WO2017192813A1 - Inhibitors of indoleamine 2,3-dioxygenase and methods of their use - Google Patents

Inhibitors of indoleamine 2,3-dioxygenase and methods of their use Download PDF

Info

Publication number
WO2017192813A1
WO2017192813A1 PCT/US2017/030997 US2017030997W WO2017192813A1 WO 2017192813 A1 WO2017192813 A1 WO 2017192813A1 US 2017030997 W US2017030997 W US 2017030997W WO 2017192813 A1 WO2017192813 A1 WO 2017192813A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
fluoroquinolin
cealkyl
aspects
acetamide
Prior art date
Application number
PCT/US2017/030997
Other languages
French (fr)
Inventor
David K. Williams
Weifang Shan
Liping Zhang
Emily Charlotte CHERNEY
James Aaron Balog
Original Assignee
Bristol-Myers Squibb Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to JP2018557904A priority Critical patent/JP2019516687A/en
Priority to US16/098,245 priority patent/US11066383B2/en
Priority to EP17793316.5A priority patent/EP3452451A4/en
Priority to CN201780040617.8A priority patent/CN109311816A/en
Priority to KR1020187034654A priority patent/KR20190004742A/en
Publication of WO2017192813A1 publication Critical patent/WO2017192813A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/12Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D215/14Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/18Halogen atoms or nitro radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • C07D215/42Nitrogen atoms attached in position 4
    • C07D215/46Nitrogen atoms attached in position 4 with hydrocarbon radicals, substituted by nitrogen atoms, attached to said nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • the invention relates generally to compounds that modulate or inhibit the enzymatic activity of indoleamine 2,3-dioxygenase (IDO), pharmaceutical compositions containing said compounds and methods of treating proliferative disorders, such as cancer, viral infections and/or autoimmune diseases utilizing the compounds of the invention.
  • IDO indoleamine 2,3-dioxygenase
  • IDO Indoleamine 2,3-dioxygenase
  • IDOl is an IFN- ⁇ target gene that plays a role in immunomodulation.
  • IDO is an oxidoreductase and one of two enzymes that catalyze the first and rate-limiting step in the conversion of tryptophan to N-formyl- kynurenine. It exists as a 41kD monomer that is found in several cell populations, including immune cells, endothelial cells, and fibroblasts. IDO is relatively well-conserved between species, with mouse and human sharing 63% sequence identity at the amino acid level.
  • IDO plays a major role in immune regulation, and its immunosuppressive function manifests in several manners. Importantly, IDO regulates immunity at the T cell level, and a nexus exists between IDO and cytokine production. In addition, tumors frequently manipulate immune function by upregulation of IDO. Thus, modulation of IDO can have a therapeutic impact on a number of diseases, disorders and conditions.
  • Treatment of cancer commonly entails surgical resection followed by chemotherapy and radiotherapy.
  • the standard treatment regimens show highly variable degrees of long-term success because of the ability of tumor cells to essentially escape by regenerating primary tumor growth and, often more importantly, seeding distant metastasis.
  • Recent advances in the treatment of cancer and cancer-related diseases, disorders and conditions comprise the use of combination therapy incorporating immunotherapy with more traditional chemotherapy and radiotherapy. Under most scenarios, immunotherapy is associated with less toxicity than traditional chemotherapy because it utilizes the patient's own immune system to identify and eliminate tumor cells.
  • IDO In addition to cancer, IDO has been implicated in, among other conditions, immunosuppression, chronic infections, and autoimmune diseases or disorders (e.g., rheumatoid arthritis). Thus, suppression of tryptophan degradation by inhibition of IDO activity has tremendous therapeutic value. Moreover, inhibitors of IDO can be used to enhance T cell activation when the T cells are suppressed by pregnancy, malignancy, or a virus (e.g., HIV). Although their roles are not as well defined, IDO inhibitors may also find use in the treatment of patients with neurological or neuropsychiatric diseases or disorders (e.g., depression).
  • a virus e.g., HIV
  • IDO inhibitors of IDO have been developed to treat or prevent IDO- related diseases.
  • the IDO inhibitors 1-methyl-DL-tryptophan; p-(3-benzofuranyl)- DL-alanine; p-[3-benzo(b)thienyl]-DL-alanine; and 6-nitro-L-tryptophan have been used to modulate T cell-mediated immunity by altering local extracellular concentrations of tryptophan and tryptophan metabolites (WO 99/29310).
  • Compounds having IDO inhibitory activity are further reported in PCT Publication No. WO 2004/094409.
  • the invention is also directed to pharmaceutical compositions comprising one or more compounds of formula I and/ or formula II.
  • the invention is also directed to methods of treating cancer using one or more compounds of formula I and/ or formula II.
  • X is CH or N. In some aspect, X is CH. In other aspects, X is N.
  • T is CH or N. In some aspects, T is CH. In other aspects, T is N.
  • X is CH and T is CH.
  • X is N and T is CH. In other embodiments, X is CH and T is N. In other embodiments, X is N and T is N.
  • R 1 is H, halo, or Ci-Cehaloalkyl.
  • R 1 is H, halo, Ci-C 6 alkyl, or Ci-Cehaloalkyl.
  • R 1 is H.
  • R 1 is halo (F, CI, Br, or I), preferably F.
  • R 1 is Ci-Cealkyl, for example, methyl, ethyl, isopropyl, butyl, and t-butyl.
  • R 1 is Ci-Cehaloalkyl, for example, -CF 3 .
  • R 1 is halo, Ci-C6alkyl, or Ci-Cehaloalkyl.
  • R 1 is halo or Ci- Cehaloalkyl.
  • the R 1 when R 1 is halo, Ci-C 6 alkyl, or Ci-Cehaloalkyl, the R 1 is preferably present at the 4-carbon position of the ring. In other aspects of the disclosure comprising compounds of formula I, when R 1 is halo, Ci- Cealkyl, or Ci-Cehaloalkyl, the R 1 can be present at the 2-carbon position of the ring. Preferably, in those aspects of the disclosure comprising compounds of formula I, when R 1 is F or CF 3 at the 4-carbon position of the ring.
  • R 1 when R 1 is halo, Ci-C 6 alkyl, or Ci-Cehaloalkyl, the R 1 is preferably present at the 2-carbon position of the ring. In other aspects of the disclosure comprising compounds of formula II, when R 1 is halo, Ci-C 6 alkyl, or Ci-Cehaloalkyl, the R 1 can be present at the 3-carbon position of the ring.
  • R 1 is F or CF 3 at the 2-carbon position of the ring.
  • R 1A is H, halo, Ci-Cealkyl, or Ci-Cehaloalkyl.
  • R 1A is H.
  • R 1A is halo (F, CI, Br, or I).
  • R 1A is Ci-Cealkyl, for example, methyl, ethyl, isopropyl, butyl, and t-butyl.
  • R 1A is Ci-Cehaloalkyl, for example, -CF 3 .
  • Y is CH or N. In some aspects, Y is CH. In other aspect, Y is N. In other embodiments, Y is -C(Ci-C 6 alkyl), for example, -C(CH 3 ) or
  • n is 0, 1, 2, 3, or 4. In preferred aspects, n is 2. In other aspects, n is 0. In other aspects, n is 1. In yet other aspects, n is 3. In still other aspects, n is 4. In some aspects, n is 0 to 2 or 1 to 2. In yet other aspects, n is 1 to 3.
  • W is N or CR 4 , wherein R 4 is H or -(Ci-C 6 alkyl)-.
  • W is N.
  • W is CR 4 and R 4 is H, e.g, W is -CH-.
  • W is CR 4 and R 4 is -(Ci-Cealkyl), methyl, methyl, propyl, isopropyl, butyl, s-butyl, t- but l, and the like (i.e., W is -C(d-C 6 alkyl)-).
  • Y is CH and W is CH. In other aspects, Y is CH and W is N. In other aspects, Y is N and W is CH. In still other aspects, Y is N and W is N. In some aspects, Y is CH and W is C(Ci-C 6 alkyl). In other aspects, Y is N and W is C(Ci-C 6 alkyl).
  • V is unsubstituted Ci-Cealkylene, for example, unsubstituted Ci-Csalkylene, Ci-C4alkylene, Ci-C3alkylene, Ci-C2alkylene, or Cialkylene.
  • V is Ci-Cealkylene, for example, Ci-Csalkylene, Ci-C4alkylene, Ci-C 3 alkylene, Ci-C2alkylene, or Cialkylene, substituted with one, two, or three substituents, preferably 1 or 2 substituents, independently selected from Ci-Cealkyl (e.g., methyl, ethyl, isopropyl) and C 3 -C 6 cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl).
  • V is a Cialkylene substituted with one or two, preferably one, Ci-Cealkyl (e.g., methyl or ethyl) substituents.
  • R 2 is H or Ci-Cealkyl. In some aspects, R 2 is H. In other aspects, R 2 is Ci-Cealkyl, for example, methyl, ethyl, isopropyl, butyl, or t-butyl.
  • Z is unsubstituted Ci-Cealkylene, for example, unsubstituted Ci-Csalkylene, Ci-C4alkylene, Ci-C 3 alkylene, Ci-C2alkylene, or Cialkylene.
  • Z is Ci-Cealkylene, for example, Ci-Csalkylene, Ci-C4alkylene, Ci-C 3 alkylene, Ci-C2alkylene, or Cialkylene, substituted with one, two, or three substituents, preferably 1 or 2 substituents, independently selected from Ci-Cealkyl (e.g., methyl, ethyl, isopropyl) and C 3 -C 6 cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl).
  • Ci-Cealkyl e.g., methyl, ethyl, isopropyl
  • C 3 -C 6 cycloalkyl e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
  • Z is a Cialkylene substituted with one or two Ci-Cealkyl (e.g., methyl or ethyl) substituents.
  • Z is preferably Cialkylene substituted with one C 3 - Cecycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl).
  • Z is a bond, e.g., Z is absent.
  • B is unsubstituted aryl, for example, unsubstituted phenyl or naphthyl.
  • B is aryl substituted with 1, 2, or 3 substituents, preferably 1 or 2 substituents.
  • the B substituents which can be referred to as one or more R groups, are preferably independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi-C 6 alkyl, -OCi-Cehaloalkyl, -CN, aryl, or -Oaryl.
  • Preferred substituted aryl moieties include phenyl and naphthyl, with phenyl being particularly preferred.
  • the aryl is substituted with at least one substituent that is -OH.
  • the aryl is substituted with at least one substituent that is Ci-Cealkyl, for example, methyl, ethyl, propyl, isopropyl, butyl, s-butyl, t-butyl, pentyl, or hexyl.
  • the aryl is substituted with at least one substituent that is Ci-Cehaloalkyl, for example, -CF 3 .
  • the aryl is substituted with at least one substituent that is halo, for example, F, CI, or Br, with F and CI substituents being particularly preferred aryl substituents.
  • the aryl is substituted with at least one substituent that is -OCi-Cealkyl, for example, methoxy, ethoxy, propoxy, isoproxy, butoxy, or t-butoxy.
  • the aryl is substituted with at least one substituent that is -OCi-Cehaloalkyl, for example, -OCF 3 .
  • the aryl is substituted with at least one substituent that is -CN.
  • the aryl is substituted with at least one substituent that is another aryl moiety, for example, a phenyl or naphthyl moiety.
  • the aryl is substituted with at least one substituent that is -Oaryl, for example, -Ophenyl.
  • B is unsubstituted C 3 - C cycloalkyl, for example, C 3 cycloalkyl (e.g., cyclopropyl), C4cycloalkyl (e.g., cyclobutyl), CiCycloalkyl (e.g., cyclopentyl), Cecycloalkyl (e.g., cyclohexyl), Cycycloalkyl (e.g., bicyclo[2.2.1]heptanyl), Cgcycloalkyl (bicyclo[2.2.2]octanyl), Cgcycloalkyl, Ciocycloalkyl (e.g., adamantyl), Cncycloalkyl, or Cncycloalkyl.
  • B is unsubstituted C3- Cecycloalkyl.
  • B is unsubstituted C7-Ci2cycloalkyl or C7-Ci
  • B is C 3 -Ci2cycloalkyl substituted with 1 , 2, or 3 substituents, preferably 1 or 2 substituents.
  • B is substituted C 3 cycloalkyl, C4cycloalkyl, Cscycloalkyl, Cecycloalkyl, Cycycloalkyl, Cgcycloalkyl, Cgcycloalkyl,
  • Ciocycloalkyl, Cncycloalkyl, or Cncycloalkyl is substituted C 3 - Cecycloalkyl. In other aspects, B is substituted C7-Ci2cycloalkyl or C7-Ciocycloalkyl.
  • the B substituents which can be referred to as one or more R groups, are preferably independently selected from -OH, Ci-C 6 alkyl, Ci-C 6 haloalkyl, halo, -OCi-C 6 alkyl, -OCi-C 6 haloalkyl, -CN, aryl, or -Oaryl.
  • C 3 -Ci2cycloalkyl moieties include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo[2.2.1]heptanyl, bicyclo[2.2.2]octanyl, and adamantyl.
  • the C 3 -Ci2cycloalkyl is substituted with at least one substituent that is -OH.
  • the C 3 -Ci2cycloalkyl is substituted with at least one substituent that is Ci-Cealkyl, for example, methyl, ethyl, propyl, isopropyl, butyl, s-butyl, t-butyl, pentyl, or hexyl.
  • the C3-Ci2cycloalkyl is substituted with at least one substituent that is Ci-Cehaloalkyl, for example, -CF 3 .
  • the C3-Ci2cycloalkyl is substituted with at least one substituent that is halo, for example, F, CI, or Br, with F and CI substituents being particularly preferred C 3 -Ci2cycloalkyl substituents.
  • the C 3 -Ci2cycloalkyl is substituted with at least one substituent that is -OCi-Cealkyl, for example, methoxy, ethoxy, propoxy, isoproxy, butoxy, or t-butoxy.
  • the C 3 -Ci2cycloalkyl is substituted with at least one substituent that is -OCi-Cehaloalkyl, for example, -OCF 3 .
  • the C 3 -Ci2cycloalkyl is substituted with at least one substituent that is -CN.
  • the C 3 - Ci2cycloalkyl is substituted with at least one substituent that is an aryl moiety, for example, a phenyl or naphthyl moiety.
  • the C 3 -Ci2cycloalkyl is substituted with at least one substituent that is -Oaryl, for example, -Ophenyl.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I and II.
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is CR 4 , V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted aryl, for exam le,
  • X is CH, R is H or Ci-C 6 alkyl, R b is H, Ci-C 6 alkyl, or C 3 -C 6 cycloalkyl, and R bA is absent, H, or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I-A and II-A are those wherein X is N, R 5 is H or Ci-Cealkyl, R 6 is H, Ci-Cealkyl, or C 3 -C 6 cycloalkyl, and R ⁇ is absent, H, or Ci-Cealkyl.
  • X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-A and II-A.
  • Sub-formulas of formula I or formula II include formulas wherein n is 2, Y is N, W is CR 4 , V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted aryl, for example,
  • X is CH, R is H or Ci-C 6 alkyl, R b is H, Ci-C 6 alkyl, or C 3 -C 6 cycloalkyl, and R bA is absent, H, or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I-B and II-B are those wherein X is N, R 5 is H or Ci-Cealkyl, R 6 is H, Ci-Cealkyl, or C 3 -C 6 cycloalkyl, and R 6A is absent, H, or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-B and II-B.
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is N, V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted aryl, for example,
  • X is CH, R is H or Ci-C 6 alkyl, R b is H, Ci-C 6 alkyl, or C 3 -C 6 cycloalkyl, and R bA is absent, H, or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I-C and II-C are those wherein X is N, R 5 is H or Ci-Cealkyl, R 6 is H, Ci-Cealkyl, or C 3 -C 6 cycloalkyl, and R 6 is absent, H, or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-C and II-C.
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is N, V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted aryl, for example,
  • X is CH, R is H or Ci-Cealkyl (e.g., methyl), R is H, Ci-Cealkyl, or C3-Cecycloalkyl, and R 6A is absent, H, or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I- D and II-D are those wherein X is N, R 5 is H or Ci-C 6 alkyl, R 6 is H, Ci-C 6 alkyl, or C 3 - Cecycloalkyl, and R 6A is absent, H, or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts,
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is CR 4 , V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted C 3 -Ci2cycloalkyl, for exam le,
  • X is CH, R is H or Ci-C 6 alkyl, R b is H, Ci-C 6 alkyl, or C 3 -C 6 cycloalkyl, and R bA is absent, H, or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I-E and II-E are those wherein X is N, R 5 is H or Ci-Cealkyl, R 6 is H, Ci-Cealkyl, or C 3 -C 6 cycloalkyl, and R f is absent, H, or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-E and II-E.
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is CR 4 , V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted C 3 - C cycloalkyl, for exam le,
  • X is CH, R is H or Ci-C 6 alkyl, R b is H, Ci-C 6 alkyl, or C 3 -C 6 cycloalkyl, and R bA is absent, H, or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I-F and II-F are those wherein X is N, R 5 is H or Ci-Cealkyl, R 6 is H, Ci-Cealkyl, or C3-C 6 cycloalkyl, and R ⁇ is absent, H, or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-F and II-F.
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is N, V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted C3- Ci2cycloalkyl, for ex mple,
  • X is CH and T is N. In other aspects, X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-G and II-G.
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is N, V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted C 3 - Ci2cycloalkyl for exam le,
  • X is CH, R is H or Ci-Cealkyl (e.g., methyl), R is H, Ci-Cealkyl, or C3-C 6 cycloalkyl, and R 6A is absent, H, or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I- H and II-H are those wherein X is N, R 5 is H or Ci-C 6 alkyl, R 6 is H, Ci-C 6 alkyl, or C 3 - Cecycloalkyl, and R 6A is absent, H, or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts,
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is CR 4 , V is Cialkylene, R 2 is H, Z is a bond, and B is substituted or unsubstituted C3- C cycloalkyl, for example,
  • X is CH, and R is H or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I-J and II-J are those wherein X is N and R 5 is H or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-J and II-J.
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is CR 4 , V is Cialkylene, R 2 is H, Z is a bond, and B is substituted or unsubstituted C 3 - C cycloalkyl, for example,
  • X is CH an R is H or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I-K and II-K are those wherein X is N and R 5 is H or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-K and II-K.
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is N, V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted C 3 - C cycloalkyl, for example,
  • X is CH and R is H or Ci-Cealkyl.
  • Other preferred embodiments of compounds of formulas I-L and II-L are those wherein X is N and R 5 is H or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-L and II-L.
  • Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is N, V is Cialkylene, R 2 is H, Z is Cialkylene, and B is substituted or unsubstituted C 3 - Ci2cycloalkyl for example,
  • X is CH and R is H or Ci-Cealkyl (e.g., methyl)
  • Other preferred embodiments of compounds of formulas I-M and II-M are those wherein X is N and R 5 is H or Ci-Cealkyl.
  • X is CH and T is CH.
  • X is N and T is CH.
  • X is CH and T is N.
  • X is N and T is N.
  • the invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-M and II- M.
  • the compounds of the invention have human IDO IC5 0 values > 50 nM. In another embodiment, the compounds of the invention have human IDO IC5 0 values ⁇ 50 nM. In another embodiment, the compounds of the invention have human IDO IC5 0 values ⁇ 5 nM.
  • the present invention provides a composition comprising one or more compounds of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and at least one of the compounds of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
  • the present invention provides a pharmaceutical composition, comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
  • the present invention provides a process for making a compound of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
  • the present invention provides an intermediate for making a compound of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
  • the present invention provides a method for the treatment and/or prophylaxis of various types of cancer, viral infections and/or autoimmune diseases, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of one or more compounds of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent, such as a chemotherapeutic agent or a signal transductor inhibitor.
  • a therapeutically effective amount of one or more compounds of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent, such as a chemotherapeutic agent or a signal transductor inhibitor.
  • the present invention provides a compound of the present invention, and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof, for use in therapy.
  • the present invention provides a combined preparation of a compound of the present invention, and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof, and additional therapeutic agent(s) for simultaneous, separate or sequential use in therapy.
  • the present invention provides a combined preparation of a compound of the present invention, and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof, and additional therapeutic agent(s) for simultaneous, separate or sequential use in the treatment and/or prophylaxis of multiple diseases or disorders associated with the enzymatic activity of IDO.
  • the invention provides a method of treating a patient suffering from or susceptible to a medical condition that is sensitive to enzymatic activity of IDO.
  • a number of medical conditions can be treated.
  • the method comprises administering to the patient a therapeutically effective amount of a composition comprising a compound described herein and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof.
  • the compounds described herein may be used to treat or prevent viral infections, proliferative diseases (e.g. , cancer), and autoimmune diseases.
  • the compounds and pharmaceutical compositions of the present invention are useful in treating or preventing any disease or conditions that are sensitive to enzymatic activity of IDO. These include viral and other infections (e.g., skin infections, GI infection, urinary tract infections, genito-urinary infections, systemic infections), proliferative diseases (e.g. , cancer), and autoimmune diseases (e.g. , rheumatoid arthritis, lupus).
  • the compounds and pharmaceutical compositions may be administered to animals, preferably mammals (e.g. , domesticated animals, cats, dogs, mice, rats), and more preferably humans. Any method of administration may be used to deliver the compound or pharmaceutical composition to the patient. In certain embodiments, the compound or pharmaceutical composition is administered orally. In other embodiments, the compound or pharmaceutical composition is administered parenterally.
  • Compounds of the invention can modulate activity of the enzyme indoleamine- 2,3-dioxygenase (IDO).
  • IDO indoleamine- 2,3-dioxygenase
  • modulate is meant to refer to an ability to increase or decrease activity of an enzyme or receptor.
