 1.46 (d, J = 6.1 Hz, 6H).
[0679] (S)- 2- (N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- l ,2,3,4-tetrahydronaphmalen-l-yl)sulfamoyl)-N,N-dimethylacetamide 293 was prepared in an analogous fashion (5)-2-(N-(5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)- l,2,3,4-tetrahydronaphthalen-l-yl)sulfamoyl)acetic acid 288.
[0680] (R)-2-isopropoxy-5-(5-(5-((2-(methylsulfonyl)ethyl)amino)-5, 6, 7,8- tetrahydronaphthalen-l-yl)-l , 3, 4-thiadiazol-2-yl)benzonitrile (Compound 296)
Figure imgf000166_0001
To a solution of (/?)-5-(5-(5-amino-5,6,7,8-tetrahydronaphthalen-l-yl)-l ,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 228 (20 mg, 0.05 mmol) in DMA (0.5 mL) were added TEA (131 iL, 0.94 mmol) and methylvinylsulfone (50 mg, 0.50 mmol). The reaction mixture was heated to 50°C for 24 h. The crude reaction mixture was purified by preparative HPLC to give 9.9 mg (43%) of (#)-2-isopropoxy-5-(5-(5-((2- (methylsulfonyl)ethyl)amino)- 5,6,7,8- tetrahydronaphthalen-l-yl)- 1,3,4- thiadiazol- 2-yl) benzonitrile 296. LCMS-ESI (m/z) calculated for C25H2gN403S2: 496.6; found 497.1 [M+H]+, tR - 2.50 min. Ή NMR (400 MHz, CDC13) δ 8.22 - 8.09 (m, 2H), 7.65 (t, J = 8.2 Hz, 1H), 7.61 (d, J = 7.7 Hz, 1H), 7.42 (t, J - 7.7 Hz, 1H), 7.10 (d, J = 8.9 Hz, 1H), 4.77 (hept, J = 6.0 Hz, 1H), 4.60 (t, J = 4.9 Hz, 1H), 3.74 - 3.51 (m, 4H), 3.16 (dt, J = 17.5, 5.3 Hz, 1H), 3.10 - 2.99 (m, 4H), 2.32 - 2.09 (m, 2H), 1.95 (d, J = 4.9 Hz, 2H), 1.47 (d, J - 6.1 Hz, 6H).
[0681] (5)-2-isopropoxy-5-(5-(5-((2-(methylsulfonyl)ethyl)amino)-5,6,7,8- tetrahydronaphmalen-l-yl)-l,3,4-tWadiazol-2-yl)benzonitrile 297 was prepared in an analogous fashion from (5)-5-(5-(5-amino-5,6,7,8-tetrahydronaphthalen-l-yl)-l,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile hydrochloride 229.
[0682] (S)- 3- (N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1, 3, 4- thiadiazol- 2- yl)- 1, 2, -tetrahydronaphthalen- l-yl)sulfamoyl)-N-methylpropanamide (Compound 298)
Figure imgf000167_0001
To a stirring solution of (5)-ethyl 3- (N- (5- (5- (3-cyano- 4- isopropoxyphenyl)- 1,3 ,4- thiadiazol-2-yl)- 1,2,3, 4-tetrahydronaphthalen- l-yl)sulfamoyl)propanoate 286 (20 mg, 0.04 mmol) in MeOH (2 mL) was added methylamine (2M solution in MeOH, 20 μί,, 0.04 mmol). After stirring at 70°C overnight, the reaction mixture was concentrated and purified by preparative HPLC to give 6.4 mg (33%) of (5)-3-(N-(5-(5-(3-cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl) sulfamoyl)- N-methylpropanamide 298. LCMS-ESI (m/z) calculated for C26H29N504S2: 539.7; found 540.1 [M+H]+, tR = 3.21 min. 1H NMR (400 MHz, CDC13) δ 8.20 (dd, J - 8.9, 2.3 Hz, 1H), 8.15 (d, J = 2.2 Hz, 1H), 7.70 (d, J = 7.0 Hz, 1H), 7.56 (dd, J = 7.6, 1.2 Hz, 1H), 7.34 (dd, J = 17.2, 9.4 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 5.71 (s, 1H), 4.84 - 4.71 (m, 3H), 3.59 - 3.45 (m, 2H), 3.11 (dt, J = 17.6, 6.1 Hz, 1H), 3.03 - 2.89 (m,lH), 2.85 (t, J = 5.1 Hz, 3H), 2.73 (t, J = 7.4 Hz, 2H), 2.16 (dd, J = 9.1, 4.3 Hz, 1H), 2.00 (dd, J = 10.7, 3.4 Hz, 1H), 1.97 - 1.81 (m, 1H), 1.75 (s, 1H), 1.46 (d, J = 6.1 Hz, 6H). [0683] (R)- 3- (N- (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2-yl)- l,2,3,4-tetrahydronaphmalen-l-yl)sulfamoyl)-N-memylpropanamide can be prepared in an analogous fashion from (i?)-ethyl 3-(N-(5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol- 2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl)sulfamoyl)propanoate.
[0684] (R)- methyl 2- ((5- (5- (3- cyano- 4- isopropoxyphenyl)- 1, 3, 4- thiadiazol- 2- yl)- 1, 2, -tetrahydronaphthalen- l-yl)amino)acetate (TDZ THN INT-2)
Figure imgf000168_0001
To a stirring solution of the free base of (R)-5-(5-(5-amino-5,6,7,8- tefrahydronaphmalen-l-yl)-l,3,4-thiadiazol-2-yl)-2-isopropoxybenzonitrile 228 (137 mg, 0.35 mmol) in CH3CN (5.0 mL) were added methyl bromoacetate (59 μί, 0.39 mmol) and K2C03 (194 mg, 1.4 mmol). After stirring for 18 h, the mixture was diluted with brine and washed with NaHC03. The organic layer was dried with Na2S04 and concentrated. The resulting crude solid was purified by chromatography (MeOH / DCM) to provide 132 mg (82%) of (/?)- methyl 2- ((5- (5- (3- cyano- 4-isopropoxyphenyl)- 1,3,4- thiadiazol- 2-yl)- 1,2,3, 4-tetrahydronaphthalen- l-yl)amino)acetate TDZ THN INT-2. LCMS-ESI (m/z) calculated for C25H26N403S: 462.6; found 463.1 [M+H]+, tR = 2 AS min.
[0685] (S)- methyl 2- ((5- (5- (3- cyano- 4-isopropoxyphenyl)- 1,3,4- thiadiazol-2-yl)- 1,2,3, 4-tetrahydronaphthalen- l-yl)amino)acetate TDZ THN INT-3 was prepared in an analogous fashion from (5)-5-(5-(5-amino- 5,6,7,8- tetrahydronaphthalen-l-yl)- 1,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile (free base) 229.
[0686] (R)- methyl 2- ((tert- butoxycarbonyl) (5- (5- (3- cyano- 4- isopropoxyphenyl)- -thiadiazol -2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)amino)acetate (TDZ THN INT-4)
To a stirred solution of (Ti)-methyl 2-((5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4- tWadiazol-2-yl)-l,2,3,4-tetrahydronaphmalen-l-yl)amino)acetate TDZ THN INT-2 (132.0 mg, 0.29 mmol) in DCM (6.0 mL) were added Boc20 (125 mg, 0.57 mmol) and TEA (120 μί, 0.86 mmol). After stirring for 18 h, the mixture was concentrated. The resulting crude solid was purified by chromatography (EA / hexanes) to provide 100.0 mg (63%) of (R)- methyl 2-((tert-butoxycarbonyl)(5-(5-(3-cyano-4-isopropoxyphenyl)- 1 ,3,4-thiadiazol-2-yl)- l,2,3,4-tetrahydronaphthalen-l-yl)amino)acetate TDZ THN INT-4. LCMS-ESI (m/z) calculated for C3oH34N405S: 562.68; no M/Z observed, tR = 4.21 min.
[0687] (5 -methyl 2-((tert-butoxycarbonyl)(5-(5-(3-cyano-4-isopropoxyphenyl)-l ,3,4- thiadiazol-2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)amino)acetate TDZ THN INT-5 was prepared in an analogous fashion from (5)-methyl 2-((5-(5-(3-cyano-4-isopropoxyphenyl)- l,3,4-tWadiazol-2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)amino)acetate TDZ THN INT-3.
[0688] (R)- tert- butyl (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- l,2 -tetrahydronaphthalen-l-yl)(2-hydroxyethyl)carbamate (TDZ THN INT-6)
Figure imgf000169_0001
To a stirring solution of (i?)-methyl 2-((tert-butoxycarbonyl)(5-(5-(3-cyano-4- isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl)amino) acetate TDZ THN INT-4 (25 mg, 0.04 mmol) in THF (1.2 mL) at 70°C was added NaBFL; (4 mg, 0.11 mmol). After stirring for 0.5 h, MeOH (7.7 μί, 0.19 mmol) was added and the mixture was heated for an additional 1.5 h. The mixture was concentrated and the resulting solid was purified by preparative HPLC to provide 6.4 mg (27%) of (i?)-tert-butyl (5-(5-(3-cyano-4- isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)- 1,2,3,4- tetrahydronaphthalen-l-yl) (2- hydroxyethyl)carbamate TDZ THN INT-6. LCMS-ESI (m/z) calculated for C29H34N404S: 534.7; found 535.2 [M+H]+, tR = 3.94 min.
[0689] {S)-tert-bu y\ (5- (5- (3- cyano- 4 -isopropoxyphenyl)- 1,3,4- thiadiazol- 2-yl)- 1,2,3 ,4-tetrahydronaphthalen- 1-yl) (2-hydroxyethyl)carbamate TDZ THN INT-7 was prepared in prepared in an analogous fashion from (5)-methyl 2-((tert-butoxycarbonyl)(5-(5- (3 -cyano-4-isopropoxyphenyl)- 1 ,3 ,4-thiadiazol-2-yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl) amino) acetate TDZ THN INT-5.
[0690] General Procedure 29: Boc Deprotection of Tetrahydronaphthalene Amines
To a stirring solution of either Boc- protected (R)- or (5)-tetrahydronaphthalene amine in MeOH or dioxane was added 4N HC1/ dioxanes (4-10 eq). The reaction mixture was heated at 50°C for 18 h. The reaction mixture was concentrated and the resulting solid was purified by preparative HPLC.
[0691] (R)-5-(5-(5-((2-hydroxyethyl)amino)-5,6, 7,8-tetrahydroriaphthalen-l-yl)-l,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile (Compound 299)
Figure imgf000170_0001
Prepared using General Procedure 29: To a stirring solution of (i?)-tert-butyl (5-(5- (3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)- 1,2,3,4-tetrahydronaphthalen-l-yl) (2- hydroxyethyl)carbamate TDZ THN INT-6 (6.4 mg, 0.01 mmol) in dioxane (1 mL) was added 4N HC1 / dioxanes (10 μί, 0.03 mmol). After heating at 50°C for 1 h, the mixture was concentrated and the resulting solid was purified by preparative HPLC to provide 6.7 mg (100%) of (R)-5-(5-(5-((2-hydroxyethyl)amino)-5,6,7,8-tetrahydronaphthalen-l-yl)- 1,3,4- thiadiazol-2-yl)- 2-isopropoxybenzonitrile 299 as the TFA salt. LCMS-ESI (m/z) calculated for C24H26N402S: 434.5; found 435.1 [M+H]+, tR = 2.39 min. 1H NMR (400 MHz, CDC13) 6 8.17 (d, J = 13.9 Hz, 2H), 7.92 (s, 1H), 7.64 (s,lH), 7.40 (d, J = 30.4 Hz, 1H), 7.09 (d, J = 7.5 Hz, 1H), 4.84 - 4.70 (m, 1H), 4.58 (s, 1H), 3.92 - 3.70 (m, 3H), 3.71 - 3.56 (m, 1H), 3.10 (d, J = 34.0 Hz, 4H), 2.23 (s, 4H), 1.45 (t, J = 8.5 Hz, 6H).
[0692] (S)-5- (5- (5- ((2-hydroxyemyl)amino)- 5,6,7,8- tetrahydronaphthalen- 1 -yl)- 1 ,3,4- thiadiazol-2-yl)-2-isopropoxybenzonitrile 300 was prepared in an analogous fashion from (5)-tert-butyl (5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)- 1,2,3 ,4- tetrahydronaphthalen- l-yl)(2-hydroxyethyl)carbamate TDZ THN INT-7.
[0693] (R)-2- ((tert-butoxycarbonyl) (5-(5-(3-cyano-4-isopropoxyphenyl)- 1,3,4- thiadiazol-2-yl)-l,2, 3,4- tetrahydronaphthalen- l-yl)amino)acetic acid (TDZ THN INT-8)
Figure imgf000171_0001
To a stirring solution of of (i?)-methyl 2-((tert-butoxycarbonyl)(5-(5-(3-cyano- 4- isopropoxyphenyl)-l,3,4- thiadiazol-2-yl)-l,2,3,4- tetrahydronaphthalen-l-yl) amino) acetate TDZ THN INT-4. (75 mg, 0.13 mmol) in MeOH (2 ml) were added 10 drops of 1 N NaOH. The mixture was stirred at 50°C for 4 h, then diluted with H20 and neutralized with 1 N HC1. The aqueous solution was extracted with DCM and EA. The combined organic layers were dried over Na2S04 and concentrated to provide 77.0 mg (108%) of crude (R)-2-((tert- butoxycarbonyl) (5- (5- (3- cyano- 4-isopropoxyphenyl)- 1,3,4- thiadiazol- 2-yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)amino)acetic acid TDZ THN INT-8 as a white solid, which was used without further purification. LCMS-ESI (m/z) calculated for C29H32N405S: 548.7; found 549.2 [M+H]+, fR = 3.8 min.
[0694] (5)-2-((tert-butoxycarbonyl) (5-(5-(3-cyano-4-isopropoxyphenyl)- 1,3,4- thiadiazol-2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)amino)acetic acid TDZ THN INT-9 was prepared in an analogous fashion from (5)-methyl 2-((tert-butoxycarbonyl)(5-(5-(3-cyano-4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- 1,2,3,4- tetrahydronaphthalen- 1- yl) amino) acetate TDZ THN INT-5.
[0695] (R)- 2- ((5- (5- (3- cyano- 4- isopropoxyphenyl)- 1, 3, 4- thiadiazol- 2- yl)- 1, 2, 3, 4-tetrahydronaphthalen-l-yl)amino)acetic acid (Compound 301)
Figure imgf000171_0002