  • compounds of the invention can be used in methods of modulating IDO by contacting the enzyme with any one or more of the compounds or compositions described herein.
  • compounds of the present invention can act as inhibitors of IDO.
  • the compounds of the invention can be used to modulate activity of IDO in cell or in an individual in need of modulation of the enzyme by administering a modulating (e.g. , inhibiting) amount of a compound of the invention.
  • Compounds of the invention can inhibit activity of the enzyme indoleamine-2,3- dioxygenase (IDO).
  • IDO indoleamine-2,3- dioxygenase
  • the compounds of the invention can be used to inhibit activity of IDO in cell or in an individual in need of modulation of the enzyme by administering an inhibiting amount of a compound of the invention.
  • the present invention further provides methods of inhibiting the degradation of tryptophan in a system containing cells expressing IDO such as a tissue, living organism, or cell culture.
  • the present invention provides methods of altering (e.g., increasing) extracellular tryptophan levels in a mammal by administering an effective amount of a compound of composition provided herein. Methods of measuring tryptophan levels and tryptophan degradation are routine in the art.
  • the present invention further provides methods of inhibiting
  • immunosuppression such as IDO-mediated immunosuppression in a patient by administering to the patient an effective amount of a compound or composition recited herein.
  • IDO-mediated immunosuppression has been associated with, for example, cancers, tumor growth, metastasis, viral infection, and viral replication.
  • the present invention further provides methods of treating diseases associated with activity or expression, including abnormal activity and/or overexpression, of IDO in an individual (e.g., patient) by administering to the individual in need of such treatment a therapeutically effective amount or dose of a compound of the present invention or a
  • Example diseases can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the IDO enzyme, such as over expression or abnormal activity.
  • An IDO-associated disease can also include any disease, disorder or condition that can be prevented, ameliorated, or cured by modulating enzyme activity.
  • IDO-associated diseases include cancer, viral infection such as HIV infection, HCV infection, depression, neurodegenerative disorders such as Alzheimer's disease and Huntington's disease, trauma, age-related cataracts, organ transplantation (e.g., organ transplant rejection), and autoimmune diseases including asthma, rheumatoid arthritis, multiple sclerosis, allergic inflammation, inflammatory bowel disease, psoriasis and systemic lupus erythematosus.
  • an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal.
  • an in vitro cell can be a cell in a cell culture.
  • an in vivo cell is a cell living in an organism such as a mammal.
  • contacting refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • "contacting" the IDO enzyme with a compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having IDO, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the IDO enzyme.
  • IDO inhibitor refers to an agent capable of inhibiting the activity of indoleamine 2,3-dioxygenase (IDO) and thereby reversing IDO-mediated immunosuppression.
  • the IDO inhibitor may inhibit IDOl and/or ID02 (INDOL1).
  • An IDO inhibitor may be a reversible or irreversible IDO inhibitor.
  • a reversible IDO inhibitor is a compound that reversibly inhibits IDO enzyme activity either at the catalytic site or at a non-catalytic site and "an irreversible IDO inhibitor” is a compound that irreversibly destroys IDO enzyme activity.
  • Types of cancers that may be treated with the compounds of this invention include, but are not limited to, brain cancers, skin cancers, bladder cancers, ovarian cancers, breast cancers, gastric cancers, pancreatic cancers, prostate cancers, colon cancers, blood cancers, lung cancers and bone cancers.
  • cancer types include neuroblastoma, intestine carcinoma such as rectum carcinoma, colon carcinoma, familiar adenomatous polyposis carcinoma and hereditary non-polyposis colorectal cancer, esophageal carcinoma, labial carcinoma, larynx carcinoma, hypopharynx carcinoma, tongue carcinoma, salivary gland carcinoma, gastric carcinoma, adenocarcinoma, medullary thyroid carcinoma, papillary thyroid carcinoma, renal carcinoma, kidney parenchymal carcinoma, ovarian carcinoma, cervix carcinoma, uterine corpus carcinoma, endometrium carcinoma, chorion carcinoma, pancreatic carcinoma, prostate carcinoma, testis carcinoma, breast carcinoma, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, Hodgkin lymphoma, non-Hodgkin lymphoma, Burkitt lymphoma, acute lymphatic leuk
  • the invention provides a method of treating an autoimmune disease by providing to a patient in need thereof a compound or composition of the present invention.
  • autoimmune diseases include, but are not limited to, collagen diseases such as rheumatoid arthritis, systemic lupus erythematosus, Sharp's syndrome, CREST syndrome (calcinosis, Raynaud's syndrome, esophageal dysmotility, telangiectasia), dermatomyositis, vasculitis (Morbus Wegener's) and Sjogren's syndrome, renal diseases such as Goodpasture's syndrome, rapidly -progressing glomerulonephritis and membranoproliferative glomerulonephritis type II, endocrine diseases such as type-I diabetes, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), autoimmune parathyroidism, pernicious anemia,
  • collagen diseases such as rheumato
  • One or more additional pharmaceutical agents or treatment methods such as, for example, anti-viral agents, chemotherapeutics or other anticancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g. , IL2 and GM-CSF), and/or tyrosine kinase inhibitors can be optionally used in combination with the compounds of the present invention for treatment of IDO-associated diseases, disorders or conditions.
  • the agents can be combined with the present compounds in a single dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
  • Suitable chemotherapeutic or other anticancer agents include, for example, alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) such as uracil mustard, chlormethine,
  • cyclophosphamide CYTOXAN®
  • ifosfamide melphalan
  • chlorambucil pipobroman
  • triethylene-melamine triethylenethiophosphoramine
  • busulfan carmustine, lomustine, streptozocin, dacarbazine, and temozolomide.
  • suitable agents for use in combination with the compounds of the present invention include: dacarbazine (DTIC), optionally, along with other chemotherapy drugs such as carmustine (BCNU) and cisplatin; the "Dartmouth regimen", which consists of DTIC, BCNU, cisplatin and tamoxifen; a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOY®.
  • DTIC dacarbazine
  • BCNU carmustine
  • cisplatin the "Dartmouth regimen” which consists of DTIC, BCNU, cisplatin and tamoxifen
  • a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOY® a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOY®.
  • Compounds according to the invention may also be combined with immunotherapy drugs, including cytokines such as interferon
  • Anti-melanoma vaccines are, in some ways, similar to the anti-virus vaccines which are used to prevent diseases caused by viruses such as polio, measles, and mumps. Weakened melanoma cells or parts of melanoma cells called antigens may be injected into a patient to stimulate the body's immune system to destroy melanoma cells.
  • Melanomas that are confined to the arms or legs may also be treated with a combination of agents including one or more compounds of the invention, using a hyperthermic isolated limb perfusion technique.
  • This treatment protocol temporarily separates the circulation of the involved limb from the rest of the body and inj ects high doses of chemotherapy into the artery feeding the limb, thus providing high doses to the area of the tumor without exposing intemal organs to these doses that might otherwise cause severe side effects.
  • the fluid is warmed to 102° to 104° F.
  • Melphalan is the drug most often used in this chemotherapy procedure. This can be given with another agent called tumor necrosis factor (TNF).
  • TNF tumor necrosis factor
  • Suitable chemotherapeutic or other anticancer agents include, for example, antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatine, and gemcitabine.
  • antimetabolites including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
  • methotrexate including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
  • methotrexate including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
  • Suitable chemotherapeutic or other anticancer agents further include, for example, certain natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) such as vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, ara-C, paclitaxel (Taxol), mithramycin, deoxycoformycin, mitomycin-C, L-asparaginase, interferons (especially IFN-a), etoposide, and teniposide.
  • certain natural products and their derivatives for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins
  • vinblastine vincristine, vindesine
  • bleomycin dactinomycin
  • daunorubicin daunor
  • cytotoxic agents include navelbene, CPT-11 , anastrazole, letrazole, capecitabine, reloxafine, and droloxafine.
  • cytotoxic agents such as epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes such as cisplatin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors.
  • anticancer agent(s) include antibody therapeutics such as trastuzumab (HERCEPTIN®), antibodies to costimulatory molecules such as CTLA-4, 4-1BB and PD-1, or antibodies to cytokines (IL-10 or TGF- ⁇ ).
  • HERCEPTIN® antibodies to costimulatory molecules
  • CTLA-4 costimulatory molecules
  • 4-1BB antibodies to PD-1
  • cytokines IL-10 or TGF- ⁇
  • anticancer agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4.
  • anticancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer.
  • Anticancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses.
  • the pharmaceutical composition of the invention may optionally include at least one signal transduction inhibitor (STI).
  • STI signal transduction inhibitor
  • a "signal transduction inhibitor” is an agent that selectively inhibits one or more vital steps in signaling pathways, in the normal function of cancer cells, thereby leading to apoptosis.
  • Suitable STIs include, but are not limited to: (i) bcr/abl kinase inhibitors such as, for example, STI 571 (GLEEVEC®); (ii) epidermal growth factor (EGF) receptor inhibitors such as, for example, kinase inhibitors (IRESSA®, SSI-774) and antibodies (Imclone: C225 [Goldstein et al., Clin.
  • ABX-EGF her-2/neu receptor inhibitors such as farnesyl transferase inhibitors (FTI) such as, for example, L-744,832 (Kohl et al, Nat. Med., 1(8): 792-797 (1995));
  • FTI farnesyl transferase inhibitors
  • Akt family kinases
  • rapamycin see, for example, Sekulic et al, Cancer Res.
  • STI and at least one IDO inhibitor may be in separate pharmaceutical compositions.
  • At least one IDO inhibitor and at least one STI may be administered to the patient concurrently or sequentially.
  • at least one IDO inhibitor may be administered first, at least one STI may be administered first, or at least one IDO inhibitor and at least one STI may be administered at the same time.
  • the present invention further provides a pharmaceutical composition for the treatment of a chronic viral infection in a patient comprising at least one IDO inhibitor, optionally, at least one chemotherapeutic drug, and, optionally, at least one antiviral agent, in a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions may include at least one IDO inhibitor of the instant invention in addition to at least one established (known) IDO inhibitor.
  • at least one of the IDO inhibitors of the pharmaceutical composition is selected from the group consisting of compounds of formulas I and (II).
  • At least one IDO inhibitor and at least one chemotherapeutic agent may be administered to the patient concurrently or sequentially.
  • at least one IDO inhibitor may be administered first, at least one chemotherapeutic agent may be administered first, or at least one IDO inhibitor and the at least one STI may be administered at the same time.
  • the compounds may be administered in any order.
  • any antiviral agent or STI may also be administered at any point in comparison to the administration of an IDO inhibitor.
  • Chronic viral infections that may be treated using the present combinatorial treatment include, but are not limited to, diseases caused by: hepatitis C virus (HCV), human papilloma virus (HPV), cytomegalovirus (CMV), herpes simplex virus (HSV), Epstein-Barr virus (EBV), varicella zoster virus, Coxsackie virus, human immunodeficiency virus (HIV).
  • HCV hepatitis C virus
  • HPV human papilloma virus
  • CMV cytomegalovirus
  • HSV herpes simplex virus
  • EBV Epstein-Barr virus
  • varicella zoster virus Coxsackie virus
  • Coxsackie virus human immunodeficiency virus
  • the pharmaceutical compositions comprising at least one IDO inhibitor of the instant invention may be administered to a patient to prevent arterial restenosis, such as after balloon endoscopy or stent placement.
  • the pharmaceutical composition further comprises at least one taxane (e.g. , paclitaxel (Taxol); see, e.g. , Scheller et al, Circulation, 110: 810-814 (2004)).
  • Suitable antiviral agents contemplated for use in combination with the compounds of the present invention can comprise nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors and other antiviral drugs.
  • NRTIs nucleoside and nucleotide reverse transcriptase inhibitors
  • NRTIs non-nucleoside reverse transcriptase inhibitors
  • protease inhibitors and other antiviral drugs.
  • suitable NRTIs include zidovudine (AZT); didanosine (ddl);
  • zalcitabine (ddC); stavudine (d4T); lamivudine (3TC); abacavir (1592U89); adefovir dipivoxil [bis(POM)-PMEA]; lobucavir (BMS-180194); BCH-I0652; emtricitabine [(-)-FTC]; beta-L-FD4 (also called beta-L-D4C and named beta-L-2',3'-dicleoxy-5-fluoro-cytidene); DAPD, ((-)-beta- D-2,6-diamino-purine dioxolane); and lodenosine (FddA).
  • NNRTIs include nevirapine (BI-RG-587); delaviradine (BHAP, U-90152); efavirenz (DMP-266); PNU-142721; AG-1549; MKC-442 (l-(ethoxy-methyl)-5-(l-methylethyl)-6-(phenylmethyl)-(2,4(lH,3H)- pyrimidinedione); and (+)-calanolide A (NSC-675451) and B.
  • Suitable protease inhibitors include saquinavir (Ro 31-8959); ritonavir (ABT-538); indinavir (MK-639); nelfinavir (AG-1343); amprenavir (141W94); lasinavir (BMS-234475); DMP-450; BMS-2322623;
  • antiviral agents include hydroxyurea, ribavirin, IL-2, IL-12, pentafuside and Yissum Project No.11607.
  • immuno-oncology agents used herein, also known as cancer immunotherapies, are effective to enhance, stimulate, and/or upregulate immune responses in a subject.
  • the Compound of formula I or formula II is sequentially administered prior to administration of the immuno-oncology agent. In another aspect, the Compound of formula I or formula II is administered concurrently with the immunology- oncology agent. In yet another aspect, the Compound of formula I or formula II is sequentially administered after administration of the immuno-oncology agent.
  • the Compound of formula I or formula II may be co- formulated with an immuno-oncology agent.
  • Immuno-oncology agents include, for example, a small molecule drug, antibody, or other biologic or small molecule.
  • biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines.
  • the antibody is a monoclonal antibody. In another aspect, the monoclonal antibody is humanized or human.
  • the immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co- inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators).
  • an agonist of a stimulatory (including a co-stimulatory) receptor or an antagonist of an inhibitory (including a co- inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators).
  • an antagonist of an inhibitory (including a co- inhibitory) signal on T cells both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators).
  • Certain of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF).
  • B7 family includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7- H6.
  • TNF family of molecules that bind to cognate TNF receptor family members which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-lBB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fnl4, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTfiR, LIGHT, DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFRl, Lymphotoxin ⁇ / ⁇ , TNFR2, TNFa, LTfiR, Lymphotoxin a 1 ⁇
  • the immuno-oncology agent is a cytokine that inhibits T cell activation (e.g. , IL-6, IL-10, TGF- ⁇ , VEGF, and other immunosuppressive cytokines) or a cytokine that stimulates T cell activation, for stimulating an immune response.
  • a cytokine that inhibits T cell activation e.g. , IL-6, IL-10, TGF- ⁇ , VEGF, and other immunosuppressive cytokines
  • a cytokine that stimulates T cell activation for stimulating an immune response.
  • T cell responses can be stimulated by a combination of the Compound of formula I or formula II and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4, and (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-lBB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H.
  • an antagonist of a protein that inhibits T cell activation e.g., immune
  • Other agents that can be combined with the Compound of formula I or formula II for the treatment of cancer include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells.
  • the Compound of formula I or formula II can be combined with antagonists of KIR, such as lirilumab.
  • agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (WO 11/70024, WO 11/107553, WO 11/131407, WO 13/87699, WO 13/119716, WO 13/132044) or FPA-008 (WO 11/140249, WO 13/169264, WO 14/036357).
  • CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (WO 11/70024, WO 11/107553, WO 11/131407, WO 13/87699, WO 13/119716, WO 13/132044) or FPA-008 (WO 11/140249, WO 13/169264, WO 14/036357).
  • the Compound of formula I or formula II can be used with one or more of agonistic agents that ligate positive costimulatory receptors, blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency of anti-tumor T cells, agents that overcome distinct immune suppressive pathways within the tumor microenvironment (e.g. , block inhibitory receptor engagement (e.g. , PD-Ll/PD-1 interactions), deplete or inhibit Tregs (e.g. , using an anti-CD25 monoclonal antibody (e.g.
  • daclizumab or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell anergy or exhaustion) and agents that trigger innate immune activation and/or inflammation at tumor sites.
  • metabolic enzymes such as IDO, or reverse/prevent T cell anergy or exhaustion
  • the immuno-oncology agent is a CTLA-4 antagonist, such as an antagonistic CTLA-4 antibody.
  • CTLA-4 antibodies include, for example, YERVOY® (ipilimumab) or tremelimumab.
  • the immuno-oncology agent is a PD-1 antagonist, such as an antagonistic PD-1 antibody.
  • Suitable PD-1 antibodies include, for example, OPDIVO® (nivolumab), KEYTRUDA® (pembrolizumab), or MEDI-0680 (AMP-514; WO 2012/145493).
  • the immuno-oncology agent may also include pidilizumab (CT-011), though its specificity for PD-1 binding has been questioned.
  • Another approach to target the PD-1 receptor is the recombinant protein composed of the extracellular domain of PD-L2 (B7-DC) fused to the Fc portion of IgGl, called AMP-224
  • the immuno-oncology agent is a PD-Ll antagonist, such as an antagonistic PD-Ll antibody.
  • PD-Ll antibodies include, for example, MPDL3280A (RG7446; WO 2010/077634), durvalumab (MEDI4736), BMS-936559 (WO 2007/005874), and MSB0010718C (WO 2013/79174).
  • the immuno-oncology agent is a LAG-3 antagonist, such as an antagonistic LAG-3 antibody.
  • LAG3 antibodies include, for example, BMS-986016 (WO 10/19570, WO 14/08218), or IMP-731 or IMP-321 (WO 08/132601, WO 09/44273).
  • the immuno-oncology agent is a CD137 (4-1BB) agonist, such as an agonistic CD 137 antibody.
  • Suitable CD 137 antibodies include, for example, urelumab and PF-05082566 (WO 12/32433).
  • the immuno-oncology agent is a GITR agonist, such as an agonistic GITR antibody.
  • GITR antibodies include, for example, BMS-986153, BMS-986156, TRX-518 (WO 06/105021, WO 09/009116) and MK-4166 (WO 11/028683).
  • the immuno-oncology agent is an IDO antagonist.
  • IDO antagonists include, for example, INCB-024360 (WO 2006/122150, WO 07/75598, WO 08/36653, WO 08/36642), indoximod, or NLG-919 (WO 09/73620, WO 09/1156652, WO 11/56652, WO 12/142237).
  • the immuno-oncology agent is an OX40 agonist, such as an agonistic OX40 antibody.
  • OX40 antibodies include, for example, MEDI-6383 or MEDI-6469.
  • the immuno-oncology agent is an OX40L antagonist, such as an antagonistic OX40 antibody.
  • OX40L antagonists include, for example, RG-7888 (WO 06/029879).
  • the immuno-oncology agent is a CD40 agonist, such as an agonistic CD40 antibody.
  • the immuno-oncology agent is a CD40 antagonist, such as an antagonistic CD40 antibody.
  • Suitable CD40 antibodies include, for example, lucatumumab or dacetuzumab.
  • the immuno-oncology agent is a CD27 agonist, such as an agonistic CD27 antibody.
  • Suitable CD27 antibodies include, for example, varlilumab.
  • the immuno-oncology agent is MGA271 (to B7H3) (WO 11/109400).
  • kits useful for example, in the treatment or prevention of IDO-associated diseases or disorders, obesity, diabetes and other diseases referred to herein which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • the combination therapy is intended to embrace administration of these therapeutic agents in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner.
  • Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each therapeutic agent or in multiple, single dosage forms for each of the therapeutic agents.
  • Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues.
  • the therapeutic agents can be administered by the same route or by different routes.
  • a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally.
  • all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection.
  • Combination therapy also can embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients and non-drug therapies (e.g., surgery or radiation treatment).
  • the combination therapy further comprises a non-drug treatment
  • the non-drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and non-drug treatment is achieved. For example, in appropriate cases, the beneficial effect is still achieved when the non- drug treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks.
  • compositions which comprise a therapeutically effective amount of one or more of the compounds of formula I and/or formula II, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents, and optionally, one or more additional therapeutic agents described above.
  • the compounds of this invention can be administered for any of the uses described herein by any suitable means, for example, orally, such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups, and emulsions; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection, or infusion techniques (e.g.
  • nasally including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories.
  • nasally including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories.
  • nasally including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories.
  • nasal membranes such as by inhalation spray
  • topically such as in the form of a cream or ointment
  • rectally such as in the form of suppositories.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
  • manufacturing aid e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid
  • solvent encapsulating material involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • composition means a composition comprising a compound of the invention in combination with at least one additional pharmaceutically acceptable carrier.
  • a “pharmaceutically acceptable carrier” refers to media generally accepted in the art for the delivery of biologically active agents to animals, in particular, mammals, including, i.e. , adjuvant, excipient or vehicle, such as diluents, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
  • Pharmaceutically acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art. These include, without limitation: the type and nature of the active agent being formulated; the subject to which the agent-containing composition is to be administered; the intended route of administration of the composition; and the therapeutic indication being targeted. Pharmaceutically acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms. Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g. , stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art.
  • the dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.
  • the daily oral dosage of each active ingredient when used for the indicated effects, will range between about 0.001 to about 5000 mg per day, preferably between about 0.01 to about 1000 mg per day, and most preferably between about 0.1 to about 250 mg per day.
  • the most preferred doses will range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion.
  • Compounds of this invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
  • the compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, e.g. , oral tablets, capsules, elixirs, and syrups, and consistent with conventional pharmaceutical practices.
  • suitable pharmaceutical diluents, excipients, or carriers suitably selected with respect to the intended form of administration, e.g. , oral tablets, capsules, elixirs, and syrups, and consistent with conventional pharmaceutical practices.
  • Dosage forms suitable for administration may contain from about 1 milligram to about 2000 milligrams of active ingredient per dosage unit.
  • the active ingredient will ordinarily be present in an amount of about 0.1-95% by weight based on the total weight of the composition.
  • a typical capsule for oral administration contains at least one of the compounds of the present invention (250 mg), lactose (75 mg), and magnesium stearate (15 mg). The mixture is passed through a 60 mesh sieve and packed into a No. 1 gelatin capsule.
  • a typical injectable preparation is produced by aseptically placing at least one of the compounds of the present invention (250 mg) into a vial, aseptically freeze-drying and sealing. For use, the contents of the vial are mixed with 2 mL of physiological saline, to produce an injectable preparation.
  • the present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of the present invention, alone or in combination with a pharmaceutical carrier.
  • compounds of the present invention can be used alone, in combination with other compounds of the invention, or in combination with one or more other therapeutic agent(s), e.g. , an anticancer agent or other pharmaceutically active material.
  • the compounds of the present invention which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • the effective daily dose of the active compound may be any suitable daily dose of the active compound.
  • dosing is one administration per day.
  • a compound of the present invention While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation
  • references made in the singular may also include the plural.
  • references made in the singular may also include the plural.
  • “a” and “an” may refer to either one, or one or more.
  • any heteroatom with unsatisfied valences is assumed to have hydrogen atoms sufficient to satisfy the valences.
  • Optically active forms may be prepared by resolution of racemic forms or by synthesis from optically active starting materials. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention. When enantiomeric or diastereomeric products are prepared, they may be separated by conventional methods, for example, by chromatography or fractional crystallization. Depending on the process conditions the end products of the present invention are obtained either in free (neutral) or salt form. Both the free form and the salts of these end products are within the scope of the invention. If so desired, one form of a compound may be converted into another form.
  • a free base or acid may be converted into a salt; a salt may be converted into the free compound or another salt; a mixture of isomeric compounds of the present invention may be separated into the individual isomers.
  • Compounds of the present invention, free form and salts thereof, may exist in multiple tautomeric forms, in which hydrogen atoms are transposed to other parts of the molecules and the chemical bonds between the atoms of the molecules are consequently rearranged. It should be understood that all tautomeric forms, insofar as they may exist, are included within the invention. [00136]
  • the symbol 3 ⁇ 4 is used in formulas and tables to show the bond that is the point of attachment of the moiety or substituent to the core/nucleus of the structure.
  • alkyl and alkylene are intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • C1-C6 alkyl or “Ci-6 alkyl” denotes alkyl having 1 to 6 carbon atoms.
  • Example alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g. , n-propyl and isopropyl), butyl (e.g. , n-butyl, isobutyl, t- butyl), and pentyl (e.g.
  • Cl -C6alkylene denotes alkylene having 1 to 6 carbon atoms.
  • Example alkylene groups include, but are not limited to, methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), and the like.
  • aryl refers to an aromatic ring system which includes, but not limited to phenyl, biphenyl, indanyl, 1 -naphthyl, 2-naphthyl and terahydronaphthyl.
  • Halo or "halogen” includes fluoro, chloro, bromo, and iodo.
  • Haloalkyl is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogens.
  • haloalkyl include, but are not limited to, fluoromethyl,
  • haloalkyl also include "fluoroalkyl” that is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more fluorine atoms.
  • cycloalkyl refers to hydrocarbon rings having the indicated number of ring atoms, e.g., C3-C 6 cycloalkyl or C3-Ci 2 cycloalkyl, and being fully saturated. "Cycloalkyl” is also meant to refer to bicyclic and polycylic hydrocarbon rings such as, for example, bicyclo[2.2.1]heptane, bicyclo[2.2.2] octane, and adamantyl. cycloalkyl is intended to include C 3 , C 4 , C5, and e cycloalkyl groups.
  • Example cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and norbomyl.
  • substituted means that at least one hydrogen atom is replaced with a non-hydrogen group, provided that normal valencies are maintained and that the substitution results in a stable compound.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic groups such as amines; and alkali or organic salts of acidic groups such as carboxylic acids.
  • the pharmaceutically acceptable salts include the conventional nontoxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric; and the salts prepared from organic acids such as acetic, propionic, succinic, gly colic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, and isethionic, and the like.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric
  • organic acids such as acetic, propionic, succinic, gly colic, stearic,
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Allen, Jr., L.V., ed., Remington: The Science and Practice of Pharmacy, 22nd Edition,
  • compounds of formula I and formula II may have prodrug forms. Any compound that will be converted in vivo to provide the bioactive agent (i. e. , a compound of formula I or II) is a prodrug within the scope and spirit of the invention.
  • a prodrug within the scope and spirit of the invention.
  • Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see:
  • Bundgaard, H. Chapter 5: “Design and Application of Prodrugs", A Textbook of Drug Design and Development, pp. 113-191 , Krogsgaard-Larsen, P. et al, eds., Harwood Academic Publishers (1991); c) Bundgaard, H., Adv. Drug Deliv. Rev. , 8: 1-38 (1992);
  • compositions containing a carboxy group can form physiologically hydrolyzable esters that serve as prodrugs by being hydrolyzed in the body to yield formula I or formula II compounds per se.
  • prodrugs are preferably administered orally since hydrolysis in many instances occurs principally under the influence of the digestive enzymes. Parenteral administration may be used where the ester per se is active, or in those instances where hydrolysis occurs in the blood.
  • physiologically hydrolyzable esters of compounds of formula I or formula II include Ci_ 6 alkyl, C h alky lbenzyl, 4-methoxybenzyl, indanyl, phthalyl, methoxymethyl, Ci_6 alkanoyloxy-Ci_ 6 alkyl (e.g.
  • Ci_ 6 alkoxycarbonyloxy-Ci_ 6 alkyl e.g. , methoxycarbonyl-oxymethyl or ethoxycarbonyloxymethyl, glycyloxymethyl, phenylglycyloxymethyl, (5-methyl-2-oxo-l,3- dioxolen-4-yl)-methyl
  • esters may be prepared by conventional techniques known in the art.
  • prodrugs are well known in the art and described in, for example, King, F.D., ed., Medicinal Chemistry: Principles and Practice, The Royal Society of Chemistry, Cambridge, UK (Second Edition, reproduced, 2006); Testa, B. et al, Hydrolysis in Drug and Prodrug Metabolism. Chemistry, Biochemistry and Enzymology, VCHA and Wiley -VCH, Zurich, Switzerland (2003); Wermuth, C.G., ed., The Practice of Medicinal Chemistry, Third Edition, Academic Press, San Diego, CA (2008).
  • the present invention is intended to include all isotopes of atoms occurring in the present compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include deuterium and tritium.
  • Isotopes of carbon include 1 C and 14 C.
  • Isotopically - labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically -labeled reagent in place of the non-labeled reagent otherwise employed.
  • solvate means a physical association of a compound of this invention with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • the solvent molecules in the solvate may be present in a regular arrangement and/or a non-ordered arrangement.
  • the solvate may comprise either a
  • Solidvate encompasses both solution-phase and isolable solvates.
  • Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are generally known in the art.
  • the term "patient” refers to organisms to be treated by the methods of the present invention.
  • Such organisms preferably include, but are not limited to, mammals (e.g. , murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably refers to humans.
  • the term "effective amount” means that amount of a drug or pharmaceutical agent, i.e., a compound of the invention, that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician.
  • therapeutically effective amount means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder.
  • An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route. The term also includes within its scope amounts effective to enhance normal physiological function
  • treating includes any effect, e.g. , lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
  • salts of the compounds of the present invention are contemplated as being pharmaceutically acceptable.
  • salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
  • the compounds of the present invention may be prepared by methods such as those illustrated in the following Schemes utilizing chemical transformations known to those skilled in the art. Solvents, temperatures, pressures, and other reaction conditions may readily be selected by one of ordinary skill in the art. Starting materials are commercially available or readily prepared by one of ordinary skill in the art. These Schemes are illustrative and are not meant to limit the possible techniques one skilled in the art may use to manufacture compounds disclosed herein. Different methods may be evident to those skilled in the art. Additionally, the various steps in the synthesis may be performed in an alternate sequence or order to give the desired compound(s).
  • PCT/US2015/059271 PCT/US2015/059311, and PCT/US2015/059316.
  • Schemes 1-3 depict methods for preparing compounds of formula I. These methods can also be applied to preparation of compounds of the invention.
  • the group R 4 can be appended by alkylation of intermediate VIII.
  • Methods for control of the absolute stereochemistry of the resulting asymmetric center are known to those familiar with the art, as are chiral separation methods.
  • Saponification of the ester by heating with aq. LiOH or a similar base, generally in the presence of an organic co-solvent such as THF affords carboxylic acids IX.
  • Treatment of acids IX with amines X under standard peptide coupling conditions will afford compounds of the invention I.
  • peptide coupling methods see: Ayman El-Faham and Fernando Albericio. ChemRev. 2011, 111, 6557-6602.
  • Scheme 3 illustrates a method for controlling the absolute stereochemistry of
  • esters XIV provides carboxylic acids XV.
  • Treatment of these acids with an acid chloride such as pivaloyl chloride provides a mixed anhydride intermediate.
  • an optically pure oxazolidinone of known stereochemistry and general structure XVI is deprotonated by treatment with a strong base such as n-BuLi.
  • bases such as NaHMDS. Alkylation of the resulting enolate proceeds with predictable control of stereochemistry at the newly-formed center to provide materials XVIII.
  • DMEM Dulbecco's Modification of Eagle's Medium
  • EDTA ethylenediaminetetraacetic acid.
  • Method A Column: Waters Acquity UPLC BEH C 18, 2.1 x 50 mm, 1.7- ⁇ particles; Mobile Phase A: 5:95 acetonitrile:water with 0.1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile:water with 0.1% trifluoroacetic acid; Temperature: 50°C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 220 nm.
  • Method B Waters Acquity SDS using the following method: Linear Gradient of 2% to98% solvent B over 1.7 min; UV visualization at 220 nm; Column: BEH C18 2.1 mm x 50 mm; 1.7 um particle (Heated to Temp. 50 °C); Flow rate: 0.8 ml/min; Mobile phase A: 100% Water, 0.05% TFA; Mobile phase B: 100% Acetonitrile, 0.05% TFA.
  • Method C Column: Waters Acquity UPLC BEH C18, 2.1 x 50 mm, 1.7- ⁇ particles; Mobile Phase A: 5:95 acetonitrile: water with 10 mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate; Temperature: 50°C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 220 nm.
  • Preparation D (100 g, 0.348 mol) was treated with 4 N HC1 ( 300 mL) in acetone (1200 mL) at 45 ° C overnight. The mixture was monitored by TLC. Then the solution was then concentrated in vacuo. The residue was adjusted to pH 9 with 6 N NaOH and the mixture was partitioned between ethyl acetate and water.
  • Preparation E (57.8 g, 237.8 mmol) was dissolved in EtOH (240 mL) and cooled to 0 °C.
  • NaBH4 (9.94 g, 261.6 mmol) was added portionwise maintaining the temperature within a range of 0-10 °C (exothermic reaction). The resulting suspension was stirred for 20 minutes.
  • the reaction was quenched at 0 °C by the slow addition of acetone (58 mL) over 15 minutes (exotherm).
  • the reaction was poured slowly onto 500 mL of saturated aqueous ammonium chloride and 500 g of ice.
  • heterogeneous mixture was quenched by the addition of saturated aqueous ammonium chloride (200 mL) and extracted with CH2CI2 (3 x 300 mL). The combined organic fractions were dried over anhydrous sodium sulfate and concentrated under reduced pressure. Excess pyridine was removed by azeotroping from toluene (3 x 300 mL). The crude material was recrystallized from LLO/MeOH as follows: 1 mL/mmol of H2O was added and the slurry was heated to 120 °C in an oil bath. MeOH was added until the solids went into solution (-0.5 L).
  • reaction mixture was rapidly diluted with saturated aqueous ammonium chloride solution (400 mL) and the biphasic mixture was stirred for 15 min.
  • 'PrOAc 100 mL was added, the layers were separated, and the aqueous layer was extracted with 'PrOAc (3 x 50 mL).
  • the combined organic extracts were dried over anhydrous magnesium sulfate filtered, and concentrated.
  • the resulting residue was recrystallized by dissolving in 400 mL hot acetone and adding H 2 0 until a milky solution formed followed to re-dissolving with heating (-3: 1 acetone/H 2 0).
  • Preparation J was obtained as white needles (15.04 g, 2 crops, 68 %).
  • Example 1 N-(4-Chloro-2-methylbenzyl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide [00178] To a mixture of the HC1 salt of 2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin- 4-yl)acetic acid (1C, 25.6 mg, 0.09 mmol) and 4-chloro-2-methylbenzylamine (15.8 mg, 0.1 mmol) in anhydrous DMF (1 mL), under nitrogen atmosphere, was added DIPEA (0.05 mL, 0.3 mmol) followed by PyBOP (44.1 mg, 0.09 mmol).
  • Examples 2-5 were prepared using 2-(l-(6-fluoroquinolin-4-yl)-4- methylpiperidin-4-yl)acetic acid (1C) and corresponding amines following the procedure described for the synthesis of Example 1.
  • Examples 7-9 were prepared using 2-(4-methyl-l-(2-(trifluoromethyl)pyridin- 4-yl)piperidin-4-yl)acetic acid and corresponding amines following the procedure described for the synthesis of Example 6.
  • Examples 11-18 were prepared from Preparation K utilizing the procedure used to make Example 10 and corresponding benzylamines.
  • Method A Waters Acquity SDS using the following method: Linear Gradient of 2% to 98% solvent B over 1.7 min; UV visualization at 220 nm; Column: BEH CI 8 2.1 mm x 50 mm; 1.7 um particle (Heated to Temp. 50 °C); Flow rate: 0.8 ml/min; Mobile phase A: 100% Water, 0.05% TFA; Mobile phase B: 100% Acetonitrile, 0.05% TFA.
  • Method B Column: Waters Acquity UPLC BEH CI 8, 2.1 x 50 mm, 1.7- ⁇ particles; Mobile Phase A: 5 :95 acetonitrile:water with 0.1 % trifluoroacetic acid; Mobile Phase B: 95 :5 acetonitrile:water with 0.1% trifluoroacetic acid; Temperature: 50 °C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.00 mL/min; Detection: UV at 220 nm.
  • Method C Column: Waters Acquity UPLC BEH C18, 2.1 x 50 mm, 1.7- ⁇ particles; Mobile Phase A: 5 :95 acetonitrile:water with 0.1 % trifluoroacetic acid; Mobile Phase B: 95 :5 acetonitrile:water with 0.1% trifluoroacetic acid; Temperature: 50 °C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 220 nm.
  • Example 20-2 Concentration of the later eluting fractions afforded Example 20-2 (5.8 mg) assigned as N-(adamantan-2-yl)-2-(l-((6-(trifluoromethyl)pyridin-2-yl)methyl)piperidin-4- yl)butanamide (Enantiomer 2).
  • MS (ES): m/z 464 [M+H] + .
  • T r 1.74 min (Method B).
  • KYN kynurenine
  • centrifugation for 5 min at 800 x g and sonicated in 150 PBS containing a protease inhibitor cocktail (Set 2; Calbiochem, EMD Biosciences, San Diego, CA).
  • the sonicate can be centrifuged for 10 min at 10,000 x g, and the supernatant can be assayed in triplicate by incubating a 40 ⁇ sample with an equal volume of 100 mmol/L potassium phosphate buffer, pH 6.5, containing 40 mmol/L ascorbic acid (neutralized to pH 7.0), 100 ⁇ /L methylene blue, 200 ⁇ g/mL catalase, and 400 ⁇ /1 L-Trp for 30 min at 37 °C.
  • the assay can be terminated by the addition of 16 ⁇ . 30% (w/v) trichloroacetic acid (TCA) and further incubated at 60 °C for 15 min to hydrolyze N-formylkynurenine to ⁇ .
  • TCA trichloroacetic acid
  • the mixture can then be centrifuged at 12,000 rpm for 15 min, and ⁇ can be quantified by mixing equal volume of supernatant with 2% (w/v) Ehrlich's reagent in glacial acetic acid in 96-well microtiter plate and reading the absorbance at 480 nm using L-KYN as standard.
  • Protein in the islet samples can be quantified by Bio-Rad Protein assay at 595 nm. For the detection of L-KYN in the islet culture
  • proteins can be precipitated with 5% (w/v) TCA and centrifuged at 12,000 rpm for 15 min, and determination of KYN in the supernatant with Ehrlich's reagent can be determined as described above.
  • IL-4 (10 ⁇ g/mL; 500-2,000 units/mL) and 1-a-methyl Trp (1-MT; 40 ⁇ /L) can be added to the incubation media as indicated.
  • This assay can also form the basis of a cell-based assay, and may be quantified via LCMS/MS as an alternative to UV/Vis detection.
  • Immunoreactive proteins can be visualized with ECL PLUS® Westem blotting detection reagent (Amersham Biosciences, Buckinghamshire, U.K.) after incubation for 1 h with anti-mouse horseradish peroxidase- conjugated secondary antibody (Jackson Immunolabs, West Grove, PA).
  • the tissues can then be reacted with mouse monoclonal anti-human IDO antibody (1 :20; Chemicon) and goat polyclonal anti-human PDX1 antibody (1 :2,000; which may be requested from Dr. Chris Wright, School of Medicine, Vanderbilt, TN) overnight at room temperature in a humid chamber.
  • Secondary antibodies anti-goat (labeled with Cy3) and anti-mouse (labeled with Cy2) can be purchased from Jackson Immunolabs and can be used at a concentration of 1 :200.
  • the nuclei can be stained with Hoechst 33258 (Molecular Probes, Eugene, OR). Images can be acquired by Intelligent Imaging System software from an Olympus 1X81 inverted motorized microscope equipped with Olympus DSU (spinning disk confocal) and Hamamatsu ORCA HER monochromatic CCD camera.
  • Biochemical Assay cDNA clones for both human and mouse IDO have been isolated and verified by sequencing and are commercially available.
  • C-terminal His-tagged IDO protein can be produced in E. coli using the IPTG-inducible pET5a vector system and isolated over a nickel column. The yield of the partially purified protein can be verified by gel electrophoresis and the concentration estimated by comparison to protein standards.
  • a 96-well plate spectrophotometric assay for kynurenine production can be run following published procedures (see, e.g., Littlejohn, T.K. et al., Prot. Exp.
  • compounds can be evaluated at a single concentration of, for example, 200 ⁇ against 50 ng of IDO enzyme in 100 reaction volumes with tryptophan added at increasing concentrations at, for example, 0, 2, 20, and 200 ⁇ . Kynurenine production can be measured at 1 hour.
  • COS-1 cells can be transiently transfected with a CMV promoter-driven plasmid expressing IDO cDNA using Lipofectamine 2000 (Invitrogen) as recommended by the manufacturer.
  • a companion set of cells can be transiently transfected with TDO-expressing plasmid.
  • the cells can be apportioned into a 96-well format at 6 ⁇ 10 4 cells per well. The following day, the wells can be washed and new media (phenol red free) containing 20 ⁇ g/mL tryptophan can be added together with inhibitor.
  • the reaction can be stopped at 5 hours and the supernatant removed and spectrophotometrically- assayed for kynurenine as previously described for the enzyme assay.
  • compounds can be evaluated at a single concentration of, for example, 100 ⁇ . More extensive dose-escalation profiles can be collected for select compounds.
  • a pharmacodynamic assay can be based on measuring serum levels of both kynurenine and tryptophan, and calculating the kynurenine/tryptophan ratio provides an estimate of IDO activity that is independent of baseline tryptophan levels.
  • Serum tryptophan and kynurenine levels can be determined by HPLC analysis, and serum compound levels can optionally also be determined in the same HPLC run.
  • Compounds can be initially evaluated by challenging mice with LPS and then subsequently administering a bolus dose of compound at the time that the serum kynurenine level plateaus. As the kynurenine pool is rapidly turned over with a half-life in serum of less than 10 minutes, pre-existing kynurenine is not expected to unduly mask the impact that an IDO inhibitor has on kynurenine production.
  • Each experiment can include non-LPS-exposed mice (to determine baseline kynurenine levels against which to compare the other mice) and a set of LPS- exposed mice dosed with vehicle alone (to provide a positive control for IDO activation). Each compound can initially be evaluated in mice at a single high i.p.
  • bolus dose in the range of at least 100 mg/kg.
  • Blood can be collected at defined time intervals (for example, 50 sample at 5, 15, 30 min., 1, 2, 4, 6, 8, and 24 hr. following compound administration) for HPLC analysis of kynurenine and tryptophan levels (pharmacodynamic analysis) as well as for the level of compound (pharmacokinetic analysis). From the pharmacokinetic data the peak serum concentration of compound achieved can be determined as well as the estimated rate of clearance. By comparing the level of compound in serum relative to the kynurenine/tryptophan ratio at various time points, the effective IC5 0 for IDO inhibition in vivo can be roughly estimated. Compounds exhibiting efficacy can be evaluated to determine a maximum dose that achieves 100% IDO inhibition at the peak concentration.
  • HEK293 cells were transfected with a pCDNA-based mammalian expression vector harboring human IDOl cDNA (NM 002164.2) by electroporation. They were cultured in medium (DMEM with 10% FBS) containing 1 mg/ml G418 for two weeks. Clones of HEK293 cells that stably expressed human IDOl protein were selected and expanded for IDO inhibition assay.