Prepared using General Procedure 29: To a solution of (i?)-2-((tert- butoxycarbonyl)(5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)amino)acetic acid TDZ THN INT-8 (15 mg, 0.03 mmol) in dioxane (1 mL) was added 4N HC1/ dioxanes (27 μΐ, 0.11 mmol). The reaction mixture was stirred at room temperature for 18 h then concentrated and the residue was purified by preparative HPLC to provide 10.3 mg (77%) of (R)- 2- ((5- (5- (3- cyano- 4- isopropoxyphenyl)- 1,3,4- thiadiazol- 2- yl)- l,2,3,4-tetrahydronaphthalen-l-yl)amino)acetic acid 301 as the HC1 salt. LCMS-ESI (m/z) calculated for C24H24N4O3S: 448.5; found 449.2 [M+H]+, tR - 2.28 min. Ή NMR (400 MHz, CDC13) δ 8.18 - 8.04 (m, 2H), 7.67 (d, J = 7.7 Hz, 1H), 7.57 (t, J = 9.8 Hz, 1H), 7.34 (dd, J = 15.4, 7.8 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 4.76 (d, J = 12.3, 6.1 Hz, 1H), 4.64 (s, 1H), 3.72 (dd, J - 31.2, 16.7 Hz, 2H), 3.12 (dd, J = 12.4, 5.8 Hz, 1H), 3.02 - 2.82 (m, 1H), 2.06 (dd, J= 26.7, 20.1 Hz, 3H), 1.81 (d, J= 5.9 Hz, 1H), 1.46 (d, J = 6.1 Hz, 6H).
[0696] (S)- 2- ((5- (5- (3- cyano- 4- isopropoxyphenyl)- 1, 3, 4- thiadiazol- 2-yl)- 1, 2, 3, 4-tetrahydronaphthalen-l-yl)amino)acetic acid 302 was prepared in an analogous fashion from (5)-2-((tert-butoxycarbonyl) (5-(5-(3-cyano-4-isopropoxyphenyl)- 1,3,4-thiadiazol- 2- yl)-l,2,3,4-tetrahydronaphthalen-l-yl)amino)acetic acid TDZ THN ENT-9.
[0697] General Procedure 30: Preparation of Tetrahydronaphthalene Amino Amides
To either boc-protected (R)- or (5)-tetrahydronaphthalene aminoacid TDZ THN INT- 8 or TDZ THN INT-9 in DMF were added HOBT (2 eq) and EDC (2 eq). After 10 min, the appropriate amine (10 eq) was added and the reaction mixture was stirred for 18 h at room temperature. The crude reaction mixture was diluted with NaHC03 and extracted with EA. The combined organic layers were dried over Na2S04 and purified by preparative HPLC. The Boc protecting group is subsequently removed by using General Procedure 29.
[0698] Compounds 303-306 were prepared using General Procedure 30 followed by General Procedure 29.
[0699] (R)- tert- butyl (5- (5- (3- cyano- 4- isopropoxyphenyl)- 1, 3, 4- thiadiazol-2-yl)- 1, 2, -tetrahydronaphthalen-l-yl)(2-oxo-2-(pyrrolidin-l-yl)ethyl)carbamate
Figure imgf000172_0001
Prepared using General Procedure 30. To a stirring solution of (R)-2-((tert- butoxycarbonyl)(5-(5-(3-cyano-4- isopropoxyphenyl)- 1,3,4- thiadiazol-2-yl)- 1,2,3,4- tetrahydronaphthalen-l-yl)amino)acetic acid TDZ THN INT-8 (25 mg, 0.05 mmol) in DMF (1 mL) were added HOBT (9 mg, 0.07 mmol) and EDC (13 mg, 0.07 mmol). After 10 min, pyrrolidine (37 μί, 0.46 mmol) was added and the reaction mixture was stirred for 18 h at room temperature. The crude mixture was purified by preparative HPLC to give 8.7 mg (32%) of (R)-tert-butyl (5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2-yl)-l,2,3,4- tetrahydronaphthalen- 1 -yl)(2-oxo-2-(pyrrolidin- 1 -yl)ethyl)carbamate. LCMS-ESI (m/z) calculated for C33H39N504S: 601.8; found 602.3 [M+H]+, tR = 4.02 min.
[0700] (S)-tert-butyl (5- (5- (3- cyano- 4- isopropoxyphenyl)- l,3,4-thiadiazol-2-yl)- l,2,3,4-tetrahydronaphthalen-l-yl)(2-oxo-2-(pyrrolidin-l-yl)ethyl)carbamate was prepared in an analogous fashion from (5)-2-((tert-butoxycarbonyl)(5-(5-(3-cyano-4-isopropoxyphenyl)- l,3,4-thiadiazol-2-yl)-l,2,3,4-tetrahydronaphthalen-l-yl)amino) acetic acid TDZ THN INT- 9.
[0701] (R)- 2 -isopropoxy- 5- (5- (5- ((2- oxo- 2- (pyrrolidin- 1-yl) ethyl) amino)- 5, 6, 7, -tetrahydronaphthalen-l-yl)-l,3,4-thiadiazol-2-yl)benzonitrile (Compound 303)
Figure imgf000173_0001
Prepared using General Procedure 29. A solution of (R)-tert-butyl (5-(5-(3-cyano-4- isopropoxyphenyl)- 1,3,4- thiadiazol-2-yl)- 1,2,3,4-tetrahydronaphthalen-l-yl) (2-oxo-2- (pyrrolidin-l-yl)ethyl)carbamate (8.7 mg, 0.02 mmol) in 4N HCl/ dioxanes (0.5 mL) was stirred at room temperature for 18 h. The reaction mixture was concentrated to provide 7.6 mg (81%) of (i?)-2-isopropoxy-5-(5-(5-((2-oxo-2-^yrrolidin-l-yl)ethyl)airrino)-5,6,7,8- tetrahydronaphmalen-l-yl)-l,3,4-thiadiazol-2-yl)benzonitrile 303 as the HCl salt. LCMS-ESI (m/z) calculated for C28H31N502S: 501.6; found 502.2 [M+H]+, tR = 2.52 min. Ή NMR (400 MHz, CDC13) δ 8.18 (d, J = 8.8 Hz, 2H), 8.08 (s, 1H), 7.64 (s, 1H), 7.44 (s, 1H), 7.10 (d, J = 8.5 Hz, 1H), 4.78 (td, J = 12.1, 6.0 Hz, 2H), 3.92 (s, 2H), 3.86 - 3.71 (m, 1H), 3.73 - 3.56 (m, 1H), 3.47 (d, J = 30.8 Hz, 3H), 3.16 (s, 1H), 3.04 (s, 1H), 2.22 (s, 3H), 2.14 - 1.67 (m, 5H), 1.57 - 1.30 (m, 6H). [0702] (S)- 2- isopropoxy- 5- (5- (5- ((2- oxo- 2- (pyrrolidin- 1- yl) ethyl) amino)-5, 6, 7,
8-tetrahydronaphthalen-l-yl)-l,3,4-tWadiazol-2-yl)benzonitrile 304 was prepared in an analogous fashion from (<S)-tert-butyl (5-(5-(3-cyano-4-isopropoxyphenyl)-l,3,4-thiadiazol-2- yl)- 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl)(2-oxo-2-(pyrrolidin- 1 -yl)ethyl)carbamate.
[0703] 2-isopropoxy-5-(4, 4, 5, 5-tetramethyl-l, 3, 2-dioxaborolan-2-yl)benzonitrile (TDZ
INT-5)
Figure imgf000174_0001
A solution of 5-bromo-2-isopropoxybenzonitrile (1.0 g, 4.1 mmol), 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(l,3,2-dioxaborolane) (1.27 g, 5.0 mmol), and potassium acetate (0.82 g, 8.3 mmol) in anhydrous DMF (10 mL) was degassed by passing N2 through the solution for 10 min before the addition of PdCl2(dppf).CH2Cl2 (198 mg, 0.24 mmol). The reaction mixture was heated at 120°C for 3 h. The reaction flask was cooled and diluted with EA (100 mL), washed with water (100 mL) and brine (100 mL) then dried over MgS04. The product was purified by chromatography (EA / hexanes) to afford 900.0 mg (75%) of 2-isopropoxy-5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzonitrile TDZ INT-5 as a light green oil which solidified to a white solid. LCMS-ESI (m/z) calculated for C16H22BN03: 287.2; found 288.2 [M+H]+, tK = 4.07 min. Ή NMR (400 MHz, CDC13) 6 7.97 (d, J = 1.5 Hz, 1H), 7.88 (dd, J= 8.5, 1.7 Hz, 1H), 6.91 (d, J= 8.5 Hz, 1H), 4.67 (dt, J = 12.2, 6.1 Hz, 1H), 1.38 (d, J= 6.1 Hz, 6H), 1.30 (s, 12H).
[0704] 5 -(3-bromo-l, 2, 4-thiadiazol-5-yl)-2 -isopropoxy benzonitrile (TDZ INT-6)
Figure imgf000174_0002
A solution of 2-isopropoxy-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)benzonitrile TDZ INT-5 (326 mg, 1.13 mmol), 3-bromo-5-chloro-l,2,4-thiadiazole (250 mg, 1.26 mmol), 3P04.3H20 (670 mg, 3.15 mmol) in DME/H20 (4:1, 5 mL) was purged with N2 for 10 min before the addition of Pd(PPh3)4 (145 mg, 0.12 mmol). The mixture was further degassed with N2 for 5 min and then heated to 90°C for 6 h under N2. Upon cooling, the reaction mixture was diluted with EA (100 mL), washed with water (100 mL) and brine (100 mL), then dried over MgS04. The solution was filtered and concentrated. The crude product was purified by silica gel column chromatography (20% EA / hexanes) to afford 143 mg (35 %) of 5-(3-bromo-l,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile TDZ INT-6 as white solid. LCMS-ESI (m/z) calculated for Ci2Hi0BrN3OS; 324.2; found 325.9 [M+2]+, tR = 3.86 min. l NMR (400 MHz, CDC13) δ 8.26 - 7.91 (m, 2H), 7.08 (d, J= 8.9 Hz, 1H), 4.77 (dt, J - 12.1, 6.1 Hz, 1H), 1.47 (t, J = 8.2 Hz, 6H). 13C NMR (101 MHz, CDC13) δ 187.77, 162.92, 146.21, 133.23, 122.11, 115.21, 113.91, 104.29, 72.99, 21.87.
[0705] General Procedure 31: Coupling of Heterocyclic Bromides to Indane Amine
A reaction pressure flask was charged sequentially with the heterocyclic bromide (1 eq), (i?)- or (5)-Boc-protected indane amine (1 eq), DME/H20 (3:1, 0.07 M) and potassium carbonate (3 eq). The mixture was degassed by bubbling N2 gas through the stirring solution for 20 min before Pd(PPh3)4 (0.07 eq) was added and the mixture was degassed for additional 5 min. The reaction flask was capped tightly and the mixture was heated at 85°C for 12-24 h. The reaction mixture was cooled to room temperature, diluted with water (2x volume), and stirred for 30 min. The resulting solid was filtered, washed with hexanes, and dried under high vacuum. The crude product was purified by silica gel column chromatography (EA / hexanes) or used in the next experiment without purification
[0706] Compounds TDZ INT-7 and TDZ INT-8 were prepared using General Procedure 31.
[0707] (S) -tert- butyl (4- (5- (3 -cyano- 4- isopropoxyphenyl) -1,2,4- thiadiazol- 3- yl) - 2,3- dihydro-lH-inden-l-yl)carbamate (TDZ INT-7)
Figure imgf000175_0001
Prepared using General Procedure 31. A suspension of 5-(3-bromo-l,2,4-thiadiazol- 5-yl)-2-isopropoxybenzonitrile TDZ INT-6 (143 mg, 0.44 mmol), (S)-tert-buty\ (4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)- 2,3-dihydro-lH-inden-l-yl) carbamate IND INT-17 (127 mg, 0.44 mmol) and 3P04.3H20 (235 mg, 1.1 mmol) in DMF and H20 (4:1, 12 mL) was degassed with N2 for 20 min before the addition of Pd(PPh3)4 (51 mg, 0.04 mmol). The mixture was degassed with N2 for an additional 5 min and the suspension was heated under N2 at 100°C for 5 h. Upon cooling, the reaction mixture was diluted with EA (50 mL) and washed with water (100 mL) and brine (100 mL). The resulting crude product was purified by chromatography (EA / hexanes) to afford 150 mg (71%) of (S)-tert-buty\ (4-(5-(3-cyano- 4-isopropoxyphenyl)- l,2,4-thiadiazol-3-yl)- 2,3-dihydro-lH-inden-l-yl) carbamate TDZ INT-7 as a white solid. LCMS-ESI (m/z) calculated for C26H28N403S: 476.2; found 499.1 [M+Na]+, ¾ = 4.42 min. 1H NMR (400 MHz, CDC13) δ 8.31 - 8.19 (m, 2H), 8.16 (dd, J = 8.9, 2.3 Hz, 1H), 7.47 (d, J - 7.4 Hz, 1H), 7.39 (d, J= 7.6 Hz, 1H), 7.09 (d, J= 9.0 Hz, 1H), 5.26 (dt, J = 33.3, 16.7 Hz, 1H), 4.77 (td, J = 12.1, 6.0 Hz, 2H), 3.70 - 3.51 (m, 1H), 3.37 - 3.16 (m, 1H), 2.75 - 2.51 (m, 1H), 1.94 - 1.75 (m, 1H), 1.51 (s, 9H), 1.49 - 1.44 (m, 6H).
[0708] (R)-tert-butyl (4-(5-(3-cyano-4-isopropoxyphenyl)- l,2,4-thiadiazol-3-yl)- 2,3- dihydro-lH-inden-l-yl)carbamate TDZ INT-8 can be prepared in a similar fashion using (R)- tert-butyl 4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-2,3-dihydro- lH-inden- 1 - ylcarbamate IND INT-18.
[0709] (S)- 5- (3- (1- amino- 2,3- dihydro- 1H- Inden-4-yl)- 1, 2, 4-thiadiazol- 5- yl)- 2- isopropoxy benzonitrile hydrochloride (Compound 307)
Figure imgf000176_0001
To a stirring solution of (4-(5-(3-cyano-4-isopropoxyphenyl)-l,2,4-thiadiazol-3-yl)- 2,3-dihydro-lH-inden-l-yl)carbamate TDZ INT-7 (150 mg, 0.31 mmol) in 1,4-dioxane (5 mL) was added 4N HCl in 1,4-dioxane (2 mL). The reaction mixture was stirred at 55°C for 3.5 h. Upon cooling to 0°C, the reaction mixture was diluted with diethyl ether (20 mL) and the resulting solid was filtered then air dried to afford 101 mg (78%) of (5)-5-(3-(l-amino- 2,3 -dihydro- 1 H-inden-4-yl)- 1 ,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 307 as a white solid. LCMS-ESI (m/z) calculated for C21H20N4OS: 376.1; found 360.1 (M- NH2). ¾ = 2.49 min. 1H NMR (400 MHz, DMSO) δ 8.53 (t, J = 5.6 Hz, 1H), 8.46 (s, 2H), 8.38 - 8.34 (m, 1H), 7.78 (d, J= 7.5 Hz, 1H), 7.60 - 7.40 (m, 2H), 5.07 - 4.89 (m, 1H), 4.82 (s, 1H), 3.73 - 3.50 (m, 1H), 3.39 (ddd, J= 17.5, 8.6, 6.2 Hz, 1H), 2.61 - 2.47 (m, 1H), 2.08 (ddd, J = 14.3, 8.8, 6.0 Hz, 1H), 1.38 (d, J = 6.0 Hz, 6H). 13C NMR (101 MHz, DMSO) 6 185.18, 172.38, 161.92, 143.78, 141.15, 134.12, 133.22, 130.02, 128.81, 127.47, 127.07, 122.56, 115.54, 114.92, 102.40, 72.34, 54.53, 31.77, 30.04, 21.52.