  • medium DMEM with 10% FBS
  • the human IDO 1/HEK293 cells were seeded at 10,000 cells per 50 ⁇ per well with RPMI/phenol red free media contains 10% FBS in a 384-well black wall clear bottom tissue culture plate (Matrix Technologies LLC) 100 nL of certain concentration of compound was then added to each well using ECHO liquid handling systems. The cells were incubated for 20 hours in 37 °C incubator with 5% C0 2 . [00224] The compound treatments were stopped by adding trichloroacetic acid (Sigma- Aldrich) to a final concentration at 0.2%. The cell plate was further incubated at 50 °C for 30 minute.
  • Compound IC5 0 values were calculated using the counts of 500 nM of a reference standard treatment as one hundred percent inhibition, and counts of no compound but DMSO treatment as zero percent inhibition.
  • HeLa (ATCC® CCL-2) cells were obtained from the ATCC® and cultured in Dulbecco's Modified Eagle Medium supplemented with 4.5 g/L glucose, 4.5 g/L L-glutamine and 4.5 g/L sodium pyruvate (#10-013-CV, Corning), 2 mM L-alanyl-L-glutamine dipeptide (#35050-061, Gibco), lOOU/mL penicillin, 100 ⁇ g/mL streptomycin (#SV30010, HyClone) and 10% fetal bovine serum (#SH30071.03 HyClone). Cells were maintained in a humidified incubator at 37 °C in 5% C0 2 .
  • IDO activity was assessed as a function of kynurenine production as follows: HeLa cells were seeded in a 96-well culture plate at a density of 5,000 cells/well and allowed to equilibrate overnight. After 24 hours, the media was aspirated and replaced with media containing IFNy (#285-IF/CF, R&D Systems) at a final concentration of 25 ng/mL. A serial dilution of each test compound was added to the cells in a total volume of 200 of culture medium. After a further 48 hour incubation, 170 ⁇ . of supernatant was transferred from each well to a fresh 96-well plate. 12.1 ⁇ . of 6.
  • X is CH or N
  • T CH or N
  • R 1 is H, halo, or Ci-Cehaloalkyl
  • R 1A is H, halo, or Ci-C 6 haloalkyl
  • Y is CH or N
  • n 0, 1 , 2, 3, or 4;
  • W is N or CR 4 wherein R 4 is H or (Ci-C 6 alkyl);
  • V is Ci-Cealkylene optionally substituted with one, two, or three substituents
  • Ci-Cealkyl and C 3 -Cecycloalkyl independently selected from Ci-Cealkyl and C 3 -Cecycloalkyl
  • R 2 is H or Ci-C 6 alkyl
  • Z is Ci-Cealkylene optionally substituted with one, two, or three substituents
  • Ci-C 6 alkyl and C 3 -C 6 cycloalkyl independently selected from Ci-C 6 alkyl and C 3 -C 6 cycloalkyl
  • B is aryl optionally substituted with 1 , 2, or 3 R substituents independently selected from -OH, Ci-C 6 alkyl, Ci-C 6 haloalkyl, halo, -OCi-C 6 alkyl, -OCi-C 6 haloalkyl, -
  • C 3 -Ci2cycloalkyl optionally substituted with 1 , 2, or 3 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi- Cealkyl, -OCi-C 6 haloalkyl, -CN, aryl, or -Oaryl;
  • Aspect 2 The compound of Aspect 1 that is a compound of formula I.
  • Aspect 3 The compound of cl Aspect aim 1 that is a compound of formula II.
  • Aspect 4 The compound of any one of the preceding Aspects, wherein R 1 is H, F, or -CF 3 .
  • Aspect 5. The compound of any one of the preceding Aspects, wherein R is H.
  • Aspect 6 The compound of any one of the preceding Aspects, wherein Y is CH.
  • Aspect 7 The compound of any one of Aspects 1 to 3, wherein Y is N.
  • Aspect 8 The compound of any one of the preceding Aspects, wherein n is 2.
  • Aspect 9 The compound of any one of the preceding Aspects, wherein W is CR 4 .
  • Aspect 10 The compound of any one of Aspects 1 to 8, wherein W is N.
  • Aspect 11 The compound of any one of the preceding Aspects, wherein V is Cialkylene optionally substituted with one substituent that is Ci-Cealkyl and C3-C 6 cycloalkyl.
  • Aspect 12 The compound of any one of the preceding Aspects, wherein Z is Cialkylene optionally substituted with one or two substituents independently selected from Ci- Cealkyl and C 3 -C 6 cycloalkyl.
  • Aspect 13 The compound of any one of the preceding claims, wherein B is aryl optionally substituted with 1, 2, or 3 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi-C 6 alkyl, -OCi-C 6 haloalkyl, -CN, aryl, or -Oaryl.
  • Aspect 14 The compound of Aspects 13, wherein B is phenyl optionally substituted with 1 or 2 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, - OCi-Cealkyl, -OCi-C 6 haloalkyl, -CN, aryl, or -Oaryl.
  • Aspect 15 The compound of any one of Aspects 1 to 12, wherein B is C 3 -Ci2cycloalkyl optionally substituted with 1 , 2, or 3 R substituents independently selected from -OH, Ci- Cealkyl, Ci-C 6 haloalkyl, halo, -OCi-C 6 alkyl, -OCi-C 6 haloalkyl, -CN, aryl, or -Oaryl.
  • Aspect 16 The compound of Aspect 1 that is
  • a pharmaceutical composition comprising a compound of any one of Aspects 1 to 16 and a pharmaceutically acceptable carrier.
  • Aspect 18 The pharmaceutical composition of Aspect 17, further comprising YERVOY, OPDIVO, or KEYTRUDA, or a combination thereof.
  • Aspect 19 A method of treating cancer in a patient in need of such treatment comprising administering to the patient a therapeutically effective amount of a compound according to any one of Aspects 1 to 16.

Abstract

There are disclosed compounds that modulate or inhibit the enzymatic activity of indoleamine 2,3-dioxygenase (IDO), pharmaceutical compositions containing said compounds and methods of treating proliferative disorders, such as cancer, viral infections and/or inflammatory disorders utilizing the compounds of the invention.

Description

INHIBITORS OF INDOLEAMINE 2.3-DIOXYGENASE AND METHODS OF THEIR USE
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application No.
62/331,871, filed May 4, 2016, the entirety of which is incorporated by reference herein.
FIELD OF THE INVENTION
[0002] The invention relates generally to compounds that modulate or inhibit the enzymatic activity of indoleamine 2,3-dioxygenase (IDO), pharmaceutical compositions containing said compounds and methods of treating proliferative disorders, such as cancer, viral infections and/or autoimmune diseases utilizing the compounds of the invention.
BACKGROUND OF THE INVENTION
[0003] Indoleamine 2,3-dioxygenase (IDO; also known as IDOl) is an IFN-γ target gene that plays a role in immunomodulation. IDO is an oxidoreductase and one of two enzymes that catalyze the first and rate-limiting step in the conversion of tryptophan to N-formyl- kynurenine. It exists as a 41kD monomer that is found in several cell populations, including immune cells, endothelial cells, and fibroblasts. IDO is relatively well-conserved between species, with mouse and human sharing 63% sequence identity at the amino acid level. Data derived from its crystal structure and site-directed mutagenesis show that both substrate binding and the relationship between the substrate and iron-bound dioxygenase are necessary for activity. A homolog to IDO (ID02) has been identified that shares 44% amino acid sequence homology with IDO, but its function is largely distinct from that of IDO. (See, e.g. , Serafini, P. et al, Semin. Cancer Biol. , 16(l):53-65 (Feb. 2006) and Ball, H.J. et al, Gene, 396(1):203-213 (Jul. 1, 2007)).
[0004] IDO plays a major role in immune regulation, and its immunosuppressive function manifests in several manners. Importantly, IDO regulates immunity at the T cell level, and a nexus exists between IDO and cytokine production. In addition, tumors frequently manipulate immune function by upregulation of IDO. Thus, modulation of IDO can have a therapeutic impact on a number of diseases, disorders and conditions.
[0005] A pathophysiological link exists between IDO and cancer. Disruption of immune homeostasis is intimately involved with tumor growth and progression, and the production of IDO in the tumor microenvironment appears to aid in tumor growth and metastasis. Moreover, increased levels of IDO activity are associated with a variety of different tumors (Brandacher, G. et al, Clin. Cancer Res. , 12(4): 1144-1151 (Feb. 15, 2006)).
[0006] Treatment of cancer commonly entails surgical resection followed by chemotherapy and radiotherapy. The standard treatment regimens show highly variable degrees of long-term success because of the ability of tumor cells to essentially escape by regenerating primary tumor growth and, often more importantly, seeding distant metastasis. Recent advances in the treatment of cancer and cancer-related diseases, disorders and conditions comprise the use of combination therapy incorporating immunotherapy with more traditional chemotherapy and radiotherapy. Under most scenarios, immunotherapy is associated with less toxicity than traditional chemotherapy because it utilizes the patient's own immune system to identify and eliminate tumor cells.
[0007] In addition to cancer, IDO has been implicated in, among other conditions, immunosuppression, chronic infections, and autoimmune diseases or disorders (e.g., rheumatoid arthritis). Thus, suppression of tryptophan degradation by inhibition of IDO activity has tremendous therapeutic value. Moreover, inhibitors of IDO can be used to enhance T cell activation when the T cells are suppressed by pregnancy, malignancy, or a virus (e.g., HIV). Although their roles are not as well defined, IDO inhibitors may also find use in the treatment of patients with neurological or neuropsychiatric diseases or disorders (e.g., depression).
[0008] Small molecule inhibitors of IDO have been developed to treat or prevent IDO- related diseases. For example, the IDO inhibitors 1-methyl-DL-tryptophan; p-(3-benzofuranyl)- DL-alanine; p-[3-benzo(b)thienyl]-DL-alanine; and 6-nitro-L-tryptophan have been used to modulate T cell-mediated immunity by altering local extracellular concentrations of tryptophan and tryptophan metabolites (WO 99/29310). Compounds having IDO inhibitory activity are further reported in PCT Publication No. WO 2004/094409.
[0009] In view of the role played by indoleamine 2,3-dioxygenase in a diverse array of diseases, disorders and conditions, and the limitations (e.g., efficacy) of current IDO inhibitors, new IDO modulators, and compositions and methods associated therewith, are needed.
SUMMARY OF THE INVENTION
[0010] The invention is directed to compounds of formula I and formula II:
Figure imgf000004_0001
wherein X is CH or N; T is CH or N; R1 is H, halo, or Ci-C6haloalkyl; R1A is H, halo, or C C6haloalkyl; Y is CH or N; n is 0, 1, 2, 3, or 4; W is N or CR4 wherein R4 is H or C(Ci-C6alkyl); V is Ci-C6alkylene optionally substituted with one, two, or three substituents independently selected from Ci-Cealkyl and C3-C6cycloalkyl; R2 is H or Ci-Cealkyl; Z is a bond or Ci- Cealkylene optionally substituted with one, two, or three substituents independently selected from Ci-C6alkyl and C3-C6cycloalkyl; and B is aryl optionally substituted with 1, 2, or 3 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi-Cealkyl, - OCi-Cehaloalkyl, -CN, aryl, or -Oaryl; or B is C3-Ci2cycloalkyl optionally substituted with 1, 2, or 3 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi-C6alkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl.
[0011] Also within the scope of the invention are pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of the compounds of formula I and formula II.
[0012] The invention is also directed to pharmaceutical compositions comprising one or more compounds of formula I and/ or formula II. The invention is also directed to methods of treating cancer using one or more compounds of formula I and/ or formula II.
DETAILED DESCRIPTION OF THE INVENTION COMPOUNDS OF THE INVENTION
[0013] The invention is directed to compounds of formula I and formula II:
Figure imgf000004_0002
(II) [0014] According to the disclosure, X is CH or N. In some aspect, X is CH. In other aspects, X is N.
[0015] According to the disclosure T is CH or N. In some aspects, T is CH. In other aspects, T is N.
[0016] In some embodiments of formula I, X is CH and T is CH. In other
embodiments, X is N and T is CH. In other embodiments, X is CH and T is N. In other embodiments, X is N and T is N.
[0017] According to the disclosure, R1 is H, halo, or Ci-Cehaloalkyl. In other aspects, R1 is H, halo, Ci-C6alkyl, or Ci-Cehaloalkyl. In some aspects, R1 is H. In some aspects, R1 is halo (F, CI, Br, or I), preferably F. In other aspects, R1 is Ci-Cealkyl, for example, methyl, ethyl, isopropyl, butyl, and t-butyl. In yet other aspects, R1 is Ci-Cehaloalkyl, for example, -CF3. In some aspects, R1 is halo, Ci-C6alkyl, or Ci-Cehaloalkyl. In other aspects, R1 is halo or Ci- Cehaloalkyl.
[0018] In those aspects of the disclosure comprising compounds of formula I, when R1 is halo, Ci-C6alkyl, or Ci-Cehaloalkyl, the R1 is preferably present at the 4-carbon position of the ring. In other aspects of the disclosure comprising compounds of formula I, when R1 is halo, Ci- Cealkyl, or Ci-Cehaloalkyl, the R1 can be present at the 2-carbon position of the ring. Preferably, in those aspects of the disclosure comprising compounds of formula I, when R1 is F or CF3 at the 4-carbon position of the ring.
[0019] In those aspects of the disclosure comprising compounds of formula II, when R1 is halo, Ci-C6alkyl, or Ci-Cehaloalkyl, the R1 is preferably present at the 2-carbon position of the ring. In other aspects of the disclosure comprising compounds of formula II, when R1 is halo, Ci-C6alkyl, or Ci-Cehaloalkyl, the R1 can be present at the 3-carbon position of the ring.
Preferably, in those aspects of the disclosure comprising compounds of formula II, when R1 is F or CF3 at the 2-carbon position of the ring.
[0020] According to the disclosure, R1A is H, halo, Ci-Cealkyl, or Ci-Cehaloalkyl. In preferred aspects, R1A is H. In some aspects, R1A is halo (F, CI, Br, or I). In other preferred aspects, R1A is Ci-Cealkyl, for example, methyl, ethyl, isopropyl, butyl, and t-butyl. In yet other aspects, R1A is Ci-Cehaloalkyl, for example, -CF3.
[0021] According to the disclosure, Y is CH or N. In some aspects, Y is CH. In other aspect, Y is N. In other embodiments, Y is -C(Ci-C6alkyl), for example, -C(CH3) or
-C(CH2CH3). [0022] According to the disclosure, n is 0, 1, 2, 3, or 4. In preferred aspects, n is 2. In other aspects, n is 0. In other aspects, n is 1. In yet other aspects, n is 3. In still other aspects, n is 4. In some aspects, n is 0 to 2 or 1 to 2. In yet other aspects, n is 1 to 3.
[0023] According to the disclosure, W is N or CR4, wherein R4 is H or -(Ci-C6alkyl)-. In some aspects, W is N. In other aspects, W is CR4 and R4 is H, e.g, W is -CH-. In some aspects, W is CR4 and R4 is -(Ci-Cealkyl), methyl, methyl, propyl, isopropyl, butyl, s-butyl, t- but l, and the like (i.e., W is -C(d-C6alkyl)-).
[0024] In some aspects, Y is CH and W is CH. In other aspects, Y is CH and W is N. In other aspects, Y is N and W is CH. In still other aspects, Y is N and W is N. In some aspects, Y is CH and W is C(Ci-C6alkyl). In other aspects, Y is N and W is C(Ci-C6alkyl).
[0025] According to the disclosure, V is unsubstituted Ci-Cealkylene, for example, unsubstituted Ci-Csalkylene, Ci-C4alkylene, Ci-C3alkylene, Ci-C2alkylene, or Cialkylene. In other aspects of the disclosure, V is Ci-Cealkylene, for example, Ci-Csalkylene, Ci-C4alkylene, Ci-C3alkylene, Ci-C2alkylene, or Cialkylene, substituted with one, two, or three substituents, preferably 1 or 2 substituents, independently selected from Ci-Cealkyl (e.g., methyl, ethyl, isopropyl) and C3-C6cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl). In a preferred aspect, V is a Cialkylene substituted with one or two, preferably one, Ci-Cealkyl (e.g., methyl or ethyl) substituents.
[0026] According to the disclosure, R2 is H or Ci-Cealkyl. In some aspects, R2 is H. In other aspects, R2 is Ci-Cealkyl, for example, methyl, ethyl, isopropyl, butyl, or t-butyl.
[0027] According to the disclosure, Z is unsubstituted Ci-Cealkylene, for example, unsubstituted Ci-Csalkylene, Ci-C4alkylene, Ci-C3alkylene, Ci-C2alkylene, or Cialkylene. In other aspects of the disclosure, Z is Ci-Cealkylene, for example, Ci-Csalkylene, Ci-C4alkylene, Ci-C3alkylene, Ci-C2alkylene, or Cialkylene, substituted with one, two, or three substituents, preferably 1 or 2 substituents, independently selected from Ci-Cealkyl (e.g., methyl, ethyl, isopropyl) and C3-C6cycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl). In a preferred aspect, Z is a Cialkylene substituted with one or two Ci-Cealkyl (e.g., methyl or ethyl) substituents. In another preferred aspects, Z is preferably Cialkylene substituted with one C3- Cecycloalkyl (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl).
[0028] In other aspects, Z is a bond, e.g., Z is absent.
[0029] According to some aspects of the disclosure, B is unsubstituted aryl, for example, unsubstituted phenyl or naphthyl. In other aspects, B is aryl substituted with 1, 2, or 3 substituents, preferably 1 or 2 substituents. The B substituents, which can be referred to as one or more R groups, are preferably independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi-C6alkyl, -OCi-Cehaloalkyl, -CN, aryl, or -Oaryl. Preferred substituted aryl moieties include phenyl and naphthyl, with phenyl being particularly preferred. In some aspects, the aryl is substituted with at least one substituent that is -OH. In some aspects, the aryl is substituted with at least one substituent that is Ci-Cealkyl, for example, methyl, ethyl, propyl, isopropyl, butyl, s-butyl, t-butyl, pentyl, or hexyl. In other aspects, the aryl is substituted with at least one substituent that is Ci-Cehaloalkyl, for example, -CF3. In other embodiments, the aryl is substituted with at least one substituent that is halo, for example, F, CI, or Br, with F and CI substituents being particularly preferred aryl substituents. In some aspects, the aryl is substituted with at least one substituent that is -OCi-Cealkyl, for example, methoxy, ethoxy, propoxy, isoproxy, butoxy, or t-butoxy. In yet other aspects, the aryl is substituted with at least one substituent that is -OCi-Cehaloalkyl, for example, -OCF3. In some embodiments, the aryl is substituted with at least one substituent that is -CN. In other aspects, the aryl is substituted with at least one substituent that is another aryl moiety, for example, a phenyl or naphthyl moiety. In some embodiments, the aryl is substituted with at least one substituent that is -Oaryl, for example, -Ophenyl.
[0030] According to some aspects of the disclosure, B is unsubstituted C3- C cycloalkyl, for example, C3cycloalkyl (e.g., cyclopropyl), C4cycloalkyl (e.g., cyclobutyl), CiCycloalkyl (e.g., cyclopentyl), Cecycloalkyl (e.g., cyclohexyl), Cycycloalkyl (e.g., bicyclo[2.2.1]heptanyl), Cgcycloalkyl (bicyclo[2.2.2]octanyl), Cgcycloalkyl, Ciocycloalkyl (e.g., adamantyl), Cncycloalkyl, or Cncycloalkyl. In some aspects, B is unsubstituted C3- Cecycloalkyl. In other aspects, B is unsubstituted C7-Ci2cycloalkyl or C7-Ciocycloalkyl.
[0031] In other aspects, B is C3-Ci2cycloalkyl substituted with 1 , 2, or 3 substituents, preferably 1 or 2 substituents. For example, in some aspects, B is substituted C3cycloalkyl, C4cycloalkyl, Cscycloalkyl, Cecycloalkyl, Cycycloalkyl, Cgcycloalkyl, Cgcycloalkyl,
Ciocycloalkyl, Cncycloalkyl, or Cncycloalkyl. In other aspects, B is substituted C3- Cecycloalkyl. In other aspects, B is substituted C7-Ci2cycloalkyl or C7-Ciocycloalkyl. The B substituents, which can be referred to as one or more R groups, are preferably independently selected from -OH, Ci-C6alkyl, Ci-C6haloalkyl, halo, -OCi-C6alkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl. Preferred C3-Ci2cycloalkyl moieties include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo[2.2.1]heptanyl, bicyclo[2.2.2]octanyl, and adamantyl. In some aspects, the C3-Ci2cycloalkyl is substituted with at least one substituent that is -OH. In some aspects, the C3-Ci2cycloalkyl is substituted with at least one substituent that is Ci-Cealkyl, for example, methyl, ethyl, propyl, isopropyl, butyl, s-butyl, t-butyl, pentyl, or hexyl. In other aspects, the C3-Ci2cycloalkyl is substituted with at least one substituent that is Ci-Cehaloalkyl, for example, -CF3. In other embodiments, the C3-Ci2cycloalkyl is substituted with at least one substituent that is halo, for example, F, CI, or Br, with F and CI substituents being particularly preferred C3-Ci2cycloalkyl substituents. In some aspects, the C3-Ci2cycloalkyl is substituted with at least one substituent that is -OCi-Cealkyl, for example, methoxy, ethoxy, propoxy, isoproxy, butoxy, or t-butoxy. In yet other aspects, the C3-Ci2cycloalkyl is substituted with at least one substituent that is -OCi-Cehaloalkyl, for example, -OCF3. In some embodiments, the C3-Ci2cycloalkyl is substituted with at least one substituent that is -CN. In other aspects, the C3- Ci2cycloalkyl is substituted with at least one substituent that is an aryl moiety, for example, a phenyl or naphthyl moiety. In some embodiments, the C3-Ci2cycloalkyl is substituted with at least one substituent that is -Oaryl, for example, -Ophenyl.
[0032] The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I and II.
[0033] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is CR4, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted aryl, for exam le,
Figure imgf000008_0001
wherein X is CH, R is H or Ci-C6alkyl, Rb is H, Ci-C6alkyl, or C3-C6cycloalkyl, and RbA is absent, H, or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I-A and II-A are those wherein X is N, R5 is H or Ci-Cealkyl, R6 is H, Ci-Cealkyl, or C3-C6cycloalkyl, and R^ is absent, H, or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-A and II-A. [0034] Sub-formulas of formula I or formula II include formulas wherein n is 2, Y is N, W is CR4, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted aryl, for example,
Figure imgf000009_0001
wherein X is CH, R is H or Ci-C6alkyl, Rb is H, Ci-C6alkyl, or C3-C6cycloalkyl, and RbA is absent, H, or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I-B and II-B are those wherein X is N, R5 is H or Ci-Cealkyl, R6 is H, Ci-Cealkyl, or C3-C6cycloalkyl, and R6A is absent, H, or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N.The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-B and II-B.
[0035] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is N, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted aryl, for example,
Figure imgf000009_0002
wherein X is CH, R is H or Ci-C6alkyl, Rb is H, Ci-C6alkyl, or C3-C6cycloalkyl, and RbA is absent, H, or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I-C and II-C are those wherein X is N, R5 is H or Ci-Cealkyl, R6 is H, Ci-Cealkyl, or C3-C6cycloalkyl, and R6 is absent, H, or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-C and II-C.
[0036] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is N, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted aryl, for example,
Figure imgf000010_0001
wherein X is CH, R is H or Ci-Cealkyl (e.g., methyl), R is H, Ci-Cealkyl, or C3-Cecycloalkyl, and R6A is absent, H, or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I- D and II-D are those wherein X is N, R5 is H or Ci-C6alkyl, R6 is H, Ci-C6alkyl, or C3- Cecycloalkyl, and R6A is absent, H, or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts,
stereoisomers, tautomers, and solvates of formulas I-D and II-D.
[0037] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is CR4, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted C3-Ci2cycloalkyl, for exam le,
Figure imgf000010_0002
wherein X is CH, R is H or Ci-C6alkyl, Rb is H, Ci-C6alkyl, or C3-C6cycloalkyl, and RbA is absent, H, or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I-E and II-E are those wherein X is N, R5 is H or Ci-Cealkyl, R6 is H, Ci-Cealkyl, or C3-C6cycloalkyl, and Rf is absent, H, or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-E and II-E.
[0038] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is CR4, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted C3- C cycloalkyl, for exam le,
Figure imgf000011_0001
wherein X is CH, R is H or Ci-C6alkyl, Rb is H, Ci-C6alkyl, or C3-C6cycloalkyl, and RbA is absent, H, or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I-F and II-F are those wherein X is N, R5 is H or Ci-Cealkyl, R6 is H, Ci-Cealkyl, or C3-C6cycloalkyl, and R^ is absent, H, or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-F and II-F.
[0039] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is N, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted C3- Ci2cycloalkyl, for ex mple,
Figure imgf000011_0002
(II-G) wherein X is CH, R is H or Ci-C6alkyl, Rb is H, Ci-C6alkyl, or C3-C6cycloalkyl, and RbA is absent, H, or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I-G and II-G are those wherein X is N, R5 is H or Ci-C6alkyl, R6 is H, Ci-C6alkyl, or C3-C6cycloalkyl, and R67 is absent, H, or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-G and II-G.
[0040] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is N, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted C3- Ci2cycloalkyl for exam le,
Figure imgf000012_0001
wherein X is CH, R is H or Ci-Cealkyl (e.g., methyl), R is H, Ci-Cealkyl, or C3-C6cycloalkyl, and R6A is absent, H, or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I- H and II-H are those wherein X is N, R5 is H or Ci-C6alkyl, R6 is H, Ci-C6alkyl, or C3- Cecycloalkyl, and R6A is absent, H, or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts,
stereoisomers, tautomers, and solvates of formulas I-H and II-H.
[0041] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is CR4, V is Cialkylene, R2 is H, Z is a bond, and B is substituted or unsubstituted C3- C cycloalkyl, for example,
Figure imgf000013_0001
wherein X is CH, and R is H or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I-J and II-J are those wherein X is N and R5 is H or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-J and II-J.
[0042] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is CR4, V is Cialkylene, R2 is H, Z is a bond, and B is substituted or unsubstituted C3- C cycloalkyl, for example,
Figure imgf000013_0002
wherein X is CH an R is H or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I-K and II-K are those wherein X is N and R5 is H or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-K and II-K.
[0043] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is CH, W is N, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted C3- C cycloalkyl, for example,
Figure imgf000014_0001
wherein X is CH and R is H or Ci-Cealkyl. Other preferred embodiments of compounds of formulas I-L and II-L are those wherein X is N and R5 is H or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-L and II-L.
[0044] Sub-formulas of formula I and formula II include formulas wherein n is 2, Y is N, W is N, V is Cialkylene, R2 is H, Z is Cialkylene, and B is substituted or unsubstituted C3- Ci2cycloalkyl for example,
Figure imgf000014_0002
wherein X is CH and R is H or Ci-Cealkyl (e.g., methyl) Other preferred embodiments of compounds of formulas I-M and II-M are those wherein X is N and R5 is H or Ci-Cealkyl. In other aspects, X is CH and T is CH. In some aspects, X is N and T is CH. In other aspects, X is CH and T is N. In other aspects, X is N and T is N. The invention also encompasses the pharmaceutically acceptable salts, stereoisomers, tautomers, and solvates of formulas I-M and II- M.
[0045] In another embodiment, the compounds of the invention have human IDO IC50 values > 50 nM. In another embodiment, the compounds of the invention have human IDO IC50 values < 50 nM. In another embodiment, the compounds of the invention have human IDO IC50 values < 5 nM. OTHER EMBODIMENTS OF THE INVENTION
[0046] In another embodiment, the present invention provides a composition comprising one or more compounds of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
[0047] In another embodiment, the present invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and at least one of the compounds of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
[0048] In another embodiment, the present invention provides a pharmaceutical composition, comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
[0049] In another embodiment, the present invention provides a process for making a compound of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
[0050] In another embodiment, the present invention provides an intermediate for making a compound of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
[0051] In another embodiment, the present invention provides a method for the treatment and/or prophylaxis of various types of cancer, viral infections and/or autoimmune diseases, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of one or more compounds of the present invention and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent, such as a chemotherapeutic agent or a signal transductor inhibitor.
[0052] In another embodiment, the present invention provides a compound of the present invention, and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof, for use in therapy.
[0053] In another embodiment, the present invention provides a combined preparation of a compound of the present invention, and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof, and additional therapeutic agent(s) for simultaneous, separate or sequential use in therapy. [0054] In another embodiment, the present invention provides a combined preparation of a compound of the present invention, and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof, and additional therapeutic agent(s) for simultaneous, separate or sequential use in the treatment and/or prophylaxis of multiple diseases or disorders associated with the enzymatic activity of IDO.
[0055] In another aspect, the invention provides a method of treating a patient suffering from or susceptible to a medical condition that is sensitive to enzymatic activity of IDO. A number of medical conditions can be treated. The method comprises administering to the patient a therapeutically effective amount of a composition comprising a compound described herein and/or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof. For example, the compounds described herein may be used to treat or prevent viral infections, proliferative diseases (e.g. , cancer), and autoimmune diseases.
THERAPEUTIC APPLICATIONS
[0056] The compounds and pharmaceutical compositions of the present invention are useful in treating or preventing any disease or conditions that are sensitive to enzymatic activity of IDO. These include viral and other infections (e.g., skin infections, GI infection, urinary tract infections, genito-urinary infections, systemic infections), proliferative diseases (e.g. , cancer), and autoimmune diseases (e.g. , rheumatoid arthritis, lupus). The compounds and pharmaceutical compositions may be administered to animals, preferably mammals (e.g. , domesticated animals, cats, dogs, mice, rats), and more preferably humans. Any method of administration may be used to deliver the compound or pharmaceutical composition to the patient. In certain embodiments, the compound or pharmaceutical composition is administered orally. In other embodiments, the compound or pharmaceutical composition is administered parenterally.
[0057] Compounds of the invention can modulate activity of the enzyme indoleamine- 2,3-dioxygenase (IDO). The term "modulate" is meant to refer to an ability to increase or decrease activity of an enzyme or receptor. Accordingly, compounds of the invention can be used in methods of modulating IDO by contacting the enzyme with any one or more of the compounds or compositions described herein. In some embodiments, compounds of the present invention can act as inhibitors of IDO. In further embodiments, the compounds of the invention can be used to modulate activity of IDO in cell or in an individual in need of modulation of the enzyme by administering a modulating (e.g. , inhibiting) amount of a compound of the invention.
[0058] Compounds of the invention can inhibit activity of the enzyme indoleamine-2,3- dioxygenase (IDO). For example, the compounds of the invention can be used to inhibit activity of IDO in cell or in an individual in need of modulation of the enzyme by administering an inhibiting amount of a compound of the invention.
[0059] The present invention further provides methods of inhibiting the degradation of tryptophan in a system containing cells expressing IDO such as a tissue, living organism, or cell culture. In some embodiments, the present invention provides methods of altering (e.g., increasing) extracellular tryptophan levels in a mammal by administering an effective amount of a compound of composition provided herein. Methods of measuring tryptophan levels and tryptophan degradation are routine in the art.
[0060] The present invention further provides methods of inhibiting
immunosuppression such as IDO-mediated immunosuppression in a patient by administering to the patient an effective amount of a compound or composition recited herein. IDO-mediated immunosuppression has been associated with, for example, cancers, tumor growth, metastasis, viral infection, and viral replication.
[0061] The present invention further provides methods of treating diseases associated with activity or expression, including abnormal activity and/or overexpression, of IDO in an individual (e.g., patient) by administering to the individual in need of such treatment a therapeutically effective amount or dose of a compound of the present invention or a
pharmaceutical composition thereof. Example diseases can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the IDO enzyme, such as over expression or abnormal activity. An IDO-associated disease can also include any disease, disorder or condition that can be prevented, ameliorated, or cured by modulating enzyme activity. Examples of IDO-associated diseases include cancer, viral infection such as HIV infection, HCV infection, depression, neurodegenerative disorders such as Alzheimer's disease and Huntington's disease, trauma, age-related cataracts, organ transplantation (e.g., organ transplant rejection), and autoimmune diseases including asthma, rheumatoid arthritis, multiple sclerosis, allergic inflammation, inflammatory bowel disease, psoriasis and systemic lupus erythematosus.
[0062] As used herein, the term "cell" is meant to refer to a cell that is in vitro, ex vivo or in vivo. In some embodiments, an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal. In some embodiments, an in vitro cell can be a cell in a cell culture. In some embodiments, an in vivo cell is a cell living in an organism such as a mammal.
[0063] As used herein, the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, "contacting" the IDO enzyme with a compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having IDO, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the IDO enzyme.
[0064] The term "IDO inhibitor" refers to an agent capable of inhibiting the activity of indoleamine 2,3-dioxygenase (IDO) and thereby reversing IDO-mediated immunosuppression. The IDO inhibitor may inhibit IDOl and/or ID02 (INDOL1). An IDO inhibitor may be a reversible or irreversible IDO inhibitor. "A reversible IDO inhibitor" is a compound that reversibly inhibits IDO enzyme activity either at the catalytic site or at a non-catalytic site and "an irreversible IDO inhibitor" is a compound that irreversibly destroys IDO enzyme activity.
[0065] Types of cancers that may be treated with the compounds of this invention include, but are not limited to, brain cancers, skin cancers, bladder cancers, ovarian cancers, breast cancers, gastric cancers, pancreatic cancers, prostate cancers, colon cancers, blood cancers, lung cancers and bone cancers. Examples of such cancer types include neuroblastoma, intestine carcinoma such as rectum carcinoma, colon carcinoma, familiar adenomatous polyposis carcinoma and hereditary non-polyposis colorectal cancer, esophageal carcinoma, labial carcinoma, larynx carcinoma, hypopharynx carcinoma, tongue carcinoma, salivary gland carcinoma, gastric carcinoma, adenocarcinoma, medullary thyroid carcinoma, papillary thyroid carcinoma, renal carcinoma, kidney parenchymal carcinoma, ovarian carcinoma, cervix carcinoma, uterine corpus carcinoma, endometrium carcinoma, chorion carcinoma, pancreatic carcinoma, prostate carcinoma, testis carcinoma, breast carcinoma, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, Hodgkin lymphoma, non-Hodgkin lymphoma, Burkitt lymphoma, acute lymphatic leukemia (ALL), chronic lymphatic leukemia (CLL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), adult T-cell leukemia lymphoma, diffuse large B-cell lymphoma (DLBCL), hepatocellular carcinoma, gall bladder carcinoma, bronchial carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, multiple myeloma, basalioma, teratoma, retinoblastoma, choroid melanoma, seminoma, rhabdomyosarcoma, craniopharyngioma, osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma, fibrosarcoma, Ewing sarcoma and plasmacytoma.
[0066] Thus, according to another embodiment, the invention provides a method of treating an autoimmune disease by providing to a patient in need thereof a compound or composition of the present invention. Examples of such autoimmune diseases include, but are not limited to, collagen diseases such as rheumatoid arthritis, systemic lupus erythematosus, Sharp's syndrome, CREST syndrome (calcinosis, Raynaud's syndrome, esophageal dysmotility, telangiectasia), dermatomyositis, vasculitis (Morbus Wegener's) and Sjogren's syndrome, renal diseases such as Goodpasture's syndrome, rapidly -progressing glomerulonephritis and membranoproliferative glomerulonephritis type II, endocrine diseases such as type-I diabetes, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), autoimmune parathyroidism, pernicious anemia, gonad insufficiency, idiopathic Morbus Addison's, hyperthyreosis, Hashimoto's thyroiditis and primary myxedema, skin diseases such as pemphigus vulgaris, bullous pemphigoid, herpes gestationis, epidermolysis bullosa and erythema multiforme major, liver diseases such as primary biliary cirrhosis, autoimmune cholangitis, autoimmune hepatitis type-1, autoimmune hepatitis type-2, primary sclerosing cholangitis, neuronal diseases such as multiple sclerosis, myasthenia gravis, myasthenic Lambert-Eaton syndrome, acquired neuromyotomy, Guillain-Barre syndrome (Muller-Fischer syndrome), stiff-man syndrome, cerebellar degeneration, ataxia, opsoclonus, sensoric neuropathy and achalasia, blood diseases such as autoimmune hemolytic anemia, idiopathic thrombocytopenic purpura (Morbus Werlhof), infectious diseases with associated autoimmune reactions such as AIDS, malaria and Chagas disease.
[0067] One or more additional pharmaceutical agents or treatment methods such as, for example, anti-viral agents, chemotherapeutics or other anticancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g. , IL2 and GM-CSF), and/or tyrosine kinase inhibitors can be optionally used in combination with the compounds of the present invention for treatment of IDO-associated diseases, disorders or conditions. The agents can be combined with the present compounds in a single dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
[0068] Suitable chemotherapeutic or other anticancer agents include, for example, alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) such as uracil mustard, chlormethine,
cyclophosphamide (CYTOXAN®), ifosfamide, melphalan, chlorambucil, pipobroman, triethylene-melamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, and temozolomide.
[0069] In the treatment of melanoma, suitable agents for use in combination with the compounds of the present invention include: dacarbazine (DTIC), optionally, along with other chemotherapy drugs such as carmustine (BCNU) and cisplatin; the "Dartmouth regimen", which consists of DTIC, BCNU, cisplatin and tamoxifen; a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOY®. Compounds according to the invention may also be combined with immunotherapy drugs, including cytokines such as interferon alpha, interleukin 2, and tumor necrosis factor (TNF) in the treatment of melanoma.
[0070] Compounds of the invention may also be used in combination with vaccine therapy in the treatment of melanoma. Anti-melanoma vaccines are, in some ways, similar to the anti-virus vaccines which are used to prevent diseases caused by viruses such as polio, measles, and mumps. Weakened melanoma cells or parts of melanoma cells called antigens may be injected into a patient to stimulate the body's immune system to destroy melanoma cells.
[0071] Melanomas that are confined to the arms or legs may also be treated with a combination of agents including one or more compounds of the invention, using a hyperthermic isolated limb perfusion technique. This treatment protocol temporarily separates the circulation of the involved limb from the rest of the body and inj ects high doses of chemotherapy into the artery feeding the limb, thus providing high doses to the area of the tumor without exposing intemal organs to these doses that might otherwise cause severe side effects. Usually the fluid is warmed to 102° to 104° F. Melphalan is the drug most often used in this chemotherapy procedure. This can be given with another agent called tumor necrosis factor (TNF).
[0072] Suitable chemotherapeutic or other anticancer agents include, for example, antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatine, and gemcitabine.
[0073] Suitable chemotherapeutic or other anticancer agents further include, for example, certain natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) such as vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, ara-C, paclitaxel (Taxol), mithramycin, deoxycoformycin, mitomycin-C, L-asparaginase, interferons (especially IFN-a), etoposide, and teniposide.
[0074] Other cytotoxic agents include navelbene, CPT-11 , anastrazole, letrazole, capecitabine, reloxafine, and droloxafine.
[0075] Also suitable are cytotoxic agents such as epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes such as cisplatin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors.
[0076] Other anticancer agent(s) include antibody therapeutics such as trastuzumab (HERCEPTIN®), antibodies to costimulatory molecules such as CTLA-4, 4-1BB and PD-1, or antibodies to cytokines (IL-10 or TGF-β).
[0077] Other anticancer agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4.
[0078] Other anticancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer.
[0079] Anticancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses.
[0080] The pharmaceutical composition of the invention may optionally include at least one signal transduction inhibitor (STI). A "signal transduction inhibitor" is an agent that selectively inhibits one or more vital steps in signaling pathways, in the normal function of cancer cells, thereby leading to apoptosis. Suitable STIs include, but are not limited to: (i) bcr/abl kinase inhibitors such as, for example, STI 571 (GLEEVEC®); (ii) epidermal growth factor (EGF) receptor inhibitors such as, for example, kinase inhibitors (IRESSA®, SSI-774) and antibodies (Imclone: C225 [Goldstein et al., Clin. Cancer Res., 1 : 1311-1318 (1995)], and Abgenix: ABX-EGF); (iii) her-2/neu receptor inhibitors such as farnesyl transferase inhibitors (FTI) such as, for example, L-744,832 (Kohl et al, Nat. Med., 1(8): 792-797 (1995)); (iv) inhibitors of Akt family kinases or the Akt pathway, such as, for example, rapamycin (see, for example, Sekulic et al, Cancer Res. , 60:3504-3513 (2000)); (v) cell cycle kinase inhibitors such as, for example, flavopiridol and UCN-Ol (see, for example, Sausville, Curr. Med. Chem. Anti- Canc. Agents, 3:47-56 (2003)); and (vi) phosphatidyl inositol kinase inhibitors such as, for example, LY294002 (see, for example, Vlahos et al, J. Biol. Chem. , 269:5241-5248 (1994)). Alternatively, at least one STI and at least one IDO inhibitor may be in separate pharmaceutical compositions. In a specific embodiment of the present invention, at least one IDO inhibitor and at least one STI may be administered to the patient concurrently or sequentially. In other words, at least one IDO inhibitor may be administered first, at least one STI may be administered first, or at least one IDO inhibitor and at least one STI may be administered at the same time.
Additionally, when more than one IDO inhibitor and/or STI is used, the compounds may be administered in any order. [0081] The present invention further provides a pharmaceutical composition for the treatment of a chronic viral infection in a patient comprising at least one IDO inhibitor, optionally, at least one chemotherapeutic drug, and, optionally, at least one antiviral agent, in a pharmaceutically acceptable carrier. The pharmaceutical compositions may include at least one IDO inhibitor of the instant invention in addition to at least one established (known) IDO inhibitor. In a specific embodiment, at least one of the IDO inhibitors of the pharmaceutical composition is selected from the group consisting of compounds of formulas I and (II).
[0082] Also provided is a method for treating a chronic viral infection in a patient by administering an effective amount of the above pharmaceutical composition.
[0083] In a specific embodiment of the present invention, at least one IDO inhibitor and at least one chemotherapeutic agent may be administered to the patient concurrently or sequentially. In other words, at least one IDO inhibitor may be administered first, at least one chemotherapeutic agent may be administered first, or at least one IDO inhibitor and the at least one STI may be administered at the same time. Additionally, when more than one IDO inhibitor and/or chemotherapeutic agent is used, the compounds may be administered in any order.
Similarly, any antiviral agent or STI may also be administered at any point in comparison to the administration of an IDO inhibitor.
[0084] Chronic viral infections that may be treated using the present combinatorial treatment include, but are not limited to, diseases caused by: hepatitis C virus (HCV), human papilloma virus (HPV), cytomegalovirus (CMV), herpes simplex virus (HSV), Epstein-Barr virus (EBV), varicella zoster virus, Coxsackie virus, human immunodeficiency virus (HIV). Notably, parasitic infections (e.g., malaria) may also be treated by the above methods wherein compounds known to treat the parasitic conditions are optionally added in place of the antiviral agents.