[0710] (R)-5-Q- (1-amino- 2,3- dihydro-lH- inden-4-yl)- 1,2,4- thiadiazol-5-yl)- 2 isopropoxy benzonitrile hydrochloride can be prepared in a similar fashion using (R)-tert- butyl (4-(5-(3-cyano-4-isopropoxyphenyl)- 1 ,2,4-thiadiazol-3-yl)-2,3-dihydro- 1 H-inden- 1 - yl)carbamate TDZ INT-8. [0711] (S)- tert-butyl (4- (3-bromo- 1,2,4-thiadiazol- 5-yl)- 2,3 -dihydro-lH-inden-l-yl) carbamate (TDZ INT-9
Figure imgf000177_0001
A solution of (5)-tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3- dihydro-lH-inden-l-ylcarbamate IND I T-17 (90.7 mg, 0.25 mmol), 3-bromo-5-chloro- 1,2,4-thiadiazole (50 mg, 0.25 mmol), and K3P04.3H20 (134 mg, 0.63 mmol) in DME/H20 (4:1, 1 mL) was purged with N2 for 10 min before the addition of Pd(PPh3)4 (29 mg, 0.025 mmol). The mixture was further degassed with N2 for 5 min and then heated under microwave irradiation at 100°C for 45 min. Upon cooling, the reaction mixture was diluted with EA (10 mL), washed with water (10 mL) and brine (10 mL), then dried over MgS04. The reaction mixture was filtered and concentrated. The crude product was purified by silica gel column chromatography (20% EA / hexanes) to afford 35 mg (35%) of (S)-tert-buty\ (4- (3-bromo-l,2,4-thiadiazol-5-yl)-2,3-dihydro-lH-inden-l-yl)carbamate TDZ INT-9 as a white solid. LCMS-ESI (m/z) calculated for Ci6H18BrN302S: 396.3; no M+ observed, tK = 4.10 min. Ή NMR (400 MHz, CDC13) 6 8.00 (d, J= 7.8 Hz, 1H), 7.54 (d, J= 7.5 Hz, 1H), 7.39 (t, J= 7.7 Hz, 1H), 5.28 (dd, J= 15.7, 7.8 Hz, 1H), 4.81 (d, J = 7.9 Hz, 1H), 3.27 (ddd, J= 16.6, 9.0, 3.4 Hz, 1H), 3.04 (dt, J = 16.5, 8.2 Hz, 1H), 2.72 (td, J = 8.1, 4.2 Hz, 1H), 2.00 - 1.74 (m, 1H), 1.49 (s, 9H).
[0712] (R)-tert-buty\ (4-(3-bromo- 1 ,2,4-thiadiazol-5-yl)-2,3-dihydro- 1 H-inden- 1 -yl) carbamate can be prepared in a similar fashion from (R)-tert-butyl 4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-2,3-dihydro-lH-inden-l-ylcarbamate IND INT-18.
[0713] (S) -tert-butyl (4-(3-(3-cyano-4-isopropoxyphenyl)-l,2,4-thiadiazol-5-yl)- 2,3- dihyd -1 H-inden- 1 -yl)carbamate
Figure imgf000177_0002
Prepared using General Procedure 31. A suspension of 2-isopropoxy-5-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)benzonitrile (13 mg, 0.04 mmol), (4-(3- bromo-l,2,4-thiadiazol-5-yl)-2,3-dihydro-lH-inden-l-yl)carbamate TDZ INT-9 (15 mg, 0.04 mmol) and K3P04.3H20 (20 mg, 0.09 mmol) in DMF and H20 (4:1, 1.25 mL) was degassed with N2 for 20 min before the addition of Pd(PPh3)4 (4.3 mg, 0.003 mmol). The mixture was degassed with N2 for an additional 5 min and the suspension was heated under microwave irradiation at 100°C for 45 min. Upon cooling, the reaction mixture was diluted with EA (10 mL) and washed with water (10 mL) and brine (10 mL). The resulting crude product was purified by silica gel column (EA and hexanes) to afford 15.0 mg (83%) of (5)-tert-butyl (4- (3-(3-cyano-4-isopropoxyphenyl)-l,2,4-tMadiazol-5-yl)-2,3-dihydro-lH-inden-l- yl)carbamate as white solid. LCMS-ESI (m/z) calculated for C26H28N403S: 476.1; no M/Z observed, tR = 4.55 min. Ή NMR (400 MHz, CDC13) δ 8.59 (d, J = 2.1 Hz, 1H), 8.50 (dd, J = 8.9, 2.2 Hz, 1H), 8.05 (d, J= 7.7 Hz, 1H), 7.53 (d, J= 7.5 Hz, 1H), 7.41 (t, J= 7.6 Hz, 1H), 7.07 (d, J= 9.0 Hz, 1H), 5.30 (dd, J= 15.9, 7.9 Hz, 1H), 4.84 (d, J = 8.7 Hz, 1H), 4.80 - 4.70 (m, 1H), 3.40 (ddd, J= 16.7, 8.9, 3.4 Hz, 1H), 3.15 (dt, J= 16.6, 8.2 Hz, 1H), 2.74 (ddd, J = 12.7, 8.0, 4.0 Hz, 1H), 1.94 (dd, J= 12.9, 8.3 Hz, 1H), 1.49 (d, J = 12.3 Hz, 9H), 1.44 (dd, J = 10.6, 6.1 Hz, 6H).
[0714] (R)-tert-bu yl (4-(3-(3-cyano-4-isopropoxyphenyl)-l,2,4-thiadiazol-5-yl)- 2,3- dihydro-lH-inden-l-yl)carbamate can be prepared in a similar fashion using (if)-tert-butyl (4- (3-bromo- 1 ,2,4-thiadiazol-5-yl)-2,3 -dihydro- 1 H-inden- 1 -yl)carbamate.
[0715] (S)-5-(5-(l-amino-2 -dihydro-lH nden-4-yl)-l,2 -thiadiazol-3-yl)-2-isopropoxy benzonitrile hydrochloride (Compound 308)
Figure imgf000178_0001
To a stirring solution of (S)-tert-buty\ (4-(3-(3-cyano-4-isopropoxyphenyl)-l,2,4- thiadiazol-5-yl)-2,3-dihydro-lH-inden-l-yl)carbamate (15 mg, 0.03 mmol) in 1,4-dioxane (1 mL) was added 4N HC1 in 1,4-dioxane (0.1 mL). The reaction mixture was stirred at 55°C for 3.5 h. Upon cooling to 0°C, the reaction mixture was diluted with diethyl ether (10 mL) and the resulting solid was filtered and dried to afford 10.0 mg (77%) of (S)-5-(5-(l-amino- 2,3-dihydro-lH-inden-4-yl)- l,2,4-thiadiazol-3-yl)-2- isopropoxybenzonitrile hydrochloride 308 as a white solid. LCMS-ESI (m/z) calculated for: C21H2oN4OS: 376.4; found 377.1 [M+H]+. tR = 2.48 min. Ή NMR (400 MHz, DMSO) 6 8.55 (dd, 7 = 5.7, 2.1 Hz, 1H), 8.52 (d, J= 2.3 Hz, 1H), 8.46 (s, 3H), 8.27 (d, J= 7.5 Hz, 1H), 7.86 (d, J= 7.6 Hz, 1H), 7.60 (t, J = 7.7 Hz, 1H), 7.50 (d, J = 8.9 Hz, 1H), 4.91 (ddd, J = 16.9, 12.2, 6.2 Hz, 2H), 3.54 - 3.39 (m, 1H), 3.35 - 3.20 (m, 1H), 2.72 - 2.55 (m, 1H), 2.25 - 2.07 (m, 1H), 1.38 (d, J= 6.0 Hz, 6H).
[0716] (i?)-5-(5- (1 -amino- 2,3- dihydro-lH- inden-4-yl)- l,2,4-thiadiazol-3-yl)-2- isopropoxybenzonitrile hydrochloride can be prepared in a similar fashion using R)-tert- butyl (4-(3-(3-cyano-4-isopropoxyphenyl)- 1 ,2,4-thiadiazol-5-yl)-2,3-dihydro- 1 H-inden- 1 - yl)carbamate.
[0717] (S)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l, 2, 4-thiadiazol-3-yl)-2, 3-dihydro-lH- inden-l-yl) sulfamide (Compound 309)
Figure imgf000179_0001
Prepared using General Procedure 24: To a solution of (5)-5-(3-(l-amino-2,3- dihydro-lH-inden-4-yl)-l,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 307 (6 mg, 0.01 mmol) in dioxane (1 mL) was added sulfamide (7 mg, 0.07 mmol) and the mixture was heated to 90°C. After 16 h, the solvent was evaporated and the residue was purified by preparative HPLC to provide 4.0 mg (66%) of (5)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)- l,2,4-thiadiazol-3-yl)-2,3-dihydro-lH-inden-l-yl) sulfamide 309. LCMS-ESI (m/z) calculated for C21H2iN503S2: 455.1; found 456.1 [M+H]+, tR = 3.55 min.
[0718] (^)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,2,4-thiadiazol-3-yl)-2,3-dihydro- 1 H-inden- 1-yl) sulfamide can be prepared in a similar fashion using (i?)-5-(3-(l-amino-2,3- dihydro- 1 H-inden-4-yl)- 1 ,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride.
[0719] (S)-N-(4-(3-(3-cyano-4-isopropoxyphenyl)-l,2,4-thiadiazol-5-yl)-2,3-dihydro-lH- inden-l- l sul amide Com ound 310
Figure imgf000179_0002
Prepared using General Procedure 24: To a solution of (5)-5-(5-(l-amino-2,3- dihydro-lH-inden-4-yl)-l,2,4-thiadiazol-3-yl)-2-isopropoxybenzonitrile hydrochloride 308 (6 mg, 0.01 mmol) in dioxane (1 mL) was added sulfamide (7 mg, 0.07 mmol) and the mixture was heated to 90°C. After 16 h, the solvent was evaporated and the residue was purified by preparative HPLC to provide 4.0 mg (66%) of (5)-N-(4-(3-(3-cyano-4-isopropoxyphenyl)- l,2,4-thiadiazol-5-yl)-2,3-dihydro-lH-inden-l-yl) sulfamide 310. LCMS-ESI (m/z) calculated for C21H2iN503S2: 455.1; found 456.1 [M+H]+, tR = 3.62 min.
[0720] (i?)-N-(4-(3-(3-cyano-4-isopropoxyphmyl)-l,2,4-thiadiazol-5-yl)-2,3-dihydro-lH- inden-l-yl) sulfamide can be prepared in a similar fashion using ( ?)-5-(5-(l-amino-2,3- dihydro- 1 H-inden-4-yl)- 1 ,2,4-thiadiazol-3-yl)-2-isopropoxybenzonitrile hydrochloride.
[0721] (S)- 5- (3- (1- ((2-((tert-butyldimethylsilyl) oxy)ethyl) amino)- 2,3-dihydro-lH- inden-4-yl)-l, 2, b4-thiadiazol- 5- yl)- 2- isopropoxybenzonitrile and (S)- 5- (3- (I- (bis (2- ((tert-butyldimethylsilyl) oxy) ethyl) amino)- 2,3-dihydro-lH-inden-4-yl)- 1,2,4- thiadiazol-5- yl)- 2-isopropoxybenzonitrile
Figure imgf000180_0001
To a suspension of (5)-5-(3-(l-amino-2,3-dihydro-lH-inden-4-yl)-l,2,4-thiadiazol-5- yl)-2-isopropoxybenzonitrile hydrochloride 307 (30 mg, 0.07 mmol) in anhydrous CH3CN (1 mL) were added potassium carbonate (60.3 mg, 0.43 mmol), tetrabutylammonium iodide (53.7 mg, 0.14 mmol) and (2-bromoethoxy)(tert-butyl)dimethylsilane (35 mg, 0.14 mmol). The reaction mixture was stirred at 85°C for 16 h. Water (5 mL) was added and the reaction mixture was extracted with EA (2 5 mL). The organic layers were washed with brine, dried, and purified by column chromatography (EA / hexanes) to provide 18 mg (46%) of (S)-5-(3- ( 1 -(bi'5(2-((tert-butyldimethylsilyl)oxy)ethyl)amino)-2,3 -dihydro- 1 H-inden-4-yl)- 1 ,2,4- thiadiazol-5-yl)-2-isopropoxybenzonitrile as a white solid. LCMS-ESI (m/z) calculated for: C37H56N403SSi2: 693.1; found 694.3 [M+H]+, iR = 3.81 min. Further elution provided 13 mg (33%) of (5 -5-(3-(l-((2-((te^butyldimemylsilyl)oxy)emyl)amino)-2,3-dihydro-lH-inden-4- yl)-l,2,4-thiadiazol-5-yl)-2 -isopropoxybenzonitrile as a white solid. LCMS-ESI (m/z) calculated for: C29H38N402SSi: 534.3; found 535.3 [M+H]+, tR = 3.16 min.
[0722] CK)-5-(3 -( 1 -((2-((te^butyldimethylsilyl)oxy)ethyl)amino)-2,3 -dihydro- 1 H-inden- 4-yl)- 1,2,4- thiadiazol- 5-yl)- 2-isopropoxybenzonitrile and (R)- 5- (3- (1- (bis (2- ((tert- butyldimethylsilyl) oxy) ethyl) amino)- 2,3-dihydro-lH-inden-4-yl)- l,2,4-thiadiazol-5-yl) - 2-isopropoxybenzonitrile can be prepared in a similar fashion using (/?)-5-(3-(l-amino-2,3- dihydro- 1 H-inden-4-yl)- 1 ,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride.
[0723J (S)-5-(3-(l-((2-hydroxyethyl)amino)-2 -dihydro-lH-inden-4-yl)^^
-yl)-2-isopropoxybenzonitrile hydrochloride (Compound 311)
Figure imgf000181_0001
To a solution of (5)-5-(3-(l-((2-((tert-butyldimethylsilyl)oxy)ethyl)amino)-2,3- dihydro-lH-inden-4-yl)-l,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile (13 mg, 0.02 mmol), in 1,4-dioxane (1 mL) was added 4N HCl in dioxanes (0.2 mL). The mixture was stirred at room temperature for 4 h and then was diluted with diethyl ether (5 mL) and filtered to afford 8.3 mg (75%) of (5 -5-(3-(l-((2-hydroxyethyl)amino)-2,3-dihydro-lH-inden-4-yl)-l,2,4- thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 311 as a white solid. LCMS-ESI (m/z) calculated for: C23H24N402S: 420.2; found 421.1 [M+H]+, tR = 2.47 min. Ή NMR (400 MHz, DMSO) δ 9.07 (d, J= 4.9 Hz, 2H), 8.54 (d, J = 2.3 Hz, 1H), 8.38 (dd, J= 8.9, 2.3 Hz, 2H), 7.86 (d, J = 7.5 Hz, 1H), 7.62 - 7.39 (m, 2H), 5.27 (s, 1H), 4.95 (ddd, J= 15.6, 9.9, 4.9 Hz, 2H), 3.75 - 3.57 (m, 3H), 3.41 (ddd, J= 18.5, 9.4, 5.2 Hz, 1H), 3.03 (ddd, J= 18.2, 12.6, 6.6 Hz, 2H), 2.60 - 2.51 (m, 1H), 2.36 - 2.18 (m, 1H), 1.38 (d, J = 6.0 Hz, 6H). 13C NMR (101 MHz, DMSO) δ 185.20, 172.32, 161.92, 144.76, 139.09, 134.13, 133.23, 128.93, 128.08, 127.42, 122.56, 115.55, 114.92, 102.41, 72.35, 61.21, 56.63, 53.42, 46.36, 27.97, 21.52.
[0724] (i?)-5-(3-(l-((2-hydroxyethyl)amino)-2,3-dihydro-lH-inden-4-yl)- 1,2,4- thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride can be prepared in a similar fashion using (i?)-5-(3-(l-((2-((tert-butyldimethylsilyl) oxy) ethyl) amino)- 2,3-dihydro-lH-inden-4- yl)- 1 ,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile.
[0725] (S)-5-(3-(l^is(2-hydroxyethyl)amino)-2,3-dihydro-lH nden-4-yl)-l,2,4- thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride (Compound 312)
Figure imgf000181_0002
To (5 -5-(3-(l -(bi5(2-((te^butyldimethylsilyl)oxy)ethyl)amino)-2,3-dihydro-l H- inden-4-yl)-l,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile (16 mg, 0.02 mmol), in 1,4- dioxane (1 mL) was added 4N HC1 in dioxanes (0.2 mL). The mixture was stirred at room temperature for 4 h, then diluted with diethyl ether (5 mL) and filtered to afford 8.3 mg (75%) of (S)-5- (3- (1- (bw(2-hydroxyethyl) amino)- 2,3- dihydro- 1H- inden-4-yl)- 1,2,4- thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 312. LCMS-ESI (m/z) calculated for: C25H28N403S: 464.2; found 465.2 [M+H]+, tR = 2.47 min.
[0726] (R)-5- (3- (1- (bis (2-hydroxyethyl)amino)-2,3 -dihydro- lH-inden-4-yl)- 1,2,4- thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride can be prepared in a similar fashion using (i?)-5-(3-(l-(bis(2-((tert-butyldimethylsilyl)oxy)ethyl)amino)- 2,3-dihydro-lH-inden-4- yl)- 1 ,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile.
[0727] (S)-methyl 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l ,2,4-thiadiazol-3-yl)-2,3- dihydro-lH-inden-l-yl)sulfamoyl)acetate (Compound 313)
Figure imgf000182_0001
Prepared using General Procedure 25: To a stirring solution of (5)-5-(3-(l-amino-2,3- dihydro- 1 H-inden-4-yl)- 1 ,2,4-thiadiazol-5-yl)-2-isopropoxybenzonitrile hydrochloride 307 (30 mg, 0.07 mmol) and DIE A (31.8 mg, 0.24 mmol) in DCM (1 mL) was added a solution of methyl-2-(chlorosulfonyl)acetate (21.8 mg, 0.12 mmol) in DCM (0.5 mL). The reaction mixture was stirred for 15 min at room temperature. The reaction mixture was partitioned between DCM (5 mL) and NaHC03 (5 mL). The organic layer was separated and dried over MgS04, filter and concentrated. Purification by column chromatography (EA/hexanes) provided 15.0 mg (42%) of (5)-methyl 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,2,4- thiadiazol-3-yl)-2,3-dihydro-lH-inden-l-yl)sulfamoyl)acetate 313 as a light yellow solid. LCMS-ESI (m/z) calculated for C24H24N405S2: 512.6; found 513.1 [M+H]+, tR = 3.86 min. 1H NMR (400 MHz, CDC13) 6 8.32 (d, J = 7.7 Hz, 1H), 8.24 (d, J= 2.2 Hz, 1H), 8.14 (dd, J = 8.9, 2.3 Hz, 1H), 7.62 (d, J= 7.5 Hz, 1H), 7.42 (t, J= 7.6 Hz, 1H), 7.09 (d, J= 9.0 Hz, 1H), 5.10 (q, J= 7.5 Hz, 2H), 4.78 (dt, J= 12.2, 6.1 Hz, 1H), 4.24 - 4.00 (m, 2H), 3.84 (d, J= 4.6 Hz, 3H), 3.66 (ddd, J= 17.3, 8.8, 3.6 Hz, 1H), 3.39 - 3.22 (m, 1H), 2.70 (ddd, J= 20.0, 10.4, 5.7 Hz, 1H), 2.15 - 2.00 (m, 1H), 1.48 (t, J= 5.3 Hz, 6H).
[0728] (^)-methyl 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l ,2,4-thiadiazol-3-yl)-2,3- dihydro-lH-inden-l-yl)sulfamoyl)acetate can be prepared in a similar fashion using (R)-5-(3- (l-amino-2,3- dihydro- 1H- inden-4-yl)- 1,2,4- thiadiazol- 5- yl)- 2- isopropoxybenzonitrile hydrochloride.
[0729] (S)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,2,4-thiadiazol-3-yl)-2,3-dihydro-lH- inden-l-yl)-2-hydroxyethanesulfonamide (Compound 314)
Figure imgf000183_0001
Prepared using General Procedure 27: To a stirring solution of (5)-methyl 2-(N-(4-(5- (3-cyano- 4- isopropoxyphenyl)- 1,2,4- thiadiazol- 3- yl)- 2,3- dihydro- 1H- inden- 1- yl)sulfamoyl)acetate (14 mg, 0.02 mmol) 313 in THF (1 mL) was NaBiL; (2.4 mg, 0.06 mmol) at room temperature. The reaction mixture was heated to 75°C for 10 min and a drop of MeOH was added. After 10 min, the reaction mixture was cooled and concentrated. Purification by preparative HPLC gave 8.5 mg (70%) of (5)-N-(4-(5-(3- cyano-4- isopropoxyphenyl)- 1,2,4- thiadiazol- 3- yl)- 2, 3- dihydro- 1H- inden- 1- yl)- 2- hydroxyethanesulfonamide 314. LCMS-ESI (m/z) calculated for C23H24N404S2: 484.5; found 485.1 [M+H]+, tK = 3.55 min. 1H NMR (400 MHz, CDC13) δ 8.33 (d, J = 7.6 Hz, 1H), 8.27 (d, J - 2.2 Hz, 1H), 8.15 (dd, J = 8.9, 2.3 Hz, 1H), 7.60 (d, J = 7.6 Hz, 1H), 7.43 (t, J = 7.7 Hz, 1H), 7.10 (d, J = 9.0 Hz, 1H), 5.08 (dd, J = 16.2, 7.8 Hz, 1H), 4.78 (dt, J - 12.1, 6.1 Hz, 1H), 4.64 (d, J - 8.9 Hz, 1H), 4.19 - 4.12 (m, 2H), 3.66 (ddd, J= 17.5, 8.8, 3.8 Hz, 1H), 3.40 (td, J- 4.8, 1.6 Hz, 2H), 3.38 - 3.26 (m, 1H), 2.77 - 2.67 (m, 1H), 2.10 - 1.99 (m, 1H), 1.47 (d, J = 6.1 Hz, 6H). 13C NMR (101 MHz, CDC13) δ 185.02, 173.71, 162.34, 143.58, 143.02, 133.41, 133.25, 130.15, 129.52, 127.71, 126.43, 123.55, 1 13.91, 104.14, 94.69, 72.81, 59.15, 57.63, 55.96, 35.13, 31.88, 21.91.
[0730] (i?)-N-(4-(5-(3-cyano-4-isopropoxyphenyl)-l,2,4-thiadiazol-3-yl)-2,3-dihydro-lH- inden-l-yl)-2-hydroxyethanesulfonamide can be prepared in a similar fashion using (R)- methyl 2-(N-(4-(5-(3-cyano-4-isopropoxyphenyl)- l,2,4-thiadiazol-3-yl)- 2,3-dihydro-lH- inden- 1 -yl)sulfamoyl)acetate.
[0731] Selected compounds and their corresponding analytical data are shown in Table 5, where the LCMS data was collected using Method 2 (see General Methods). The enantiomeric purity was determined for key intermediates and selected final compounds and is presumed from the synthesis for the remaining compounds. [0732] Selected compounds and their corresponding analytical data is shown in Table 1, where the LCMS data was collected using Method 2 (see General Methods). The enantiomeric purity was determined for key intermediates and selected final compounds and is presumed from the synthesis for the remaining compounds.
TABLE 1
Figure imgf000184_0001
5.98
7.82
7.78
6.18
6.18
8.68
8.70
6.43
Figure imgf000186_0001
ı85
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
209
Figure imgf000211_0001
210
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Biological Assays
Assay Procedures
Generation of SIP i-mediated inhibition of cAMP reporter assay
[0733] A mammalian expression plasmid containing SIPj/EDGl cloned into pcDNA3.1 was purchased from Missouri S&T cDNA Resource Centre. The nucleotide and amino acid sequence of human SIPj/EDGl are published in Hla and Maciag (J Biol Chem, 265(1990), 9308-9313). SlP!/pcDNA3.1 was transfected into the CRE-bla CHO Kl (Invitrogen) cell line, and stable single cell clones were selected using standard techniques. Expression of functional SlPi/EDGl receptor was confirmed by cell surface FACS with a SI Pi antibody (R&D Systems, clone 218713) and S IP-mediated inhibition of Forskolin induced cAMP. SIP] CRE-bla CHOKl reporter assay - characterization of SIP/ agonists
[0734] Cells were seeded into 384-well black wall/clear bottom plates at 104 cells/well/19.5 μΐ assay media (DMEM-phenol free, 0.5% charcoal/dextran stripped serum, 2 mM glutamine, 0.1 mM NEAA, 1 mM Na-Pyruvate, 25 mM Hepes) and incubated for 18 hrs at 37°C in 5% C02. Dose response curves (10-point) were generated in 10 mM Hepes, 0.1% Pluronic F127, in the presence of Forskolin. Cells were treated with 0.5 μΐ compound in the presence of 2 μΜ Forskolin for 4 hrs at 37°C. The FRET-based β-lactamase fluorescent substrate (LiveBLAzer™-FRET B/G Loading Kit CC4-AM; Invitrogen) was prepared according to manufacturer's directions, and incubated with cells for 2 hrs at room temperature. Plates were read at Ex:410/Em:458 and Ex:410/Em:522, and the response ratio determined. Data was analyzed by non-linear regression to determine the EC50 for inhibition of Forskolin induced cAMP.
Specificity over other SIP receptors
[0735] To assess compound specificity on other SIP receptors the following cell lines were used: S1P2 CRE-bla CHOKl, SlP3-Gal5 NFAT-bla HEK293T (Invitrogen), SlP4-bla TANGO U20S (Invitrogen), SlP5-bla TANGO U20S (Invitrogen). The same assay set up for SIPi was used but without Forskolin. S1P4 and S1P assays were performed in FreeStyle Expression medium (Invitrogen). S1P5 cells were incubated for 48 hrs in prior to treatment with compound.
Reported SIPi Activity
[0736] Activity data for selected SIPi agonists is displayed in Table 2. The activity range is denoted as follows: I I I I denotes agonist activity <0.05 nM. +++ denotes agonist activity between 0.05 to 0.50 nM, and ++ denotes agonist activity between 0.50-5.00 nM, and + denotes agonist activity > 5.00 nM. N/A denotes not available.
TABLE 2
Figure imgf000224_0001
Figure imgf000224_0002
++ 163 -H-+
M i l 164 +++
+++ 165 ++
+++ 166 +++
+++ 167 +++
+++ 168 +++
++ 169 ++
++ 170 ++
+++ 171 +++
+++ 172 +++
++ 173 ++
+++ 174 +++
++ 175 ++
++ 176 ++
++ 177 +++
++ 178 +++
+++ 179 ++
++ 180 +
++ 181 +
+++ 182 ++
+++ 183 ++
+++ 184 +
+-H- 185 +
+++ 186 +++
++ 187 ++
+++ 188 ++
++++ 189 +
-H-+ 190 ++
+++ 191 +
+++ 192 +
+++ 193 +
++ 194 ++
++ 195 ++ +++ 196 ++
+++ 197 ++ l i 1 1 198 +++
+++ 199 +++
+++ 200 +++
1 1 1 1 201 +++
+++ 202 ++
+++ 203 ++
1 1 1 1 204 ++
1 1 1 1 205 ++
++ 206 +++
+++ 207 ++
+++ 208 ++
+++ 209 ++
++ 210 ++
+++ 211 ++
++ 212 +
+++ 213 ++
++ 214 ++
-t-H- 215 ++
++ 216 ++
++ 217 +
+++ 218 +
++ 219 +++
++ 220 +++
++ 221 +++
+++ 222 +
+++ 223 ++
++ 224 +
+++ 225 +
+++ 226 ++
++ 227 +
++ 228 ++ 72 ++ 229 ++
73 +++ 230 ++
74 -H-+ 231 +++
75 +++ 232 ++
76 ++ 233 ++
77 +++ 234 ++
78 ++ 235 ++
79 +++ 236 ++
80 -H-+ 237 +++
81 ++ 238 ++
82 ++ 239 ++
83 + 240 +++
84 + 241 ++
85 + 242 ++
86 + 243 +
87 +++ 244 ++
88 ++ 245 +++
89 ++ 246 +++
90 ++ 247 ++
91 ++ 248 +++
92 ++ 249 ++
93 ++ 250 ++
94 ++ 251 ++
95 ++ 252 +++
96 +++ 253 ++
97 +++ 254 ++
98 ++ 255 +++
99 ++ 256 ++
100 +++ 257 +
101 + 258 ++
102 +++ 259 +++
103 +++ 260 ++
104 ++ 261 + 105 + 262 ++
106 + 263 ++
107 ++ 264 ++
108 + 265 ++
109 +-+ 266 +++
110 ++ 267 ++
111 +++ 268 ++
112 +++ 269 ++
113 +++ 270 ++
114 +++ 271 ++
115 +++ 272 ++
116 ++ 273 ++
117 +++ 274 ++
118 +++ 275 +++
119 +++ 276 +++
120 ++ 277 +++
121 ++ 278 -HH-
122 ++ 279 +++
123 ++ 280 ++
124 ++ 281 +++
125 ++ 282 ++
126 ++ 283 +++
127 ++ 284 ++
128 + 285 ++
129 + 286 4++
130 ++ 287 +++
131 ++ 288 +++
132 ++ 289 +++
133 ++ 290 +++
134 ++ 291 +++
135 + 292 +++
136 + 293 +++
137 + 294 +++ 138 + 295 +++
139 + 296 +++
140 + 297 ++
141 ++ 298 +++
142 ++ 299 ++
143 ++ 300 +++
144 ++ 301 -t-H-
145 + 302 +++
146 ++ 303 +++
147 ++ 304 ++
148 ++ 305 +++
149 ++ 306 +++
150 ++ 307 ++
151 + 308 ++
152 + 309 -H-+
153 + 310 +++
154 +++ 311 ++
155 ++ 312 ++
156 +++ 313 +++
157 +++ 314 ++
[0737] S I P1-SIP5 data for specific compounds is presented in Table 3. The agonist values (EC50) are reported in nM.
[0738] Table 3
Figure imgf000229_0001
172 0.19 > 10000 >10000 > 10000 203
186 0.41 > 10000 > 10000 > 10000 126
In Vivo Assays
Determination of absolute oral bioavailability in rats.
[0739J All pharmacokinetic studies were conducted in non-fasted female Sprague- Dawely rats (Simonsen Laboratories or Harlan Laboratories). Rats were housed in an ALAAC accredited facility and the research was approved by the facilities Institutional Animal Care and Use Committee (IACUC). The animals were acclimated to the laboratory for at least 48 h prior to initiation of experiments.