[0085] In yet another embodiment, the pharmaceutical compositions comprising at least one IDO inhibitor of the instant invention may be administered to a patient to prevent arterial restenosis, such as after balloon endoscopy or stent placement. In a particular embodiment, the pharmaceutical composition further comprises at least one taxane (e.g. , paclitaxel (Taxol); see, e.g. , Scheller et al, Circulation, 110: 810-814 (2004)).
[0086] Suitable antiviral agents contemplated for use in combination with the compounds of the present invention can comprise nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors and other antiviral drugs. [0087] Examples of suitable NRTIs include zidovudine (AZT); didanosine (ddl);
zalcitabine (ddC); stavudine (d4T); lamivudine (3TC); abacavir (1592U89); adefovir dipivoxil [bis(POM)-PMEA]; lobucavir (BMS-180194); BCH-I0652; emtricitabine [(-)-FTC]; beta-L-FD4 (also called beta-L-D4C and named beta-L-2',3'-dicleoxy-5-fluoro-cytidene); DAPD, ((-)-beta- D-2,6-diamino-purine dioxolane); and lodenosine (FddA). Typical suitable NNRTIs include nevirapine (BI-RG-587); delaviradine (BHAP, U-90152); efavirenz (DMP-266); PNU-142721; AG-1549; MKC-442 (l-(ethoxy-methyl)-5-(l-methylethyl)-6-(phenylmethyl)-(2,4(lH,3H)- pyrimidinedione); and (+)-calanolide A (NSC-675451) and B. Typical suitable protease inhibitors include saquinavir (Ro 31-8959); ritonavir (ABT-538); indinavir (MK-639); nelfinavir (AG-1343); amprenavir (141W94); lasinavir (BMS-234475); DMP-450; BMS-2322623;
ABT-378; and AG-1549. Other antiviral agents include hydroxyurea, ribavirin, IL-2, IL-12, pentafuside and Yissum Project No.11607.
Combination with an Immuno-Oncology Agent
[0088] Further provided herein are methods of treatment wherein a compound of formula I or formula II is administered with one or more immuno-oncology agents. The immuno-oncology agents used herein, also known as cancer immunotherapies, are effective to enhance, stimulate, and/or upregulate immune responses in a subject.
[0089] In one aspect, the Compound of formula I or formula II is sequentially administered prior to administration of the immuno-oncology agent. In another aspect, the Compound of formula I or formula II is administered concurrently with the immunology- oncology agent. In yet another aspect, the Compound of formula I or formula II is sequentially administered after administration of the immuno-oncology agent.
[0090] In another aspect, the Compound of formula I or formula II may be co- formulated with an immuno-oncology agent.
[0091] Immuno-oncology agents include, for example, a small molecule drug, antibody, or other biologic or small molecule. Examples of biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines. In one aspect, the antibody is a monoclonal antibody. In another aspect, the monoclonal antibody is humanized or human.
[0092] In one aspect, the immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co- inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators). [0093] Certain of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF). One important family of membrane-bound ligands that bind to co-stimulatory or co-inhibitory receptors is the B7 family, which includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7- H6. Another family of membrane bound ligands that bind to co-stimulatory or co-inhibitory receptors is the TNF family of molecules that bind to cognate TNF receptor family members, which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-lBB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fnl4, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTfiR, LIGHT, DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFRl, Lymphotoxin α/ΤΝΡβ, TNFR2, TNFa, LTfiR, Lymphotoxin a 1β2, FAS, FASL, RELT, DR6, TROY, NGFR.
[0094] In another aspect, the immuno-oncology agent is a cytokine that inhibits T cell activation (e.g. , IL-6, IL-10, TGF-β, VEGF, and other immunosuppressive cytokines) or a cytokine that stimulates T cell activation, for stimulating an immune response.
[0095] In one aspect, T cell responses can be stimulated by a combination of the Compound of formula I or formula II and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4, and (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-lBB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H.
[0096] Other agents that can be combined with the Compound of formula I or formula II for the treatment of cancer include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells. For example, the Compound of formula I or formula II can be combined with antagonists of KIR, such as lirilumab.
[0097] Yet other agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (WO 11/70024, WO 11/107553, WO 11/131407, WO 13/87699, WO 13/119716, WO 13/132044) or FPA-008 (WO 11/140249, WO 13/169264, WO 14/036357).
[0098] In another aspect, the Compound of formula I or formula II can be used with one or more of agonistic agents that ligate positive costimulatory receptors, blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency of anti-tumor T cells, agents that overcome distinct immune suppressive pathways within the tumor microenvironment (e.g. , block inhibitory receptor engagement (e.g. , PD-Ll/PD-1 interactions), deplete or inhibit Tregs (e.g. , using an anti-CD25 monoclonal antibody (e.g. , daclizumab) or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell anergy or exhaustion) and agents that trigger innate immune activation and/or inflammation at tumor sites.
[0099] In one aspect, the immuno-oncology agent is a CTLA-4 antagonist, such as an antagonistic CTLA-4 antibody. Suitable CTLA-4 antibodies include, for example, YERVOY® (ipilimumab) or tremelimumab.
[00100] In another aspect, the immuno-oncology agent is a PD-1 antagonist, such as an antagonistic PD-1 antibody. Suitable PD-1 antibodies include, for example, OPDIVO® (nivolumab), KEYTRUDA® (pembrolizumab), or MEDI-0680 (AMP-514; WO 2012/145493). The immuno-oncology agent may also include pidilizumab (CT-011), though its specificity for PD-1 binding has been questioned. Another approach to target the PD-1 receptor is the recombinant protein composed of the extracellular domain of PD-L2 (B7-DC) fused to the Fc portion of IgGl, called AMP-224
[00101] In another aspect, the immuno-oncology agent is a PD-Ll antagonist, such as an antagonistic PD-Ll antibody. Suitable PD-Ll antibodies include, for example, MPDL3280A (RG7446; WO 2010/077634), durvalumab (MEDI4736), BMS-936559 (WO 2007/005874), and MSB0010718C (WO 2013/79174).
[00102] In another aspect, the immuno-oncology agent is a LAG-3 antagonist, such as an antagonistic LAG-3 antibody. Suitable LAG3 antibodies include, for example, BMS-986016 (WO 10/19570, WO 14/08218), or IMP-731 or IMP-321 (WO 08/132601, WO 09/44273).
[00103] In another aspect, the immuno-oncology agent is a CD137 (4-1BB) agonist, such as an agonistic CD 137 antibody. Suitable CD 137 antibodies include, for example, urelumab and PF-05082566 (WO 12/32433).
[00104] In another aspect, the immuno-oncology agent is a GITR agonist, such as an agonistic GITR antibody. Suitable GITR antibodies include, for example, BMS-986153, BMS-986156, TRX-518 (WO 06/105021, WO 09/009116) and MK-4166 (WO 11/028683).
[00105] In another aspect, the immuno-oncology agent is an IDO antagonist. Suitable IDO antagonists include, for example, INCB-024360 (WO 2006/122150, WO 07/75598, WO 08/36653, WO 08/36642), indoximod, or NLG-919 (WO 09/73620, WO 09/1156652, WO 11/56652, WO 12/142237).
[00106] In another aspect, the immuno-oncology agent is an OX40 agonist, such as an agonistic OX40 antibody. Suitable OX40 antibodies include, for example, MEDI-6383 or MEDI-6469.
[00107] In another aspect, the immuno-oncology agent is an OX40L antagonist, such as an antagonistic OX40 antibody. Suitable OX40L antagonists include, for example, RG-7888 (WO 06/029879).
[00108] In another aspect, the immuno-oncology agent is a CD40 agonist, such as an agonistic CD40 antibody. In yet another embodiment, the immuno-oncology agent is a CD40 antagonist, such as an antagonistic CD40 antibody. Suitable CD40 antibodies include, for example, lucatumumab or dacetuzumab.
[00109] In another aspect, the immuno-oncology agent is a CD27 agonist, such as an agonistic CD27 antibody. Suitable CD27 antibodies include, for example, varlilumab.
[00110] In another aspect, the immuno-oncology agent is MGA271 (to B7H3) (WO 11/109400).
[00111] The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of IDO-associated diseases or disorders, obesity, diabetes and other diseases referred to herein which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
[00112] The combination therapy is intended to embrace administration of these therapeutic agents in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each therapeutic agent or in multiple, single dosage forms for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally. Alternatively, for example, all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection. Combination therapy also can embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients and non-drug therapies (e.g., surgery or radiation treatment). Where the combination therapy further comprises a non-drug treatment, the non-drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and non-drug treatment is achieved. For example, in appropriate cases, the beneficial effect is still achieved when the non- drug treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks.
PHARMACEUTICAL COMPOSITIONS AND DOSING
[00113] The invention also provides pharmaceutically acceptable compositions which comprise a therapeutically effective amount of one or more of the compounds of formula I and/or formula II, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents, and optionally, one or more additional therapeutic agents described above.
[00114] The compounds of this invention can be administered for any of the uses described herein by any suitable means, for example, orally, such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups, and emulsions; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection, or infusion techniques (e.g. , as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice. [00115] The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[00116] The phrase "pharmaceutically acceptable carrier" as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
[00117] The term "pharmaceutical composition" means a composition comprising a compound of the invention in combination with at least one additional pharmaceutically acceptable carrier. A "pharmaceutically acceptable carrier" refers to media generally accepted in the art for the delivery of biologically active agents to animals, in particular, mammals, including, i.e. , adjuvant, excipient or vehicle, such as diluents, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
[00118] Pharmaceutically acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art. These include, without limitation: the type and nature of the active agent being formulated; the subject to which the agent-containing composition is to be administered; the intended route of administration of the composition; and the therapeutic indication being targeted. Pharmaceutically acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms. Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g. , stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art. Descriptions of suitable pharmaceutically acceptable carriers, and factors involved in their selection, are found in a variety of readily available sources such as, for example, Allen, Jr., L.V. et al, Remington: The Science and Practice of Pharmacy (2 Volumes), 22nd Edition, Pharmaceutical Press (2012).
[00119] The dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.
[00120] By way of general guidance, the daily oral dosage of each active ingredient, when used for the indicated effects, will range between about 0.001 to about 5000 mg per day, preferably between about 0.01 to about 1000 mg per day, and most preferably between about 0.1 to about 250 mg per day. Intravenously, the most preferred doses will range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion. Compounds of this invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
[00121] The compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, e.g. , oral tablets, capsules, elixirs, and syrups, and consistent with conventional pharmaceutical practices.
[00122] Dosage forms (pharmaceutical compositions) suitable for administration may contain from about 1 milligram to about 2000 milligrams of active ingredient per dosage unit. In these pharmaceutical compositions the active ingredient will ordinarily be present in an amount of about 0.1-95% by weight based on the total weight of the composition.
[00123] A typical capsule for oral administration contains at least one of the compounds of the present invention (250 mg), lactose (75 mg), and magnesium stearate (15 mg). The mixture is passed through a 60 mesh sieve and packed into a No. 1 gelatin capsule.
[00124] A typical injectable preparation is produced by aseptically placing at least one of the compounds of the present invention (250 mg) into a vial, aseptically freeze-drying and sealing. For use, the contents of the vial are mixed with 2 mL of physiological saline, to produce an injectable preparation.
[00125] The present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of the present invention, alone or in combination with a pharmaceutical carrier. Optionally, compounds of the present invention can be used alone, in combination with other compounds of the invention, or in combination with one or more other therapeutic agent(s), e.g. , an anticancer agent or other pharmaceutically active material.
[00126] Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
[00127] Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
[00128] The selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
[00129] A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
[00130] In general, a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
[00131] If desired, the effective daily dose of the active compound may be
administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. In certain aspects of the invention, dosing is one administration per day. [00132] While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation
(composition).
DEFINITIONS
[00133] Unless specifically stated otherwise herein, references made in the singular may also include the plural. For example, "a" and "an" may refer to either one, or one or more.
[00134] Unless otherwise indicated, any heteroatom with unsatisfied valences is assumed to have hydrogen atoms sufficient to satisfy the valences.
[00135] Throughout the specification and the appended claims, a given chemical formula or name shall encompass all stereo and optical isomers and racemates thereof where such isomers exist. Unless otherwise indicated, all chiral (enantiomeric and diastereomeric) and racemic forms are within the scope of the invention. Many geometric isomers of C=C double bonds, C=N double bonds, ring systems, and the like can also be present in the compounds, and all such stable isomers are contemplated in the present invention. Cis- and trans- (or E- and Z-) geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms. The present compounds can be isolated in optically active or racemic forms. Optically active forms may be prepared by resolution of racemic forms or by synthesis from optically active starting materials. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention. When enantiomeric or diastereomeric products are prepared, they may be separated by conventional methods, for example, by chromatography or fractional crystallization. Depending on the process conditions the end products of the present invention are obtained either in free (neutral) or salt form. Both the free form and the salts of these end products are within the scope of the invention. If so desired, one form of a compound may be converted into another form. A free base or acid may be converted into a salt; a salt may be converted into the free compound or another salt; a mixture of isomeric compounds of the present invention may be separated into the individual isomers. Compounds of the present invention, free form and salts thereof, may exist in multiple tautomeric forms, in which hydrogen atoms are transposed to other parts of the molecules and the chemical bonds between the atoms of the molecules are consequently rearranged. It should be understood that all tautomeric forms, insofar as they may exist, are included within the invention. [00136] For purposes of clarity and in accordance with standard convention in the art, the symbol ¾ is used in formulas and tables to show the bond that is the point of attachment of the moiety or substituent to the core/nucleus of the structure.
[00137] Additionally, for purposes of clarity, where a substituent has a dash (-) that is not between two letters or symbols; this is used to indicate a point of attachment for a substituent. For example, -CONH2 is attached through the carbon atom.
[00138] Additionally, for purposes of clarity, when there is no substituent shown at the end of a solid line, this indicates that there is a methyl (CH3) group connected to the bond.
[00139] As used herein, the terms "alkyl" and "alkylene" (also refered to as "alk") are intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. For example, "C1-C6 alkyl" or "Ci-6 alkyl" denotes alkyl having 1 to 6 carbon atoms. Example alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g. , n-propyl and isopropyl), butyl (e.g. , n-butyl, isobutyl, t- butyl), and pentyl (e.g. , n-pentyl, isopentyl, neopentyl). "Cl -C6alkylene" denotes alkylene having 1 to 6 carbon atoms. Example alkylene groups include, but are not limited to, methylene (-CH2-), ethylene (-CH2CH2-), and the like.
[00140] As used herein, "aryl" refers to an aromatic ring system which includes, but not limited to phenyl, biphenyl, indanyl, 1 -naphthyl, 2-naphthyl and terahydronaphthyl.
[00141] "Halo" or "halogen" includes fluoro, chloro, bromo, and iodo.
[00142] "Haloalkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogens. Examples of haloalkyl include, but are not limited to, fluoromethyl,
difiuoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2,2- trifluoroethyl, heptafluoropropyl, and heptachloropropyl. Examples of haloalkyl also include "fluoroalkyl" that is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more fluorine atoms.
[00143] The term "cycloalkyl" refers to hydrocarbon rings having the indicated number of ring atoms, e.g., C3-C6cycloalkyl or C3-Ci2cycloalkyl, and being fully saturated. "Cycloalkyl" is also meant to refer to bicyclic and polycylic hydrocarbon rings such as, for example, bicyclo[2.2.1]heptane, bicyclo[2.2.2] octane, and adamantyl. cycloalkyl is intended to include C3, C4, C5, and e cycloalkyl groups. Example cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and norbomyl. [00144] As referred to herein, the term "substituted" means that at least one hydrogen atom is replaced with a non-hydrogen group, provided that normal valencies are maintained and that the substitution results in a stable compound.
[00145] As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic groups such as amines; and alkali or organic salts of acidic groups such as carboxylic acids. The pharmaceutically acceptable salts include the conventional nontoxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric; and the salts prepared from organic acids such as acetic, propionic, succinic, gly colic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, and isethionic, and the like.
[00146] The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Allen, Jr., L.V., ed., Remington: The Science and Practice of Pharmacy, 22nd Edition,
Pharmaceutical Press, London, UK (2012). The disclosure of which is hereby incorporated by reference.
[00147] In addition, compounds of formula I and formula II may have prodrug forms. Any compound that will be converted in vivo to provide the bioactive agent (i. e. , a compound of formula I or II) is a prodrug within the scope and spirit of the invention. Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see:
a) Bundgaard, H., ed., Design of Prodrugs, Elsevier (1985), and Widder, K. et al., eds., Methods in Enzymology, 1 12:309-396, Academic Press (1985);
b) Bundgaard, H., Chapter 5: "Design and Application of Prodrugs", A Textbook of Drug Design and Development, pp. 113-191 , Krogsgaard-Larsen, P. et al, eds., Harwood Academic Publishers (1991); c) Bundgaard, H., Adv. Drug Deliv. Rev. , 8: 1-38 (1992);
d) Nielsen, N.M. et al, J. Pharm. Sci., 77:285 (1988);
e) Kakeya, N. et al, Chem. Pharm. Bull , 32:692 (1984); and
g) Rautio, J., ed., Prodrugs and Targeted Delivery ftlethods and Principles in Medicinal Chemistry), Vol. 47, Wiley -VCH (2011).
[00148] Compounds containing a carboxy group can form physiologically hydrolyzable esters that serve as prodrugs by being hydrolyzed in the body to yield formula I or formula II compounds per se. Such prodrugs are preferably administered orally since hydrolysis in many instances occurs principally under the influence of the digestive enzymes. Parenteral administration may be used where the ester per se is active, or in those instances where hydrolysis occurs in the blood. Examples of physiologically hydrolyzable esters of compounds of formula I or formula II include Ci_6alkyl, Chalky lbenzyl, 4-methoxybenzyl, indanyl, phthalyl, methoxymethyl, Ci_6 alkanoyloxy-Ci_6alkyl (e.g. , acetoxymethyl, pivaloyloxymethyl or propionyloxymethyl), Ci_6alkoxycarbonyloxy-Ci_6alkyl (e.g. , methoxycarbonyl-oxymethyl or ethoxycarbonyloxymethyl, glycyloxymethyl, phenylglycyloxymethyl, (5-methyl-2-oxo-l,3- dioxolen-4-yl)-methyl), and other well-known physiologically hydrolyzable esters used, for example, in the penicillin and cephalosporin arts. Such esters may be prepared by conventional techniques known in the art.
[00149] Preparation of prodrugs is well known in the art and described in, for example, King, F.D., ed., Medicinal Chemistry: Principles and Practice, The Royal Society of Chemistry, Cambridge, UK (Second Edition, reproduced, 2006); Testa, B. et al, Hydrolysis in Drug and Prodrug Metabolism. Chemistry, Biochemistry and Enzymology, VCHA and Wiley -VCH, Zurich, Switzerland (2003); Wermuth, C.G., ed., The Practice of Medicinal Chemistry, Third Edition, Academic Press, San Diego, CA (2008).
[00150] The present invention is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include deuterium and tritium. Isotopes of carbon include 1 C and 14C. Isotopically - labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically -labeled reagent in place of the non-labeled reagent otherwise employed.
[00151] The term "solvate" means a physical association of a compound of this invention with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. The solvent molecules in the solvate may be present in a regular arrangement and/or a non-ordered arrangement. The solvate may comprise either a
stoichiometric or nonstoichiometric amount of the solvent molecules. "Solvate" encompasses both solution-phase and isolable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Methods of solvation are generally known in the art.
[00152] As used herein, the term "patient" refers to organisms to be treated by the methods of the present invention. Such organisms preferably include, but are not limited to, mammals (e.g. , murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably refers to humans.
[00153] As used herein, the term "effective amount" means that amount of a drug or pharmaceutical agent, i.e., a compound of the invention, that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician. Furthermore, the term "therapeutically effective amount" means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route. The term also includes within its scope amounts effective to enhance normal physiological function
[00154] As used herein, the term "treating" includes any effect, e.g. , lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.
[00155] For therapeutic use, salts of the compounds of the present invention are contemplated as being pharmaceutically acceptable. However, salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
METHODS OF PREPARATION
[00156] The compounds of the present invention may be prepared by methods such as those illustrated in the following Schemes utilizing chemical transformations known to those skilled in the art. Solvents, temperatures, pressures, and other reaction conditions may readily be selected by one of ordinary skill in the art. Starting materials are commercially available or readily prepared by one of ordinary skill in the art. These Schemes are illustrative and are not meant to limit the possible techniques one skilled in the art may use to manufacture compounds disclosed herein. Different methods may be evident to those skilled in the art. Additionally, the various steps in the synthesis may be performed in an alternate sequence or order to give the desired compound(s). Further, the representation of the reactions in these Schemes as discrete steps does not preclude their being performed in tandem, either by telescoping multiple steps in the same reaction vessel or by performing multiple steps without purifying or characterizing the intermediate(s). In addition, many of the compounds prepared by the methods below can be further modified using conventional chemistry well known to those skilled in the art.
[00157] Reference can also be made to International Application Nos.
PCT/US2015/059271, PCT/US2015/059311, and PCT/US2015/059316.
[00158] References to many of these chemical transformations employed herein can be found in Smith, M.B. et al., March's Advanced Organic Chemistry Reactions, Mechanisms, and Structure, Fifth Edition, Wiley-Interscience, New York (2001), or other standard texts on the topic of synthetic organic chemistry. Certain transformations may require that reactive functional groups be masked by protecting group(s). A convenient reference which provides conditions for introduction, removal, and relative susceptibility to reaction conditions of these groups is Greene, T.W. et al, Protective Groups in Organic Synthesis, Third Edition, Wiley- Interscience, New York (1999).
[00159] Schemes 1-3 depict methods for preparing compounds of formula I. These methods can also be applied to preparation of compounds of the invention.
Scheme 1
Figure imgf000037_0001
[00160] Treatment of a phosphonoacetate ester (IV), with a base such as sodium hydride in a solvent such as THF (Scheme 1) followed by a ketone of the general structure III affords a trisubstituted olefin. Substituted analogs of IV (R4 is not H) afford tetrasubstitued olefins. This method and additional methods described below are transformations familiar to those skilled in the art of organic/medicinal chemistry. Alternative methods for olefination and the transformations described below are known and will be selected by one skilled in the art based on their applicability to the specific substrate under consideration. Reduction is accomplished by stirring or shaking a solution of the olefin in a suitable solvent under an atmosphere or more of H2 in the presence of a catalyst, normally palladium on carbon.
Hydrolysis of the ketal group affords a ketone of the general structure V. Typically, this is accomplished by heating with an aqueous acid such as HCl in the presence of a co-solvent such as THF. In addion to the cyclic ethylene gly col-based ketal shown, other cyclic and acyclic ketal protecting groups could be used. Ketones are deprotonated with bases such as LiHMDS and react with N-phenyltrifluoromethanesulfonimide or similar reagents to afford inflates of the general structure VI. These inflates participate in Suzuki couplings (T. Ishiyama, M. Murata, N. Miyaura, J. Org. Chem., 1995, 60, 7508-7510) with boronic acids or esters Ar-B(OR)2 such as VII to afford coupled products. Many variations on this reaction are known, but generally it involves heating the two substrates and a catalyst such as (Ph3P)4Pd in a solvent such as DMF with a base such as aq. potassium carbonate. Reduction of the olefin provides intermediate VIII. Intermediates VIII (and later intermediates) may be obtained as mixtures of cis and trans isomers. Methods for control of the stereochemical outcome of the above reactions are known to those familiar in the art of organic/medicinal chemistry. Additionally, methods for the separation of these isomers are known and described in detail in the synthetic examples. If required, the group R4 can be appended by alkylation of intermediate VIII. Methods for control of the absolute stereochemistry of the resulting asymmetric center are known to those familiar with the art, as are chiral separation methods. Saponification of the ester by heating with aq. LiOH or a similar base, generally in the presence of an organic co-solvent such as THF affords carboxylic acids IX. Treatment of acids IX with amines X under standard peptide coupling conditions will afford compounds of the invention I. For a recent review of peptide coupling methods see: Ayman El-Faham and Fernando Albericio. ChemRev. 2011, 111, 6557-6602.
Scheme 2
Figure imgf000038_0001
Piperidine and pyrrolidine esters XI are known compounds and can undergo SNAT reactions with heteroaryl halides of general structure XII to afford intermediates XIII (Y = N). These intermediates may be transformed to compounds of the invention as described herein. Scheme 3
Figure imgf000039_0001
Scheme 3 illustrates a method for controlling the absolute stereochemistry of
intermediate XIX and materials arising from it. Saponification of esters XIV provides carboxylic acids XV. Treatment of these acids with an acid chloride such as pivaloyl chloride provides a mixed anhydride intermediate. In a separate vessel, an optically pure oxazolidinone of known stereochemistry and general structure XVI is deprotonated by treatment with a strong base such as n-BuLi. These activated species are combined to form the acyloxazolidinone XVII which is deprotonated by bases such as NaHMDS. Alkylation of the resulting enolate proceeds with predictable control of stereochemistry at the newly-formed center to provide materials XVIII. Removal of the chiral auxiliary to give optically-active carboxylic acids XIX is accomplished by treatment with a solution of basic hydrogen peroxide. For a review of the history and scope scope of this reaction see: D. A. Evans, M. D. Ennis, D. J. Mathre. J. Am. Chem. Soc, 1982, 104 (6), pp 1737-1739. Compounds of general structure XIX can be converted to compounds of the invention by the methods described herein.
EXAMPLES
[00161] The following Examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention, nor are they intended to represent that the experiments below were performed or that they are all of the experiments that may be performed. It is to be understood that exemplary descriptions written in the present tense were not necessarily performed, but rather that the descriptions can be performed to generate data and the like of a nature described therein. Efforts have been made to ensure accuracy with respect to numbers used (e.g. , amounts, temperature, etc.), but some experimental errors and deviations should be accounted for.
[00162] Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Celsius (°C), and pressure is at or near atmospheric. Standard abbreviations are used, including the following: wt = wildtype; bp = base pair(s); kb = kilobase(s); nt = nucleotides(s); aa = amino acid(s); s or sec = second(s); min = minute(s); h or hr = hour(s); ng = nanogram; μg = microgram; mg = milligram; g = gram; kg = kilogram; dl or dL = deciliter; ul or = microliter; ml or mL = milliliter; 1 or L = liter; μΜ = micromolar; mM = millimolar; M = molar; kDa = kilodalton; i.m. = intramuscular(ly); i.p. = intraperitoneal(ly); SC or SQ = subcutaneous(ly); QD = daily; BID = twice daily; QW = weekly; QM = monthly; HPLC = high performance liquid chromatography; BW = body weight; U = unit; ns = not statistically significant; PBS = phosphate-buffered saline; IHC =
immunohistochemistry; DMEM = Dulbecco's Modification of Eagle's Medium; EDTA = ethylenediaminetetraacetic acid.
[00163] Analytical HPLC/MS was performed using the following methods:
Method A: Column: Waters Acquity UPLC BEH C 18, 2.1 x 50 mm, 1.7-μιη particles; Mobile Phase A: 5:95 acetonitrile:water with 0.1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile:water with 0.1% trifluoroacetic acid; Temperature: 50°C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 220 nm.
Method B: Waters Acquity SDS using the following method: Linear Gradient of 2% to98% solvent B over 1.7 min; UV visualization at 220 nm; Column: BEH C18 2.1 mm x 50 mm; 1.7 um particle (Heated to Temp. 50 °C); Flow rate: 0.8 ml/min; Mobile phase A: 100% Water, 0.05% TFA; Mobile phase B: 100% Acetonitrile, 0.05% TFA.
Method C: Column: Waters Acquity UPLC BEH C18, 2.1 x 50 mm, 1.7-μιη particles; Mobile Phase A: 5:95 acetonitrile: water with 10 mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate; Temperature: 50°C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 220 nm. Preparation of Intermediates
Preparation A:
Figure imgf000041_0001
[00164] To a stirred solution of l,4-dioxaspiro[4.5]decan-8-one (300 g, 1920.86 mmol, l.Oeq) and phenyltrifluoromethanesulfonimide (823.47 g, 2305.03 mmol, 1.2eq) in MTBE (7.5 L) under N2 at -78 °C was added 2.0 M NaHMDS in THF (1152.2 mL, 2305.03 mmol, 1.2eq) over 70 minutes, and the mixture was stirred for an additional 60 minutes. The reaction mixture was warmed to room temperature and stirred overnight until TLC showed complete consumption of the starting material. The mixture was quenched with aqueous KHSO4 (100 ml), filtrated to remove the solid and concentrated the filtrate completely. To the residue was added 3 L MTBE, then washed with 5% NaOH (1.5 L X 3). The organic phase was concentrated to obtain 567 g crude Preparation A (light yellow oil, yield 102%). The crude can be used directly in next step without further purification.
Preparation A: ^ NMR^OO MHz ,CDCh): δ (ppm) 5.65 (t, J=4.0 Hz, 1H), 3.98 (d, J=1.5 Hz, 4H), 2.53 (s,2H), 2.40 (s, 2H), 1.90 (t, J=6.6 Hz, 2H)
Preparation B:
Figure imgf000041_0002
[00165] A mixture of crude Preparation A (600 g,2.08mol ,leq), Β2ΡΠ12 (687.1 g, 2.71 mol, 1.3eq), KOAc (613 g, 6.24 mol, 3eq) , NaBr (86 g 0.833mol,0.4 eq) and Pd(dppf)Cb (76 g, 0.1 mol, 0.05eq) in dioxane (6.5 L) was heated to reflux overnight. Once the reaction was complete, the mixture was concentrated and purified by FCC (2% - 10% - 20% EtOAc/PE) to give Preparation B (369g, 66%).
Preparation B: LC-MS: 267.1 (M+l)+, XH NMR (400 MHz, CDCb) δ 6.46 (s, 1H), 3.98 (s, 4H), 2.37-2.35 (m, 4H), 1.74-1.60 (t, 2H), 1.24 (s, 12H). Preparation C:
Figure imgf000042_0001
[00166] A mixture of Preparation B (368g, 1.38 mol, 1.3eq), 4-Chloro-6- fluoroquinoline (195 g, 1.07 mol, leq), K2CO3 (445 g, 3.22 mol,3eq) and Pd(PPh3)4 (25 g, 22 mmol, 0.02eq) in dioxane-water (3L, 4: 1) was heated to reflux overnight. The solution was then concentrated and extracted with EtOAc. Purification by FCC (38% EtOAc/petrolium ether) gave Preparation C (236 g, 77%).
Preparation C: LC-MS: 286.1 (M+l)+, XH NMR (400 MHz, CDCh) δ 8.80-8.29 (d, 1H), 8.11-8.07 (q, 1H), 7.63-7.61 (q, 1H), 7.47-7.46 (q, 1H), 7.26-7.22(m,lH), 5.75-5.74 (m, 1H), 4.08-4.05 (m, 4H), 2.63-2.59 (m, 2H),2.59-2.53(m,2H), 2.0-1.97(m,2H).
Preparation D:
Figure imgf000042_0002
[00167] To Preparation C (125 g, 0.44 mol) in IPA (2 L) at 55 °C was added 10% Pd/C and the mixture was stirred under an atmosphere of H2 overnight. The mixture was filtered and concentrated to give crude Preparation D (130 g), which was used directly in the next step.
Preparation E:
Figure imgf000042_0003
[00168] Preparation D (100 g, 0.348 mol) was treated with 4 N HC1 ( 300 mL) in acetone (1200 mL) at 45 °C overnight. The mixture was monitored by TLC. Then the solution was then concentrated in vacuo. The residue was adjusted to pH 9 with 6 N NaOH and the mixture was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over anhydrous Na2S04, filtered and concentrated to give light yellow solid, which was then purified by silica gel column using hexanes and ethyl acetate (from 20 percent ethyl acetate to 70% ethyl acetate) to afford Preparation E as a white solid, (47 g+ 20 g mixture, yield >55%). Preparation E: LC-MS: 244.0 (M+l)+, lH NMR (400 MHz, CDCb) δ 8.84 (d, J = 4.6 Hz, 1H), 8.16 (dd, J = 9.3, 5.7 Hz, 1H), 7.72 (dd, J = 10.3, 2.8 Hz, 1H), 7.52 (ddd, J = 9.2, 7.8, 2.7 Hz, 1H), 7.29 (d, J = 4.6 Hz, 1H), 3.69 (ddd, J = 12.1, 9.0, 3.3 Hz, 1H), 2.77 - 2.54 (m, 4H), 2.37 (ddd, J = 13.4, 5.9, 3.0 Hz, 2H), 2.04 (qd, J = 12.6, 5.3 Hz, 2H).
Preparation F:
Figure imgf000043_0001
[00169] Preparation E (57.8 g, 237.8 mmol) was dissolved in EtOH (240 mL) and cooled to 0 °C. NaBH4 (9.94 g, 261.6 mmol) was added portionwise maintaining the temperature within a range of 0-10 °C (exothermic reaction). The resulting suspension was stirred for 20 minutes. An LC/MS of an aliquot of the reaction mixture indicated consumption of ketone (m/z ( +H)+ = 244). The reaction was quenched at 0 °C by the slow addition of acetone (58 mL) over 15 minutes (exotherm). The reaction was poured slowly onto 500 mL of saturated aqueous ammonium chloride and 500 g of ice. The resulting aqueous solution was extracted with EtOAc (3 x 300 mL) and the combined organic fractions were washed with saturated aqueous ammonium chloride (250 mL) and saturated aqueous sodium chloride (250 mL). The organic portion was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. Sufficient silica to adsorb the oil was added and diluted with 10 % MeOH in CH2CI2. A similar quantity of silica was used as a silica plug to purify the material. The silica plug was washed with 10 % MeOH in CH2CI2 until UV-active material no longer could be detected by TLC (7:3 EtOAc/Hexanes, Rf = 0.4). The filtrate was concentrated then suspended in 500 mL of toluene and concentrated again. Crude Preparation F was isolated as a yellow solid (58.2 g) that was used in the subsequent step without further purification.
Preparation G:
Figure imgf000043_0002
[00170] To Preparation F (58.2 g, 237.8 mmol) was added MeCN (125 mL) and pyridine (38.7 mL, 480 mmol) and the reaction mixture was cooled to 5 °C using an ice/water bath. Methanesulfonyl chloride (26.0 mL, 336 mmol) was added dropwise at 5 °C (exothermic reaction), the reaction mixture stirred for 1 hr at 5 °C and then brought up to room temperature and stirred for an additional 16 h during which time a white precipitate formed. The
heterogeneous mixture was quenched by the addition of saturated aqueous ammonium chloride (200 mL) and extracted with CH2CI2 (3 x 300 mL). The combined organic fractions were dried over anhydrous sodium sulfate and concentrated under reduced pressure. Excess pyridine was removed by azeotroping from toluene (3 x 300 mL). The crude material was recrystallized from LLO/MeOH as follows: 1 mL/mmol of H2O was added and the slurry was heated to 120 °C in an oil bath. MeOH was added until the solids went into solution (-0.5 L). After cooling white crystals were collected by filtration to give Preparation G (58.6 g, >20: 1 dr, 76 % over two steps), m/z (M+H)+ = 324.1. H-NMR (400 MHz; CDCL): δ 8.82 (dd, J = 4.6, 0.2 Hz, 1H), 8.15- 8.11 (m, 1H), 7.64-7.61 (m, 1H), 7.52-7.46 (m, 1H), 7.25 (s, 1H), 4.78 (tt, J = 10.9, 5.2 Hz, 1H), 3.24-3.16 (m, 1H), 3.07 (d, J= 1.0 Hz, 3H), 2.42-2.38 (m, 2H), 2.16-2.12 (m, 2H), 1.93-1.66 (m, 4H).
Preparation H:
Figure imgf000044_0001
[00171] Di-/er/-butyl malonate (33.5 mL, 150 mmol) was added dropwise to a stirred suspension of NaH (6.0 g, 60 % suspension in oil, 150 mmol) in 1,2-dimethoxy ethane (100 mL) under Ar, cooled in a water-ice bath. After stirring for 10 min, Preparation G (16.2 g, 50 mmol) was added and the reaction was heated at 85 °C for 20 h. After this time, acetic acid (100 mL) was added, the reaction flask was fitted with a distillation head and the temperature was raised to
130 °C. 1,2-dimethoxy ethane was distilled off under atmospheric pressure until the distillate was acidic (-100 mL). The distillation head was removed, a reflux condenser was attached, water (20 mL) was added and the reaction heated at 130 °C for 12 h. The reaction was concentrated under reduced pressure and poured onto 200 g of ice and 100 mL of saturated aqueous NaOAc.
Preparation H was isolated as a white solid by filtration and further dried by refluxing with toluene in a Dean-Stark apparatus (11.0 g, 76 %). m/z (M+H)+ = 288.2. ^-NMR (400 MHz; DMSO-d6): δ 12.05 (bs, 1H), 8.79 (d, J= 4.5 Hz, 1H), 8.06 (dd, J= 9.2, 5.8 Hz, 1H), 7.94 (dd, J = 11.0, 2.8 Hz, 1H), 7.66-7.61 (m, 1H), 7.50 (d, J = 4.6 Hz, 1H), 2.41 (d, J= 7.6 Hz, 2H), 2.28- 2.23 (m, 1H), 1.87-1.78 (m, 2H), 1.73-1.64 (m, 6H).
Preparation I:
Figure imgf000045_0001
[00172] To a solution of Preparation H (1.4 g, 4.8 mmol) in THF (15 mL) was added NEt (1.3 mL, 9.6 mmol). The reaction mixture was cooled to 0 °C and trimethylacetyl chloride (0.713 mL, 5.8 mmol) was added dropwise and the resulting solution stirred for 30 min at 0 °C. In a separate flask, (i?)-4-phenyloxazolidin-2-one (3, 1.01 g, 6.24 mmol) in THF (45 mL) at 0 °C was treated with 1 M LiHMDS solution in THF (dropwise addition of 6.24 mL, 6.24 mmol) and stirred at 0°C. The lithiate was added via cannula to the first flask. The reaction mixture was allowed to warm to room temperature and was stirred for 3 hours. The reaction mixture was poured onto saturated aqueous ammonium chloride (50 mL) and the layers were separated. The aqueous layer was extracted with EtOAc (3 x 50 mL). The combined organic extracts were dried over anhydrous sodium sulfate and chromatographed on silica using EtOAc/Hexanes 0 to 100% gradient to give Preparation I as a white foam in 83% yield, m/z (M+H)+ = 433.3. XH-NMR (400 MHz; CDC13): δ 8.80 (d, J = 4.5 Hz, 1H), 8.11 (dd, J = 9.1, 5.7 Hz, 1H), 7.63 (dd, J = 10.5, 2.5 Hz, 1H), 7.48-7.43 (m, 1H), 7.40-7.30 (m, 6H), 5.47-5.44 (m, 1H), 4.71 (t, J = 8.9 Hz, 1H), 4.31- 4.28 (m, 1H), 3.20-3.11 (m, 3H), 2.49-2.46 (m, 1H), 1.82-1.67 (m, 6H).
Preparation J:
Figure imgf000046_0001
[00173] A solution of Preparation I (21.6 g, 50 mmol) in anhydrous THF (200 mL) was cooled to -40 °C (using acetonitrile/dry ice bath, some precipitation occurs) and 2 M NaHMDS solution in THF (30 mL, 60 mmol) was added over 5 min (a 5-8 °C rise in temperature was observed). The resulting yellow reaction mixture was stirred for 10 min, became homogeneous, and Mel (10.6 g, 75 mmol) was added dropwise over 2 min (a 10 °C rise in temperature was observed). The reaction mixture was stirred for 1 h at -40 °C and LC/MS indicated the complete consumption of the starting material and formation of the desired methyl imide. The reaction mixture was rapidly diluted with saturated aqueous ammonium chloride solution (400 mL) and the biphasic mixture was stirred for 15 min. 'PrOAc (100 mL) was added, the layers were separated, and the aqueous layer was extracted with 'PrOAc (3 x 50 mL). The combined organic extracts were dried over anhydrous magnesium sulfate filtered, and concentrated. The resulting residue was recrystallized by dissolving in 400 mL hot acetone and adding H20 until a milky solution formed followed to re-dissolving with heating (-3: 1 acetone/H20). Preparation J was obtained as white needles (15.04 g, 2 crops, 68 %). m/z (M+H)+ = 447.3. XH-NMR (400 MHz; CDC13): δ 8.81 (d, J = 4.6 Hz, 1H), 8.10 (dd, J= 9.2, 5.7 Hz, 1H), 7.65 (dd, J= 10.6, 2.7 Hz, 1H), 7.47-7.42 (m, 1H), 7.41-7.29 (m, 6H), 5.47 (dd, J = 8.8, 3.8 Hz, 1H), 4.69 (t, J = 8.9 Hz, 1H), 4.38-4.30 (m, 1H), 4.26 (dd, J= 8.9, 3.9 Hz, 1H), 3.26-3.21 (m, 1H), 2.18-2.15 (m, 1H), 1.93-1.64 (m, 8H), 1.09 (d, J= 6.9 Hz, 3H).
Preparation K:
Figure imgf000046_0002
[00174] To a solution of Preparation J (82.0 g, 183.6 mmol) in THF (610 mL) at 0 °C was added aqueous H202 (35 wt%, 82 mL) and LiOH (7.04 g, 293.8 mmol) in H20 (189 mL). The resulting reaction mixture was allowed to slowly warm to rt and stirred overnight. The reaction was cooled to 0 °C and saturated aqueous sodium bisulfite solution (250 mL) was added. After stirring for 30 min, the THF was removed under reduced pressure. Acetic acid (34 mL) was added followed by EtOAc (300 mL). The layers were separated, and the aqueous layer was extracted with EtOAc (3 x 500 mL). The combined organic extracts were dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. The brown crude reaction mixture was suspended in MeCN (400 mL) and the suspension was brought to reflux with vigorous stirring. After cooling to rt, the solids were collected by filtration washing with additional MeCN. Preparation K was obtained as a white solid (45.4 g, 82 %). m/z (M+H)+ = 302.2. lH- NMR (400 MHz; DMSO-d6): δ 12.10 (s, 1H), 8.79 (d, J = 4.5 Hz, 1H), 8.07 (dd, J= 9.2, 5.9 Hz, 1H), 7.97-7.94 (m, 1H), 7.67-7.62 (m, 1H), 7.49 (d, J= 4.5 Hz, 1H), 3.41-3.36 (m, 1H), 2.73-2.65 (m, 1H), 1.83-1.61 (m, 9H), 1.08 (d, J= 6.8 Hz, 3H). Chiral HPLC, >99 % ee
(ChiralPak IC-3, 3μΜ, 4.6 x 250 mm, 15 min isocratic 70 % heptane 30% z-PrOH with 230 nm detection) at a flow rate of 0.75 mL/min the desired enantiomer had a retention time of 8.6 min with the undesired enantiomer eluting at 9.5 min.