[0740] Compounds were formulated in 5%DMSO/5%Tween20 and 90% purified water (intravenous infusion) or 5%DMSO/5%Tween20 and 90% 0.1N HCL (oral gavage). Depending upon the solubility properties of the compound, alternate oral formulations were used (e.g. 0.5% carboxymethylcellulose). The concentration of the dosing solutions was verified by HPLC-UV. For intravenous dosing, compounds were administered by an infusion pump into the jugular vein over one minute to manually restrained animals (n=4 rats/compound). Oral dosing was by gavage using a standard stainless steel gavage needle (n=2-4 rats/compound). For both routes of administration, blood was collected at eight time- points after dosing with the final sample drawn 24 h post dose. Aliquots of the blood and/or plasma samples were transferred to polypropylene 96-well plate and frozen at -20°C until analysis.
[0741] After thawing the blood and/or plasma samples at room temperature, 5μΙ, of DMSO was added to each well. Proteins were precipitated by adding 150 μL acetonitrile containing 200 nM internal standard (4-hydroxy-3-(alpha-iminobenzyl)-l-methyl-6- phenylpyrindin-2-(lH)-one) and 0.1% formic acid. Plates were mixed for 1 min on a plate shaker to facilitate protein precipitation and then centrifuged at 3,000 rpm for 10 min to pellet protein. The supernatant was transferred to a clean plate and centrifuged at 3,000 rpm for 10 min to pellet any remaining solid material prior to LC MS/MS analysis. Calibration curve standards were prepared by spiking 5μΙ. compound stock in DMSO into freshly collected EDTA rat blood. An eight point standard curve spanning a range of 5 nM to 10,000 nM was included with each bio-analytical run. The standards were processed identically to the rat pharmacokinetic samples. [0742] Concentrations in the rat pharmacokinetic samples were determined using a standardized HPLC-LC/MS/MS method relative to the eight point standard curve. The system consisted of a Leap CTC Pal injector, Agilent 1200 HPLC with binary pump coupled with an Applied Biosystems 3200 QTrap. Compounds were chromatographed on a Phenomenex Synergy Fusion RP 20x2mm 2um Mercury Cartridge with Security Guard. A gradient method was used with mobile phase A consisting of 0.1% formic acid in water and mobile phase B consisting of 0.1% formic acid in acetonitrile at flow rates varying from 0.7 to 0.8 mL/min. Ions were generated in positive ionization mode using an electrospray ionization (ESI) interface. Multiple reaction monitoring (MRM) methods were developed specific to each compound. The heated nebulizer was set at 325°C with a nebulizer current of 4.8 μΑ. Collision energies used to generate daughter ions ranged between 29 and 39 V. Peak area ratios obtained from MRM of the mass transitions specific for each compound were used for quantification. The limit of quantification of the method was typically 5 nM. Data were collected and analyzed using Analyst software version 1.4.2.
[0743] Blood and/or plasma concentration versus time data were analyzed using non- compartmental methods (WinNonlin version 5.2; model 200 for oral dosing and model 202 for intravenous infusion). Absolute oral bioavailability (%) was calculated using the following expression: (Oral AUC χ IV Dose)/(IV AUC * Oral Dose)x 100.
Lymphopenia
[0744] In mice: Female C57BL6 mice (Simonsen Laboratories, Gilroy CA) were housed in an ALAAC accredited facility and the research was approved by the facilities Institutional Animal Care and Use Committee (IACUC). The animals were acclimated to the laboratory for at least 5 days prior to initiation of experiments. Mice (n=3/compound/time-point) were dosed by oral gavage with 1 mg/kg compound formulated in a vehicle consisting of 5%DMSO/5%Tween 20 and 90% 0.1N HC1. Control mice were dosed PO with the vehicle. Terminal whole blood samples were collected from isoflurane anesthetized mice by cardiac puncture into EDTA. Whole blood was incubated with rat anti-mouse CD16/CD32 (Mouse BD Fc Block, #553141), PE-Rat anti-mouse CD45R/B220 (BD #553089), APC-Cy7-Rat anti-mouse CD8a (BD #557654), and Alexa Fluor647-Rat anti-mouse CD4 (BD #557681) for 30 min on ice. Red blood cells were lysed using BD Pharm Lyse Lysing buffer (#555899) and white blood cells were analyzed by FACS. Lymphopenia was expressed as the % of white blood cells that were CD4 or CD8 positive T cells. The overall lymphopenia response over 24 h was estimated by calculating the area under the effect curve (AUEC) using the linear trapezoidal rule.
[0745] In rats: Female rats (Simonsen Laboratories, Gilroy CA) were housed in an ALAAC accredited facility and the research was approved by the facilities Institutional Animal Care and Use Committee (IACUC). The animals were acclimated to the laboratory for at least 5 days prior to initiation of experiments. Rats (n=3/compound/time-point) were dosed by oral gavage with 1 mg/kg compound formulated in a vehicle consisting of 5%DMSO/5%Tween 20 and 90% 0.1N HCL. Control rats were dosed PO with the vehicle. Whole blood was collected from isoflurane anesthetized rats via the retro-orbital sinus and terminal samples were collected by cardiac puncture into EDTA. Whole blood was incubated with mouse anti-rat CD32 (BD #550271), PE-mouse anti-rat CD45R/B220 (BD #554881), PECy5-mouse anti-rat CD4 (BD #554839), and APC-mouse anti-rat CD8a (eBioscience #17- 0084) for 30 minutes on ice. Red blood cells were lysed using BD Pharm Lyse Lysing buffer (#555899) and white blood cells were analyzed with a BD FACSArray. Lymphopenia was expressed as the % of white blood cells that were CD4 or CD8 positive T cells. The overall lymphopenia response over 24 h was estimated by calculating the area under the effect curve (AUEC) using the linear trapezoidal rule. In some experiments, total lymphocyte counts were determined using a standard impediance based veterinary hematology analyzer (IDEXX Preclinical Research Services, Sacramento, CA).
[0746] Rat lymphopenia data for specific compounds is presented in Table 4. The percentage of lymphopenia at 24 h after a 0.2 mg kg single dose regiment is reported. The estimated dose needed to produce 50% lymphopenia (ED50) at 24 h after a 3-5 day dosing regimen is also reported. N/A is not available.
[0747] Table 4
Figure imgf000232_0001
172 22 0.20
186 25 0.20
Evaluation of Therapeutic Index in Rats
[0748] Studies may be conducted in non-fasted male and female Sprague-Dawely rats (Simonsen Laboratories). Rats may be housed in an AAALAC accredited facility and the research can be approved by the facilities Institutional Animal Care and Use Committee (IACUC). The animals should be acclimated to the laboratory for at least 5 days prior to initiation of experiments.
[0749] The compounds may be formulated as suspensions in a vehicle consisting of 0.5% carboxymethyl cellulose (Acres Organics) in purified water (pH adjusted to ~2.2 with hydrochloric acid). The same formulation is used in the rat lymphopenia and toxicology studies described below. The concentration of each compound in suspension should be verified to be within ± 10% of the target concentration by HPLC-UV.
[0750] Prior to the conduct of toxicology studies, the effect of three to five daily doses of each compound on peripheral T-cell counts of female rats may be determined (see lymphopenia measurements in rats above). In these lymphopenia studies, blood samples are collected onto EDTA at intervals after the final study dose. The collection times need not be identical for each study, however, all studies may include a sample collected 24 hours after the final dose.. The lymphopenia data is used as a biomarker to select equally pharmacologically active doses for the subsequent toxicology study. The low dose for the toxicology study is the dose of each compound that resulted in a 50% reduction of T-cell count 24 h after the final dose in the lymphopenia study relative to vehicle treated rats.
[0751] In the toxicology studies, three male and three female rats per group are assigned to dosing groups using body weight based randomization. A control group in each study receives vehicle. All animals are dosed orally by gavage on 5 or 14-consecutive days at a dose volume of 5 mL/kg/day. The animals are observed daily for any manifestations of adverse effect. Twenty-four hours after the final study dose, the rats are anesthetized with isoflurane and a terminal blood sample is taken by intra-cardiac puncture for hematology and clinical chemistry evaluation (IDEXX Laboratories, Sacramento, CA). The lungs with trachea are collected, weighed, and then prepared for histology by perfusion with 10% neutral buffered formalin via the trachea. The internally fixed lungs are then preserved in 10% neutral buffered formalin and submitted for histological examination (IDEXX).
[0752] The dose of each compound resulting in a 10% increase in the lung to terminal body weight ratio can be estimated for each compound by linear interpolation. The therapeutic index can then be estimated as the ratio of the dose producing 10% lung weight increase to the dose producing 50% T-Cell depletion.
Description of the TNBS Crohn 's Colitis Model in Rats
[0753] Male Sprague-Dawley rats (180-200 g) are acclimatized for seven days and then assigned to 8 rats per group so that each group has approximately the same mean weight. Twenty-four hours prior to disease initiation, rats are deprived of food. Rats are anaesthetized and weighed, then 80 mg/kg TNBS solution (50% TNBS: 50% 200 proof ethanol) is instilled into colon via a 20g feeding needle inserted into the anus. The rats are maintained in head down position until recovery from anesthesia. Daily oral dosing is initiated 2 h post TNBS- instillation for six days. Prednisolone serves as a positive control and is administered orally daily at 10 mg/kg. Body weights are monitored daily and 24 h after the last dose, all groups are terminated. The colon is removed, flushed of fecal matter and examined for gross changes including strictures, adhesions and ulcers. The colon length, weight of the distal 2 cm, and wall thickness is recorded.
Description of Influenza A H1N1 Model in Mice
[0754] Male C57B1/6 (6-8 weeks of age) may be acclimatized for seven days and then assigned to 5-8 mice per group so that each group has approximately the same mean weight. Mice may be infected with 104 PFUs mouse-adapted influenza A virus (A/WSN/33) via the intra-tracheal route. Mice may then be treated with 0.2-1.5 mg/kg compound p.o. 1 hr postinfection. Forty eight hours after infection mice may be euthanized by cervical dislocation and bronchoalveolar lavage fluid can be collected. Quantitative cytokine analysis may be performed via ELISA. In some experiments whole body perfusion can be performed and lungs can be collected for cellular enumeration of inflammatory cells. Longevity studies may be performed by infection with 3-10xl04 PFUs mouse-adapted influenza A virus over 14 days.

Claims

We claim:
1. A compound of Formula (I) or a pharmaceutically acceptable salt, ester, prodrug, homolog, tautomer, stereoisomer, or hydrate, or solvate thereof:
Figure imgf000235_0001
(I) wherein
a dashed line signifies that a single bond or a double bond can be present, provided that there are two double bonds and three single bonds in the ring comprising A1, A2, and
A3;
A1, A2, and A3 each independently is CH or S or N; provided that one of A1, A2, and A3 is
S;
R1 is di-substituted phenyl or di-substituted pyridinyl where the phenyl and pyridinyl substituents are each independently selected from the group consisting of halo, nitro, cyano, perfluromethyl, fluorinated methyl, and Ci-4-alkoxy; provided that if R1 is di- substituted phenyl, such phenyl is para-substituted with Ci_4-alkoxy;
Figure imgf000235_0002
wherein a wavy line indicates a point of attachment; X is -NR'R" or -OR'"; R' is H, CM alkyl, n-hydroxy CM alkyl, -S02-R3, or -CO-R3;
R" is H, -SQ2-R5 , C alkyl optionally substituted with 1 or more R4, or a ring moiety optionally substituted with R6 wherein such ring moiety is pyridinyl, oxazolyl, piperidinyl, cyclohexyl, morpholinyl, thiazolyl, pyrazolyl, pyrrolidinyl, imidazolyl, or phenyl;
R'" is H, C alkyl, or -CO-R3 or R' and R" taken together with the nitrogen atom to which they are bound form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle is optionally singly or multiply substituted with substituents independently selected from the group consisting of -OH, oxo, -NH2, n-hydroxy-C1-4 alkyl, -COOH, -(CH2)m-COOH, - (CH2)m-COOR3, -N(R3R3), and -(CH2)m-CO-N(R7R7);
each R is independently C1-4 alkyl or H; each R4 is independently H, halo, OH, oxo, =NH, NH2, -COOH, F, -NHR3, - N(R7R7),-S02-R3, -S02-N(R7R7), -N(R3>-S02-R3, -COOR3, -OCO-R3, -CO-N(R7R7), -N(R3)-COR3, Ci-3 alkyl, C1-3 alkoxy, and a ring moiety optionally substituted with R6 wherein such ring moiety is pyridinyl, oxazolyl, piperazinyl, piperidinyl, morpholinyl, pyrrolidinyl, pyrazolyl, imidazolyl, benzimidazolyl, azetidinyl, cyclobutinyl, or phenyl; each R5 is independently R4, C alkyl, C -6 cycloalkyl, or C alkyl optionally substituted with 1 or more R4; each R6 is independently halo, OH, -NH2, -NHR3, -N(R3R3), -COOH, -COOR3, - NHCO-R3; each R7 is independently CM alkyl or H, or two R7 taken together with the nitrogen atom to which they are bound form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle is optionally substituted with -OH, - NH2, -N(R3R3), n-hydroxy C1-4 alkyl, -(CH2)m-COOH, -(CH2)m-COOR3; each m is independently 0, 1, 2, or 3.
2. A compound of Formula (I) or a pharmaceutically acceptable salt, ester, prodrug, homolog, tautomer, stereoisomer, or hydrate, or solvate thereof:
Figure imgf000237_0001
(I) wherein
a dashed line signifies that a single bond or a double bond can be present, provided that there are two double bonds and three single bonds in the ring comprising A1, A2, and A3;
A1, A2, and A3 each independently is CH or S or N; provided that one of A1, A2, and A3 is
S;
R1 is di-substituted phenyl or di-substituted pyridinyl where the phenyl and pyridinyl substituents are each independently selected from the group consisting of halo, nitro, cyano, perfluromethyl, fluorinated methyl, and Ci-4-alkoxy; provided that if R1 is di- substituted phenyl, such phenyl is para-substituted with d-4-alkoxy; 2 is
Figure imgf000237_0002
wherein a wavy line indicates a point of attachment; X is -NR'R" or -OR'";
R' is H, C1-4 alkyl, n-hydroxy C alkyl, -S02-R3, or -CO-R3; R" is H, -SO2-R5 , C alkyl optionally substituted with 1 or more R4, or a ring moiety optionally substituted with R wherein such ring moiety is piperidinyl, cyclohexyl, morpholinyl, thiazolyl, pyrazolyl, pyrrolidinyl, imidazolyl, or phenyl; R'" is H, CM alkyl, or -CO-R3 or R' and R" taken together with the nitrogen atom to which they are bound form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle is optionally singly or multiply substituted with substituents independently selected from the group consisting of -OH, oxo, -NH2, n-hydroxy-CM alkyl, -COOH, -(CH2)m-COOH, - (CH2)m-COOR3, -N(R3R3), and -(CH2)m-CO-N(R7R7);
each R is independently C alkyl or H; each R4 is independently H, halo, OH, oxo, =NH, NH2, -COOH, F, -NHR3, - N(R7R7),-S02-R3, -S02-N(R7R7), -N(R3>-S02-R3, -COOR3, -OCO-R3, -CO-N(R7R7), -N(R3)-COR3, Ci-3 alkyl, Ci-3 alkoxy, and a ring moiety optionally substituted with R6 wherein such ring moiety is piperazinyl, piperidinyl, morpholinyl, pyrrolidinyl, pyrazolyl, imidazolyl, benzimidazolyl, azetidinyl, cyclobutinyl, or phenyl; each R5 is independently R4, C alkyl, C3-6 cycloalkyl, or C1-4 alkyl optionally substituted with 1 or more R4; each R6 is independently halo, OH, -NH2, -NHR3, -N(R3R3), -COOH, -COOR3, - NHCO-R3; each R7 is independently CM alkyl or H, or two R7 taken together with the nitrogen atom to which they are bound form a 4, 5, or 6 membered saturated heterocyclic ring containing 0 or 1 additional heteroatoms where such additional heteroatom is O or N wherein such heterocycle is optionally substituted with -OH, - NH2, -N(R3R3), n-hydroxy CM alkyl, -(CH2)m-COOH, -(CH2)m-COOR3; each m is independently 0, 1, 2, or 3.
3. The compound of claim 1 or 2 wherein the structure of Formula I is selected from the group consisting of formulas a-i through a-x:
Figure imgf000239_0001
a-i a-ii a-iii a-iv
Figure imgf000239_0002
a-v a-vi a-vii a-viii
Figure imgf000239_0003
a-ix a-x
4. The compound of claim 1 or 2 wherein A1 is S.
5. The compound of claim 1 or 2 wherein A2 is S.
6. The compound of claim 1 or 2wherein A3 is S.
7. The compound of claim 6 wherein A1 is N and A2 is CH or N.
8. The compound of claim 7 wherein A2 is CH.
9. The compound of claim 7 wherein A2 is N.
10. The compound of claims 1-9 wherein R1 is
Figure imgf000239_0004
C2-4 alkyl; and Y is -CN, -CI, -O-R3, or -CF3.
11. The compound of claim 10 wherein R3 is isopropyl or ethyl.
12. The compound of claims 10-11 wherein Y is -CN or -0-C2H5.
13. The compound of claims 1-12 wherein R is
Figure imgf000240_0001
14. The compound of claims 1-12 wherein R2 is
Figure imgf000240_0002
15. The compound of claim 13 wherein R is
Figure imgf000240_0003
Figure imgf000241_0001
17. The compound of claim 15 wherein R2 is
Figure imgf000241_0002
18. The compound of claim 15 wherein R2 is
Figure imgf000241_0003
19. The compound of claim 16 wherein R2 is
Figure imgf000241_0004
20. The compound of claim 16 wherein R2 is
Figure imgf000242_0001
The com ound of claim 14 having the following formula:
Figure imgf000242_0002
wherein
A2 is CH or N;
Rm is H, -S02-R5, or C 1 -4 alkyl optionally substituted with 1 or more R4; each R4 is independently H, OH, oxo, NH2, -COOH, S02-R3, -N(R3>-S02-R3, - COOR3, -CO-N(R7R7), N(R7R7), C1-3 alkyl, C1-3 alkoxy; or a ring moiety selected from pyridinyl, oxazolyl, pyrrolidinyl, or imidazolyl.
22. The compound of claim 21 having the following formula:
Figure imgf000242_0003
23. The compound of claim 21 having the following formula:
Figure imgf000243_0001
24. The compound of claims 17-20, 22 and 23 wherein the compound is substantially enantiomerically pure.
25. The compound of claims 1-24 wherein the compound has an EC5o as an SIP receptor subtype 1 agonist which is at least ten times smaller than its EC50 as an agonist of a mutant SIP receptor subtype 1 having a single mutation with respect to wild type SIP receptor subtype 1 such that the 101st amino acid residue is changed from asparagine to alanine.
26. The compound of claim 25 wherein the compound has an EC50 as an SIP receptor subtype 1 agonist which is at least twenty times smaller than its EC50 as an agonist of a mutant form of S 1 P receptor subtype 1 having a single mutation with respect to wild type SIP receptor subtype 1 such that the 101st amino acid residue is changed from asparagine to alanine.
27. The compound of claims 1-26 wherein the compound has a therapeutic index of at least 5 as measured in rats following 5 or 14 days of dosing with the compound where the therapeutic index is the ratio of the dose achieving less than or equal to 10% increase in lung to terminal body weight at the conclusion of such 5 or 14 days and the dose achieving 50% lymphopenia.
28. The compound of claim 27 wherein the therapeutic index is at least 10.
29. The compound of claim 27 wherein the therapeutic index is at least 20.
30. The compound of claims 27-29 wherein the therapeutic index for the compound is greater than the therapeutic index for the enantiomer of the compound.
31. The compound of claim 30 wherein the therapeutic index for the compound is at least 150% of the therapeutic index for the enantiomer of the compound.
32. The compound of claims 1-20 and 24-31 wherein Y is CI.
33. The compound of claims 1-20 and 24-31 wherein Y is CF3.
34. The compound of claims 1-20 and 24-31 wherein Y is CN.
35. The compound of claims 1 -20 and 24-34 wherein X is -NR'R".
36. The compound of claims 1-20 and 24-34 wherein X is -OR'".
37. The compound of claim 36 wherein X is -OH.
38. The compound of claim 36 wherein X is -OCO-R .
39. The compound of claim 38 wherein R3 is C1-3 alkyl.
40. The compound of claim 35 wherein R' is H.
41. The compound of claim 35 wherein R' is -COR .
42. The compound of claim 35 wherein R' is -S02-R .
43. The compound of claim 35 wherein R" is H.
44. The compound of claim 35 wherein R" is -S02-R5.
45. The compound of claim 35 wherein R" is C alkyl optionally substituted with 1 or more R4.
46. The compound of claim 35 wherein R" is -{CR^^n-R4 ; each RA and each RB is independently selected from the group consisting of H, hydroxyl and methyl or RA and RB bound to the same carbon taken together are oxo; and n is 0, 1 , 2, or 3.
47. The compound of claim 46 wherein n is 2.
48. The compound of claim 47 wherein R4 is -OH, -NH2, -NHR3, -N(R7R7), or -COOH.
49. The compound of claim 44 wherein R5 is C alkyl optionally substituted with 1 or more R .
50. The compound of claim 44 wherein Y is CN.
51. The compound of claim 49 wherein R5 is -C2H5-N((R7R7) or -CH2-CO-N(R7R7).
52. The compound of claim 50 wherein R is C2¾-0-R .
53. The compound of claim 36 wherein X is -NH-CO-N(R7R7).
54. The compound of claim 21 -31 wherein A is N.
55. The compound of claim 54 wherein RM is -S02-R5.
56. The compound of claim 55 wherein R5 is NH2, or C alkyl optionally substituted with 1 or more R4.
57. The compound of claim 56 wherein R4 is OH, C1-3 alkoxy, -COOH, -COOR3 , CO- N(R7R7) or N(R7R7).
58. The compound of claim 54 wherein RM is C1-4 alkyl optionally substituted with 1 or more R4.
59. The compound of claim 58 wherein at least one of the 1 or more R4 is oxo.
60. The compound of claim 59 wherein one of the 1 or more R4 is OH, or C1-3 alkoxy.
61. The compound of claims 1, 2 and 24-31 wherein the compound is selected from compounds 1-227:
Figure imgf000245_0001
Figure imgf000246_0001
245
Figure imgf000247_0001
246
Figure imgf000248_0001
247
Figure imgf000249_0001
248
Figure imgf000250_0001
Figure imgf000250_0002
249
Figure imgf000251_0001
Figure imgf000251_0002
250
Figure imgf000252_0001
Figure imgf000252_0002
Figure imgf000252_0003
Figure imgf000252_0004
Figure imgf000252_0005
251
Figure imgf000253_0001
Figure imgf000253_0002
Figure imgf000253_0003
Figure imgf000253_0004
252
Figure imgf000254_0001
Figure imgf000254_0002
Figure imgf000254_0003
Figure imgf000254_0004
Figure imgf000254_0005
253
. The method of claims 76-77 wherein the compound comprising an indane moiety provided in step (i) is contacted with the chiral reagent to form in step (iii) Formula VI-R or VI-S:
Figure imgf000255_0001
wherein Z is CI, Br or I.
79. The method of claim 78 wherein the method further comprises the step of protecting the hydroxy group of Formula VI-R or VI-S by treating Formula VI-R or VI-S with a protecting agent to form Formula Vla-R or Vla-S:
Figure imgf000255_0002
O-PG via-R O-PG via-S wherein PG is a protecting group.
80. The method of claim 79 wherein the protecting agent is TBSC1.
81. The method of claims 79-80 wherein the method further comprises the step of reacting Formula VTa-R or Vla-S with boronic acid or bis(pinacolato)diboron to form a boronic acid or boronate ester of Formula Vlb-R or Vlb-S:
Figure imgf000255_0003
82. The method of claim 75 wherein the chiral reagent is RS(=0)NH2 and the compound comprising an indane moiety is enantiomerically enriched with respect to a carbon- nitrogen bond on a ring carbon of the five-membered ring of the indane moiety.
83. The method of claim 82 wherein the chiral reagent is t-Bu-S(=0)NH2.
278
84. The method of claims 82-83 wherein the compound comprising an indane moiety provided in step (i) is contacted with the chiral reagent to form in step (ii) Formula VII:
Figure imgf000256_0001
wherein Z is CI, Br or I.
85. The method of claims 82-84 wherein a compound of Formula VIII-R or VIII-S is formed in step (iii):
Figure imgf000256_0002
86. The method of claim 85 wherein the method further comprises the step of contacting Formula VIII-R or VIII-S with 1,4-dioxane in the presence of an acid to form Formula VIb-R or VIb-S or Formula IX-R or IX-S:
Figure imgf000256_0003
87. The method of claims 86 wherein the method further comprises the step of protecting the amino group by treating Formula IX-R or IX-S with a protecting agent to form Formula IXa-R or IXa-S:
279
Figure imgf000257_0001
88. The method of claims 87 wherein the protecting agent is di-/ert-butyldicarbonate.
89. The method of claims 87-88 wherein the method further comprises the step of reacting Formula IXa-R or IXa-S with boronic acid or bis(pinacolato)diboron to form a boronic acid or boronate ester of Formula IXb-R or IXb-S:
Figure imgf000257_0002
. The method of claims 81 or 89 wherein the method further comprises the step of reacting Formula VIb-R, Formula VTb-S, Formula IXb-R or Formula IXb-S with Formula XI to form Formula XH-R or XII-S:
Figure imgf000257_0003
to form Formula XII-R or XII-S:
Figure imgf000257_0004
X PG xii.R X-PG XII.S
1 2 1
wherein each A and each A is independently N, or CH; R is di-substituted phenyl or di-substituted pyridinyl where the phenyl and pyridinyl substituents are each independently selected from the group consisting of halo, nitro, cyano, perfluromethyl, fluorinated methyl, and
Figure imgf000257_0005
provided that if R1 is di-
280 substituted phenyl, such phenyl is para-substituted with Ci- -alkoxy; and X is NH or O.
91. The method of claim 90 wherein R1 is di-substituted phenyl where the phenyl substituents are F and Y, wherein Y is -CN, -CI, or -CF3.
92. The method of claim 91 wherein the method further comprises the step of reacting Formula XII-R or XII-S with /PrOH in the presence of NaOz'Pr to from Formula XIII-R or XIII-S:
Figure imgf000258_0001
93. The method of claim 92 wherein the method further comprises the step of deprotecting the hydroxyl group wherein X is O, or the amino group wherein X is NH, by treating Formula XIII-R or XIII-S with a deprotecting agent.
94. The method of claim 93 wherein the method further comprises the step of converting the deprotected amino group to a secondary amine.
95. The method of claims 91-94 wherein Y is CN.
96. The method of claims 90-95 wherein A1 is N and A2 is N.
97. The method of claim 96 wherein Formula XI is prepared following the process comprising the step of a) treating a di-substituted benzaldehyde with potassium phosphate monobasic to form a di-substituted benzoic acid; b) contacting the di-substituted benzoic acid with H2NNHCSNH2 to form an amino- 1,3-4-thiadizole having a di-substituted phenyl group substituted on the thiadiazole moiety; and
281 c) treating the amino-l,3-4-thiadizole in step b) with a mixture of copper bromide and isoamylnitrite.
98. The method of claims 90-95 wherein A1 is N and A2 is CH.
99. The method of claim 98 wherein Formula XI is prepared following the process comprising the step of a) contacting 2-bromothiazole with a (di-substituted phenyl)boronic acid to form a 2- (di-substituted phenyl)thiazole; and b) treating the 2-(di-substituted phenyl)thiazole with NBS.
100. The method of claims 90-95 wherein A1 is CH and A2 is N.
101. The method of claim 100 wherein Formula XI is prepared following the process comprising the step of a) contacting 5-(tributylstannyl)thiazole with an iodobenzene having two other substituents to form a 5-(di-substituted phenyl)thiazole; and b) treating the 2-(di-substituted phenyl)thiazole with NBS.
102. The method of claims 75-101 wherein a compound of claims 1-13, 15, 17, 18 and 25-53 is prepared.
103. A method for the synthesis of a compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six-membered saturated ring of the tetrahydronaphthalene moiety where the compound is enantiomerically enriched with respect to the chiral carbon, the method comprising the steps of
(i) providing a compound comprising a tetrahydronaphthalene moiety where the ring carbon of the six-membered saturated ring of the tetrahydronaphthalene moiety where chiral substitution is desired is oxo substituted at such carbon and where a ring carbon of the phenyl ring of the tetrahydronaphthylene moiety is bromo substituted;
(ii) reacting such compound with a chiral reagent to form a chiral center at the tetrahydronaphthalene moiety carbon previously bound to the oxo group.
282
104. The method of claim 103 wherein the chiral reagent is RuCl( -cymene)[(/?,R)-Ts- DPEN] or RuCl(p-cymene)[(.S'(5)-Ts-DPEN].
105. The method of claims 103-104 wherein the compound comprising a tetrahydronaphthalene moiety provided in step (i) is contacted with the chiral reagent to form in step (ii) Formula XTV-R or XTV-S:
Figure imgf000260_0001
106. The method of claim 105 wherein the method further comprises the step of reversing the chiral configuration of the chiral carbon in the six-membered saturated ring of the tetrahydronaphthalene moiety that was previously bound to the oxo group by treating Formula XTV-R or XTV-S with diphenylphosphoryl azide (DPP A) to form an azido tetrahydronaphthalene of Formula XV-S or XV-R:
Figure imgf000260_0002
xv-R, where the azido substituent in the six-membered saturated ring of the tetrahydronaphthalene moiety replacing the hydroxy substituent and the chiral carbon that is bound to the azido substituent has a reverse chiral configuration of the chiral carbon when it was previously bound to the hydroxy substituent.
107. The method of claim 106 wherein the method further comprises the step of converting the azido tetrahydronaphthalene of Formula XV-S or XV-R to an amine substituted tetrahydronaphthalene of Formula XVI-S or XVI-R:
Figure imgf000260_0003
wherein Xm is NH-PG, NH2 or NR'R"; R' is H, C alkyl, n-hydroxy C alkyl, -S02- R3, or -CO-R3; R" is H, -S02-R5 , C alkyl optionally substituted with 1 or more R4, or a ring moiety optionally substituted with R6 wherein such ring moiety is pyridinyl,
283 oxazolyl, piperidinyl, cyclohexyl, morpholinyl, thiazolyl, pyrazolyl, pyrrolidinyl, imidazolyl, or phenyl; and PG is a protecting group.
108. The method of claim 107 wherein Xm is NH-PG.
109. The method of claim 108 wherein the step of converting comprises contacting the azido tetrahydronaphthalene of Formula XV-S or XV-R with a carbonate ester.
110. The method of claim 109 wherein the carbonate ester is di-tert-butyl dicarbonate.
111. The method of claim 107 wherein the method further comprises the step of reacting Formula XVI-S or XVI-R with a boronic acid to form a boronic acid of Formula XVII-S or XVII-R:
Figure imgf000261_0001
112. The method of claim 111 wherein the boronic acid is pinacolborane.
113. The method of claim 112 wherein the method further comprises the step of forming a substituted 1,3,4-thiadiazole on the tetrahydronaphthalene moiety by reacting an intermediate of XVII-S or XVII-R with a substituted 2-bromo- 1,3,4-thiadiazole of Formula XVTV and a coupling reagent to form a compound of Formula XW-S or XW-R:
Figure imgf000261_0002
wherein Ym is F or -ORa; and Ra is lower alkyl.
114. The method of claim 113 wherein the compound of Formula XW-S or XVV-R have the structures below:
284
Figure imgf000262_0001
115. The method of claims 113-114 wherein the coupling reagent comprises a palladium.
116. The method of claims 113-115 wherein the coupling reagent comprises tetrakis (triphenylphosphine) palladium.
117. The method of claim 103 wherein the compound provided in step (i) is
Figure imgf000262_0002
118. The method of claims 103-117 wherein the resulting compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six-membered saturated ring of the tetrahydronaphthalene moiety is enantiomerically enriched at least 90%.
119. The method of claim 118 wherein the resulting compound is enantiomerically enriched at least 95%.
120. The method of claim 118 wherein the resulting compound is enantiomerically enriched at least 98%.
121. The method of claim 118 wherein the resulting compound is enantiomerically enriched at least 99%.
122. A compound selected from the group consisting of:
285
Figure imgf000263_0001
. A method for the synthesis of a compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six-membered saturated ring of the
286 tetrahydronaphthalene moiety where the compound is enantiomerically enriched with respect to the chiral carbon, the method comprising a step of providing a compound of claim 122.
. A method of claims 103-121 where the compound comprising a tetrahydronaphthalene moiety having a chiral carbon in the six-membered saturated ring of the tetrahydronaphthalene moiety where the compound is enantiomerically enriched with respect to the chiral carbon is a compound of claims 1-12, 14, 16, 19, 20-31 and 54-60, the method comprising a step of providing a compound of claim 122.
287
PCT/US2012/037609 2011-05-13 2012-05-11 Selective heterocyclic sphingosine 1 phosphate receptor modulators WO2012158550A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2014510511A JP6129159B2 (en) 2011-05-13 2012-05-11 Selective heterocyclic sphingosine 1-phosphate receptor modulator
ES12785231T ES2758841T3 (en) 2011-05-13 2012-05-11 Selective heterocyclic sphingosine-1-phosphate receptor modulators
US14/117,514 US9481659B2 (en) 2011-05-13 2012-05-11 Selective heterocyclic sphingosine 1 phosphate receptor modulators
EP12785231.7A EP2706999B1 (en) 2011-05-13 2012-05-11 Selective heterocyclic sphingosine 1 phosphate receptor modulators

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161486184P 2011-05-13 2011-05-13
US61/486,184 2011-05-13

Publications (3)

Publication Number Publication Date
WO2012158550A2 true WO2012158550A2 (en) 2012-11-22
WO2012158550A3 WO2012158550A3 (en) 2014-05-01
WO2012158550A9 WO2012158550A9 (en) 2017-10-26

Family

ID=47177578

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/037609 WO2012158550A2 (en) 2011-05-13 2012-05-11 Selective heterocyclic sphingosine 1 phosphate receptor modulators

Country Status (5)

Country Link
US (1) US9481659B2 (en)
EP (1) EP2706999B1 (en)
JP (1) JP6129159B2 (en)
ES (1) ES2758841T3 (en)
WO (1) WO2012158550A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9115054B2 (en) 2013-02-21 2015-08-25 Bristol-Myers Squibb Company Tetrahydronaphthalenyl compounds useful as sipi agonists
CN106187945A (en) * 2016-07-22 2016-12-07 辽宁师范大学 A kind of synthetic method of organic synthesis intermediate
WO2018157813A1 (en) * 2017-02-28 2018-09-07 南京明德新药研发股份有限公司 Spiro compound and use thereof
EP3492465A4 (en) * 2016-07-22 2020-03-04 Shijiazhuang Sagacity New Drug Development Co., Ltd. S1p1 agonist and application thereof
WO2020053334A1 (en) * 2018-09-12 2020-03-19 Pharmazell Gmbh A process for the preparation of ozanimod and its intermediate (s)-1-amino-2,3-dihydro-1h-indene-4-carbonitrile
WO2021047553A1 (en) 2019-09-09 2021-03-18 上海长森药业有限公司 Aromatic heterocyclic compound having tricyclic structure, and preparation method therefor and application thereof
WO2021175223A1 (en) * 2020-03-04 2021-09-10 南京明德新药研发有限公司 Benzo 2-azaspiro[4.4]nonane compound and use thereof
WO2021228000A1 (en) 2020-05-11 2021-11-18 上海长森药业有限公司 Preparation of biaryl ring-linked aromatic heterocyclic derivative as immunomodulator and use thereof
WO2022213935A1 (en) * 2021-04-09 2022-10-13 南昌弘益药业有限公司 Oxadiazole-substituted spirocyclic compound and application thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA024801B1 (en) 2009-11-13 2016-10-31 Рецептос Ллк Selective sphingosine 1 phosphate receptor modulators

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040058894A1 (en) * 2002-01-18 2004-03-25 Doherty George A. Selective S1P1/Edg1 receptor agonists
US7605171B2 (en) * 2003-12-17 2009-10-20 Merck & Co., Inc. (3,4-disubstituted)propanoic carboxylates as S1P (Edg) receptor agonists
US20100010001A1 (en) * 2008-05-14 2010-01-14 Edward Roberts Novel modulators of sphingosine phosphate receptors

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1479544A (en) 1974-02-07 1977-07-13 American Cyanamid Co 1,2,3,4-tetrahydro-1-naphthylurea derivatives their preparation and their use
FR2628103B1 (en) 1988-03-03 1991-06-14 Roussel Uclaf NOVEL PYRETHRINOID ESTERS CARRYING AN INDANYL CORE, THEIR PREPARATION PROCESS AND THEIR APPLICATION AS PESTICIDES
US5039802A (en) 1990-04-18 1991-08-13 Merck & Co., Inc. Arylation process for preparation of chiral catalysts for ketone reduction
EP0583346A1 (en) 1991-04-12 1994-02-23 Schering Corporation Bicyclic amides as inhibitors of acyl-coenzyme a: cholesterol acyl transferase
GB2290790A (en) 1994-06-30 1996-01-10 Merck & Co Inc Asymmetric synthesis of 6-substituted 2-amino-1,2,3,4-tetrahydronaphthalenes
DE69911176T2 (en) 1998-01-23 2004-06-24 Sankyo Co., Ltd. Spiropiperidine DERIVATIVES
AU2003297232B2 (en) 2002-12-20 2010-02-04 Merck Sharp & Dohme Corp. 1-(amino)indanes and (1,2-dihydro-3-amino)-benzofurans, benzothiophenes and indoles
ES2770035T3 (en) 2003-04-11 2020-06-30 Ptc Therapeutics Inc 1,2,4-Oxadiazole benzoic acid compound and its use for senseless suppression and treatment of diseases
PE20050420A1 (en) * 2003-04-21 2005-06-13 Elan Pharm Inc PHENACILO 2-HYDROXY-3-DIAMINOALKANOS
CN1859908A (en) 2003-10-01 2006-11-08 默克公司 3,5-aryl, heteroaryl or cycloalkyl substituted-1,2,4-oxadiazoles as S1P receptor agonists
US7585881B2 (en) 2004-02-18 2009-09-08 Astrazeneca Ab Additional heteropolycyclic compounds and their use as metabotropic glutamate receptor antagonists
US20060173183A1 (en) 2004-12-31 2006-08-03 Alantos Pharmaceuticals, Inc., Multicyclic bis-amide MMP inhibitors
JP2008531546A (en) 2005-02-22 2008-08-14 テバ ファーマシューティカル インダストリーズ リミティド An improved process for the synthesis of enantiomeric indanylamine derivatives
KR100667075B1 (en) 2005-07-22 2007-01-10 삼성에스디아이 주식회사 Scan driver and organic electroluminescence display device of having the same
EP1924546A1 (en) * 2005-09-14 2008-05-28 Amgen, Inc Conformationally constrained 3- (4-hydroxy-phenyl) - substituted-propanoic acids useful for treating metabolic disorders
EP1963509A4 (en) 2005-12-21 2009-07-29 Joseph Gabriele Catecholamine regulated protein
US20090163482A1 (en) 2006-03-13 2009-06-25 Mchardy Stanton Furst Tetralines antagonists of the h-3 receptor
US20080009534A1 (en) 2006-07-07 2008-01-10 Bristol-Myers Squibb Company Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
AU2007323540A1 (en) 2006-11-21 2008-05-29 University Of Virginia Patent Foundation Hydrindane analogs having sphingosine 1-phosphate receptor agonist activity
AU2007334436A1 (en) 2006-12-15 2008-06-26 Abbott Laboratories Novel oxadiazole compounds
JO2701B1 (en) 2006-12-21 2013-03-03 جلاكسو جروب ليميتد Compounds
JP2010513457A (en) * 2006-12-21 2010-04-30 ファイザー・プロダクツ・インク Compound having both angiotensin II receptor antagonism and PPARγ activation activity
WO2008106224A1 (en) 2007-02-28 2008-09-04 Rib-X Pharmaceuticals, Inc. Macrolide compounds and methods of making and using the same
WO2009048474A1 (en) * 2007-10-12 2009-04-16 Pharmacopeia, Inc. 2,7,9-substituted purinone derivatives for immunosuppression
US20090298894A1 (en) 2008-04-21 2009-12-03 Asahi Kasei Pharma Corporation Amino acid compounds
WO2010117662A1 (en) * 2009-03-30 2010-10-14 Exelixis, Inc. Modulators of s1p and methods of making and using
GB0910674D0 (en) 2009-06-19 2009-08-05 Glaxo Group Ltd Novel compounds
GB0910691D0 (en) * 2009-06-19 2009-08-05 Glaxo Group Ltd Novel compounds
WO2011005290A1 (en) 2009-06-23 2011-01-13 Arena Pharmaceuticals, Inc. Disubstituted oxadiazole derivatives useful in the treatment of autoimmune and inflammatory disorders
GB0911130D0 (en) 2009-06-26 2009-08-12 Glaxo Group Ltd Novel compounds
EA024801B1 (en) 2009-11-13 2016-10-31 Рецептос Ллк Selective sphingosine 1 phosphate receptor modulators
CN102724880B (en) * 2009-11-13 2016-09-14 瑞塞普托斯有限责任公司 S1P receptor modulators and Chiral Synthesis
EP2498609B1 (en) 2009-11-13 2018-04-18 Celgene International II Sàrl Selective heterocyclic sphingosine 1 phosphate receptor modulators
WO2013055907A1 (en) 2011-10-12 2013-04-18 Teva Pharmaceutical Industries Ltd. Treatment of multiple sclerosis with combination of laquinimod and fingolimod

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040058894A1 (en) * 2002-01-18 2004-03-25 Doherty George A. Selective S1P1/Edg1 receptor agonists
US7605171B2 (en) * 2003-12-17 2009-10-20 Merck & Co., Inc. (3,4-disubstituted)propanoic carboxylates as S1P (Edg) receptor agonists
US20100010001A1 (en) * 2008-05-14 2010-01-14 Edward Roberts Novel modulators of sphingosine phosphate receptors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GONZALEZ-CABRERA ET AL.: 'Full Pharmacoiogical Efficacy of a Novel S1P1 Agonist That Does Not Require S1 P-Like Headgroup Interactions' MOLECULAR PHARMACOLOGY vol. 74, no. 5, November 2008, pages 1308 - 1318, XP009123605 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9359286B2 (en) 2013-02-21 2016-06-07 Bristol-Myers Squibb Company Bicyclic compounds
US9487481B2 (en) 2013-02-21 2016-11-08 Bristol-Myers Squibb Company Bicyclic compounds
US9115054B2 (en) 2013-02-21 2015-08-25 Bristol-Myers Squibb Company Tetrahydronaphthalenyl compounds useful as sipi agonists
EP3492465A4 (en) * 2016-07-22 2020-03-04 Shijiazhuang Sagacity New Drug Development Co., Ltd. S1p1 agonist and application thereof
CN106187945A (en) * 2016-07-22 2016-12-07 辽宁师范大学 A kind of synthetic method of organic synthesis intermediate
US11192886B2 (en) 2016-07-22 2021-12-07 Medshine Discovery Inc. S1P1 agonist and application thereof
WO2018157813A1 (en) * 2017-02-28 2018-09-07 南京明德新药研发股份有限公司 Spiro compound and use thereof
CN110325517B (en) * 2017-02-28 2021-03-02 南京明德新药研发有限公司 Spiro compound and application thereof
CN110325517A (en) * 2017-02-28 2019-10-11 南京明德新药研发有限公司 Volution compound and its application
WO2020053334A1 (en) * 2018-09-12 2020-03-19 Pharmazell Gmbh A process for the preparation of ozanimod and its intermediate (s)-1-amino-2,3-dihydro-1h-indene-4-carbonitrile
WO2021047553A1 (en) 2019-09-09 2021-03-18 上海长森药业有限公司 Aromatic heterocyclic compound having tricyclic structure, and preparation method therefor and application thereof
WO2021175223A1 (en) * 2020-03-04 2021-09-10 南京明德新药研发有限公司 Benzo 2-azaspiro[4.4]nonane compound and use thereof
CN115279740A (en) * 2020-03-04 2022-11-01 南昌弘益药业有限公司 Benzo 2-azaspiro [4.4] nonane compound and application thereof
US11760751B2 (en) 2020-03-04 2023-09-19 Helioeast Science & Technology Co., Ltd. Benzo 2-azaspiro[4.4]nonane compound and use thereof
WO2021228000A1 (en) 2020-05-11 2021-11-18 上海长森药业有限公司 Preparation of biaryl ring-linked aromatic heterocyclic derivative as immunomodulator and use thereof
WO2022213935A1 (en) * 2021-04-09 2022-10-13 南昌弘益药业有限公司 Oxadiazole-substituted spirocyclic compound and application thereof

Also Published As

Publication number Publication date
US9481659B2 (en) 2016-11-01
JP2014517836A (en) 2014-07-24
WO2012158550A3 (en) 2014-05-01
EP2706999B1 (en) 2019-08-28
JP6129159B2 (en) 2017-05-17
EP2706999A2 (en) 2014-03-19
WO2012158550A9 (en) 2017-10-26
US20150299179A1 (en) 2015-10-22
EP2706999A4 (en) 2015-07-15
ES2758841T3 (en) 2020-05-06

Similar Documents

Publication Publication Date Title
DK2498609T3 (en) SELECTIVE HETEROCYCLIC SPHINGOSIN-1 PHOSPHATRECEPTOR MODULATORS
EP3406142B1 (en) Selective sphingosine 1 phosphate receptor modulators and methods of chiral synthesis
EP2706999B1 (en) Selective heterocyclic sphingosine 1 phosphate receptor modulators
DK2498611T3 (en) SPHINGOSIN-1-PHOSPHATE RECEPTOR MODULATORS AND METHODS FOR CHIRAL SYNTHESIS
WO2015066515A1 (en) Selective sphingosine 1 phosphate receptor modulators and combination therapy therewith
AU2015202660B2 (en) Selective sphingosine 1 phosphate receptor modulators and methods of chiral synthesis

Legal Events

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

Ref document number: 12785231

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase in:

Ref document number: 2014510511

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase in:

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2012785231

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 14117514

Country of ref document: US