Example 1
N-(4-Chloro-2-methylbenzyl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide
Figure imgf000047_0001
1A. Methyl 2-(4-methylpiperidin-4-yl)acetate
[00175] To a flask charged with MeOH (7.5 mL), at 0°C under nitrogen atmosphere, was slowly added acetyl chloride (1.1 mL, 15.2 mmol). After the addition was complete, the mixture was stirred at 0°C for 5 minutes before a homogeneous mixture of 2-(4-methylpiperidin- 4-yl)acetic acid, HC1 (675.0 mg, 3.5 mmol) in MeOH (1.5 mL) was added slowly dropwise. The resultant homogeneous mixture was stirred at 0°C for 5 minutes then at 60 °C for 8 hours, before being concentrated in vacuo to afford the HC1 salt of the title compound as a white solid (718.0 mg; 99% yield) which was used without further purification. MS (ES): m/z = 172
[M+H]+. tR = 0.46 min (Method BY lH NMR (400MHz, DMSO-d6) δ 9.41 - 9.12 (m, 1H), 3.60 (s, 3H), 3.25 - 3.15 (m, 2H), 2.93 - 2.82 (m, 2H), 2.39 - 2.30 (m, 2H), 1.74 - 1.64 (m, 4H), 1.02 (s, 3H).
IB. Methyl 2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)acetate
[00176] To a homogeneous mixture of 4-chloro-6-fluoroquinoline (350.0 mg, 1.9 mmol) in anhydrous NMP (5 mL), in a sealable vial, was added the HCl salt of methyl 2-(4- methylpiperidin-4-yl)acetate (1A, 480.0 mg, 2.3 mmol) followed by DIPEA (1.6 mL, 9.2 mmol). The vial was sealed and and the mixture was stirred at 120 °C. After 26 hours, the reaction mixture was cooled to room temperature then partitioned between water and EtOAc. The layers were separated and the aqueous layer was extracted once more with EtOAc. The organic layers were combined, washed with brine, then concentrated in vacuo to afford the crude product. Purification by Isco chromatography afforded methyl 2-(l-(6-fluoroquinolin-4-yl)-4- methylpiperidin-4-yl)acetate as an oil (565.8 mg; 93% yield). MS (ES): m/z = 317 [M+H]+. tR = 0.66 min (Method B). lH NMR (400MHz, DMSO-d6) δ 8.38 (d, J=5.4 Hz, 1H), 7.96 (dd, J=11.7, 2.8 Hz, 1H), 7.89 - 7.84 (m, 1H), 7.55 - 7.49 (m, 1H), 6.54 (d, J=5.5 Hz, 1H), 3.82 - 3.63 (m, 2H), 3.59 (s, 3H), 3.54 - 3.34 (m, 2H), 2.45 - 2.38 (m, 2H), 1.87 - 1.72 (m, 4H), 1.05 (s, 3H).
IC. 2-(l-(6-Fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)acetic acid
[00177] To a homogeneous mixture of methyl 2-(l-(6-fluoroquinolin-4-yl)-4- methylpiperidin-4-yl)acetate (321.0 mg, 1.0 mmol) in MeOH (5 mL), under nitrogen
atmosphere, was added dropwise 2M NaOH aqueous solution (1 mL, 2.0 mmol). The reaction was then stirred at ambient temperature for 20 hours before being treated with IN HCl (aq) until pH 5-6 to pH test strips. The mixture was then partitioned between water and EtOAc, the layers were separated and the aqueous layer was twice extracted with EtOAc. The aqueous layer from the extraction was lyophilized to afford the HCl salt of the crude product as an off-white solid (312.1 mg, 91% yield) which was used without further purification. MS (ES): m/z = 303
[M+H]+. tR = 0.59 min (Method B). XH NMR (400MHz, DMSO-d6) δ 12.12 (br.s, 1H), 8.41 (d, J=6.1 Hz, 1H), 8.12 (dd, J=11.5, 2.7 Hz, 1H), 8.04 - 7.94 (m, 1H), 7.75 - 7.64 (m, 1H), 6.64 (d, J=6.2 Hz, 1H), 3.98 - 3.87 (m, 1H), 3.87 - 3.78 (m, 1H), 3.69 - 3.49 (m, 2H), 2.38 - 2.29 (m, 2H), 1.92 - 1.70 (m, 4H), 1.06 (s, 3H).
Example 1. N-(4-Chloro-2-methylbenzyl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide [00178] To a mixture of the HC1 salt of 2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin- 4-yl)acetic acid (1C, 25.6 mg, 0.09 mmol) and 4-chloro-2-methylbenzylamine (15.8 mg, 0.1 mmol) in anhydrous DMF (1 mL), under nitrogen atmosphere, was added DIPEA (0.05 mL, 0.3 mmol) followed by PyBOP (44.1 mg, 0.09 mmol). The resulting mixture was stirred at ambient temperature for 156 hours, before being diluted with DMF, filtered through a syringe filter, then purified via preparative HPLC/MS to afford the title compound (22.2 mg; 60% yield). MS (ES): m/z = 440 [M+H]+. U = 1.52 min (Method A). XH NMR (500MHz, DMSO-d6) δ 8.37 - 8.30 (m, 2H), 7.99 - 7.92 (m, 1H), 7.85 (dd, J=9.1, 6.1 Hz, 1H), 7.58 - 7.49 (m, 1H), 7.24 - 7.13 (m, 3H), 6.51 (d, J=5.6 Hz, 1H), 4.17 (d, J=5.5 Hz, 2H), 3.80 - 3.60 (m, 2H), 3.54 - 3.32 (m, 2H), 2.26 - 2.19 (m, 5H), 1.84 - 1.67 (m, 4H), 1.02 (s, 3H).
Examples 2-5
Figure imgf000049_0001
[00179] Examples 2-5 were prepared using 2-(l-(6-fluoroquinolin-4-yl)-4- methylpiperidin-4-yl)acetic acid (1C) and corresponding amines following the procedure described for the synthesis of Example 1.
Figure imgf000049_0002
methylpiperidin-4-yl)acetamide
N-(4-chlorobenzyl)-2-(l-(6-
5
fluoroquinolin-4-yl)-4- 1.44 A 426.2 methylpiperidin-4-yl)acetamide
Example 6
(S)-2-(4-Methyl-l-(2-(trifluoromethyl)pyridin-4-yl)piperidin-4-yl)-N-(l- ph de
Figure imgf000050_0001
[00180] The title compound (28.0 mg) was prepared following a procedure analogous to that for the synthesis of Example 1 except that 4-chloro-2-(trifluoromethyl)pyridine (300.0 mg, 1.7 mmol) was used instead of 4-chloro-6-fluoroquinoline and (S)-l-phenylpropan-l-amine (14 mg, 0.104 mmol) was used instead of 4-chloro-2-methylbenzylamine. MS (ES): m/z = 420 [M+H]+. tR = 1.36 min (Method A). XH NMR (500MHz, DMSO-d6) δ 8.28 (d, J=8.2 Hz, 1H), 8.17 (t, J=6.3 Hz, 1H), 7.33 - 7.25 (m, 4H), 7.22 - 7.17 (m, 1H), 6.76 (s, 1H), 6.65 - 6.48 (m, 1H), 4.71 - 4.61 (m, 1H), 3.50 - 3.34 (m, 2H), 3.14 - 3.01 (m, 2H), 2.30 - 2.1 1 (m, 2H), 1.84 - 1.69 (m, 2H), 1.69 - 1.59 (m, 4H), 1.00 (s, 3H), 0.84 - 0.78 (m, 3H).
Examples 7-9
Figure imgf000051_0001
[00181] Examples 7-9 were prepared using 2-(4-methyl-l-(2-(trifluoromethyl)pyridin- 4-yl)piperidin-4-yl)acetic acid and corresponding amines following the procedure described for the synthesis of Example 6.
Figure imgf000051_0003
Example 10
(i?)-A^-(2-chlorobenzyl)-2-((cis)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propan amide
Figure imgf000051_0002
Example 10: (i?)-A^-(2-chlorobenzyl)-2-((cis)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide
[00182] Preparation K (see above) (20 mg, 0.066 mmol) was dissolved in thionyl chloride (48.4 μΐ, 0.664 mmol) and DMF (2.57 μΐ, 0.033 mmol) was added. The reaction was stirred at room temperature. After 1 hour, the reaction was concentrated in vacuo, taken up in toluene, concentrated again and placed on high vacuum to remove excess thionyl chloride. After 15 minutes, the crude acyl chloride was taken up in ACN (664 μΐ) and added to a solution of (2- chlorophenyl)methanamine (18.79 mg, 0.133 mmol) in ACN (664 μΐ) and TEA (46.3 μΐ, 0.332 mmol) at 0 °C. After 16 hours, the reaction was concentrated in vacuo, taken up in DMF (2 mL) filtered, and purified via HPLC to give Example 10 (26.4, 0.062 mmol, 94% yield). LC-MS Anal. Calc'd for C25H26CIFN2O 424.17, found [M+H] 425.2 Tr = 0.78 min (Method B). lH NMR (500 MHz, DMSO-d6) δ: 8.82 (d, J=4.5 Hz, 1H), 8.48 (t, J=5.3 Hz, 1H), 8.08 (dd, J=9.0, 5.8 Hz, 1H), 7.96 (dd, J=10.9, 2.3 Hz, 1H), 7.61-7.69 (m, 1H), 7.52 (d, J=4.5 Hz, 1H), 7.41 (d, J=7.6 Hz, 1H), 7.20-7.36 (m, 3H), 4.27-4.42 (m, 2H), 3.36 (br. s., 1H), 2.77 (dd, J=10.8, 6.7 Hz, 1H), 1.80-1.98 (m, 3H), 1.50-1.80 (m, 6H), 1.06 (d, J=6.6 Hz, 3H)
Examples 11-18
Figure imgf000052_0001
[00183] Examples 11-18 were prepared from Preparation K utilizing the procedure used to make Example 10 and corresponding benzylamines.
Figure imgf000052_0002
Figure imgf000053_0001
yl)cyclohexyl)propanamide
[00184] Analytical HPLC/MS was performed using the following methods for Examples 19-24:
Method A: Waters Acquity SDS using the following method: Linear Gradient of 2% to 98% solvent B over 1.7 min; UV visualization at 220 nm; Column: BEH CI 8 2.1 mm x 50 mm; 1.7 um particle (Heated to Temp. 50 °C); Flow rate: 0.8 ml/min; Mobile phase A: 100% Water, 0.05% TFA; Mobile phase B: 100% Acetonitrile, 0.05% TFA.
Method B: Column: Waters Acquity UPLC BEH CI 8, 2.1 x 50 mm, 1.7-μιη particles; Mobile Phase A: 5 :95 acetonitrile:water with 0.1 % trifluoroacetic acid; Mobile Phase B: 95 :5 acetonitrile:water with 0.1% trifluoroacetic acid; Temperature: 50 °C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.00 mL/min; Detection: UV at 220 nm.
Method C: Column: Waters Acquity UPLC BEH C18, 2.1 x 50 mm, 1.7-μιη particles; Mobile Phase A: 5 :95 acetonitrile:water with 0.1 % trifluoroacetic acid; Mobile Phase B: 95 :5 acetonitrile:water with 0.1% trifluoroacetic acid; Temperature: 50 °C; Gradient: 0-100% B over 3 minutes, then a 0.75-minute hold at 100% B; Flow: 1.0 mL/min; Detection: UV at 220 nm.
Example 19-1
N-((3R,5R,7R)-Adamantan-l-yl)-2-(4-(6-fluoroquinolin-4-yl)piperazin-l-yl)pentanamide
(Enantiomer 1 not assigned)
Figure imgf000054_0001
and
Example 19-2
N-((3R,5R,7R)-Adamantan-l-yl)-2-(4-(6-fluoroquinolin-4-yl)piperazin-l-yl)pentanamide
(Enantiomer 2 not assigned)
Figure imgf000054_0002
19A. tert-Butyl 4-(6-fluoroquinolin-4-yl)piperazine-l-carboxylate
[00185] To a mixture of 4-chloro-6-fluoroquinoline (500.0 mg, 2.8 mmol) in anhydrous NMP (5 mL), in a sealable vial, was added 1-Boc-piperazine (750.0 mg, 4.0 mmol) followed by DIPEA (2.0 mL, 11.5 mmol). The vial was capped and the mixture was stirred at 120 °C for 15.5 hours. The reaction mixture was cooled to room temperature before being partitioned between water and EtjO. The layers were separated and the aqueous layer was extracted twice more with Et^O. These organic extracts were combined with the original organic layer and were washed with brine, dried (Na2SC>4), filtered and concentrated in vacuo to afford the crude product. Purification by Isco chromatography afforded tert-butyl 4-(6-fluoroquinolin-4- yl)piperazine-l-carboxylate as an oil (719.3 mg; 77% yield). MS (ES): m/z = 332 [M+H] . tR = 0.70 min (Method A). lH NMR (400MHz, DMSO-d6) δ 8.70 (d, J=4.9 Hz, 1H), 8.09 - 8.00 (m, 1H), 7.77 - 7.67 (m, 1H), 7.67 - 7.62 (m, 1H), 7.07 (d, J=4.9 Hz, 1H), 3.67 - 3.57 (m, 4H), 3.15 - 3.06 (m, 4H), 1.44 (s, 9H).
19B. 6-Fluoro-4-(piperazin- l-yl)quinoline
[00186] To a homogeneous mixture of tert-butyl 4-(6-fluoroquinolin-4-yl)piperazine-l- carboxylate (600.0 mg, 1.8 mmol) in dioxane (4 mL), under nitrogen atmosphere, was added 4M HCl in dioxane (10 mL, 40.0 mmol). The resultant mixture was stirred at ambient temperature for 2.5 hours, during which time a precipitate formed. The heterogeneous mixture was concentrated to approximately 1/2 of its original volume. Vacum filtration afforded the HCl salt of the title compound as an off-white solid (490.0 mg; 100% yield) which was used without further purification. MS (ES): m/z = 232 [M+H]+. tR = 0.38 min (Method A).
19C. (±)-Ethyl 2-(4-(6-fluoroquinolin-4-yl)piperazin- l-yl)pentanoate
[00187] To a heterogeneous mixture of the HCl salt of 6-fluoro-4-(piperazin-l- yl)quinoline (19B, 200.0 mg, 0.75 mmol) in anhydrous DMF (5 mL), in a sealable reaction vial, was added K2C03 (288.0 mg, 2.08 mmol) followed by ethyl 2-bromovalerate (234.0 mg, 1.12 mmol). The flask was then sealed and the mixture was stirred at 60 °C. After 16 hours, the reaction was cooled to room temperature then partitioned between water and EtOAc. The layers were separated and the aqueous layer was extracted once more with EtOAc. The organic layers were combined, washed with brine, dried (anhydrous Na2S04), filtered and concentrated in vacuo to afford the product as a yellow oil which was used in the next step without further purification. MS (ES): m/z = 360 [M+H]+. tR = 0.62 min (Method A).
19D. (±)-2-(4-(6-Fluoroquinolin-4-yl)piperazin-l-yl)pentanoic acid
[00188] To a homogeneous mixture of ethyl 2-(4-(6-fluoroquinolin-4-yl)piperazin-l- yl)pentanoate (19C, 0.75 mmol) in MeOH (4 mL), under nitrogen atmosphere, was added dropwise 2M NaOH (aq) (0.8 mL, 1.6 mmol). The resultant mixture was stirred at ambient temperature for 64 hours before 2M NaOH (aq) (0.8 mL, 1.6 mmol) was added. After 6 hours of stirring, 2M NaOH (aq) (0.8 mL, 1.6 mmol) was added and stirring was continued. After 115 hours, the reaction was treated with HCl (4N in dioxane) until pH 5 to pH test strips. The mixture was then partitioned between water and EtOAc. The layers were separated and the aqueous layer was lyophilized to afford a pale yellow solid. Purification by RP Preparative HPLC (YMC-ODS 5μ 250 x 30 column. Conditions: 30mL/min flow rate; 40 minute gradient from 30-100%B (Solvent A = 95:5 H20/MeCN with 0.05%TFA. Solvent B = 5:95 H20/MeCN with 0.05%TFA) afforded the TFA salt of 2-(4-(6-fluoroquinolin-4-yl)piperazin-l-yl)pentanoic acid as a pale yellow solid (160.0 mg; 58% yield). MS (ES): m/z = 332 [M+H]+. tR = 0.46 min
(Method A). lH NMR (400MHz, DMSO-d6) δ 8.77 (d, J=6.4 Hz, 1H), 8.10 (dd, J=10.1, 5.2 Hz,
1H), 7.93 - 7.86 (m, 2H), 7.28 (d, J=6.5 Hz, 1H), 3.84 - 3.73 (m, 4H), 3.72 - 3.51 (m, 1H), 3.38 -
2.99 (m, 4H), 1.86 - 1.68 (m, 2H), 1.46 - 1.33 (m, 2H), 0.94 (t, J=7.3 Hz, 3H).
19E. (±)-N-((3R,5R,7R)-Adamantan- l-yl)-2-(4-(6-fluoroquinolin-4-yl)piperazin- 1- yl)pentanamide
[00189] o a jfljxtm-g 0f (±)-2-(4-(6-fluoroquinolin-4-yl)piperazin-l-yl)pentanoic acid (19D, 32.0 mg, 0.10 mmol) in anhydrous DMF (1.5 mL), in a sealable vial, was added PyBOP (50.3 mg, 0.10 mmol) followed by DIPEA (0.06 mL, 0.34 mmol). The resulting mixture was stirred at ambient temperature for 10 minutes before (3s,5s,7s)-adamantan-l -amine (17.5 mg, 0.12 mmol) was added. The vial was sealed and the reaction was stirred at ambient temperature for 63 hours, before being diluted with DMF, passed through a syringe filter, then purified via preparative HPLC/MS to afford the title compound as a racemate (10.3 mg; 23% yield). MS (ES): m/z = 465 [M+H]+. tR = 1.36 min (Method B).
Example 19-1. N-((3R,5R,7R)-Adamantan-l-yl)-2-(4-(6-fluoroquinolin-4-yl)piperazin-l- yl)pentanamide (Enantiomer 1, absolute stereochemistry not assigned)
and
Example 19-2. N-((3R,5R,7R)-Adamantan-l-yl)-2-(4-(6-fluoroquinolin-4-yl)piperazin-l- yl)pentanamide (Enantiomer 2, absolute stereochemistry not assigned)
[00190] Racemic Compound 19E (10.3 mg) was purified by chiral SFC (80/20 C02/MeOH with 0.1% DEA mobile phase, Chiral AD 25 X 3 cm, 5μιη column, 85 ml/min, detector wavelength = 220 nm). Concentration of the appropriate (earlier eluting) fractions afforded Example 19-1 (4.7 mg) assigned as N-((3R,5R,7R)-adamantan-l-yl)-2-(4-(6- fluoroquinolin-4-yl)piperazin-l-yl)pentanamide (Enantiomer 1). MS (ES): m/z = 465 [M+H]+. Tr = 1.53 min (Method B). lH NMR (500MHz, DMSO-d6) δ 8.63 (d, J=4.9 Hz, 1H), 8.00 (dd, J=9.0, 5.6 Hz, 1H), 7.64 - 7.53 (m, 2H), 7.41 (s, 1H), 7.02 (d, J=4.9 Hz, 1H), 3.19 - 3.03 (m, 4H), 2.97 (dd, J=8.8, 5.2 Hz, 1H), 2.85 - 2.69 (m, 4H), 2.00 - 1.91 (m, 9H), 1.62 - 1.56 (m, 7H), 1.50 - 1.41 (m, 1H), 1.28 - 1.19 (m, 2H), 0.87 (t, J=7.3 Hz, 3H).
[00191] Concentration of the later eluting fractions afforded Example 19-2 (4.9 mg) assigned as N-((3R,5R,7R)-adamantan-l-yl)-2-(4-(6-fluoroquinolin-4-yl)piperazin-l- yl)pentanamide (Enantiomer 2). MS (ES): m/z = 465 [M+H]+. Tr = 1.53 min (Method B). l NMR (500MHz, DMSO-d6) δ 8.65 (d, J=4.7 Hz, 1H), 8.04 - 7.98 (m, 1H), 7.64 - 7.53 (m, 2H), 7.39 (s, 1H), 7.02 (d, J=4.6 Hz, 1H), 3.20 - 3.01 (m, 4H), 2.98 (dd, J=8.2, 5.2 Hz, 1H), 2.85 - 2.69 (m, 4H), 2.02 - 1.92 (m, 9H), 1.64 - 1.56 (m, 7H), 1.50 - 1.41 (m, 1H), 1.28 - 1.21 (m, 2H), 0.88 (t, J=7.2 Hz, 3H).
Example 20-1
N-(Adamantan-2-yl)-2-(l-((6-(trifluoromethyl)pyridin-2-yl)methyl)piperidin-4- yl)butanamide (Enantiomer 1, absolute stereochemistry not assigned)
Figure imgf000057_0001
Example 20-2
N-(Adamantan-2-yl)-2-(l-((6-(trifluoromethyl)pyridin-2-yl)methyl)piperidin-4- yl)butanamide (Enantiomer 2, absolute stereochemistry not assigned)
Figure imgf000057_0002
20A. tert-Butyl 4-(l-ethoxy-l-oxobutan-2-ylidene)piperidine-l-carboxylate
[00192] To a suspension of NaH (0.29 g, 7.18 mmol) in anhydrous THF (10 mL), under nitrogen atmosphere, was added triethyl 2-phosphonobutyrate (1.81 g, 7.18 mmol) over 5 minutes. The resultant mixture was stirred at ambient temperature for 20 minutes, during which time it became homogeneous. To this solution was added dropwise a homogeneous mixture of l -Boc-4-piperidone (1.10 g, 5.52 mmol) in anhydrous THF (2.5 mL). After stirring for 1.5 hours, the reaction was quenched with saturated NH4C1 (aq) solution before being thoroughly extracted with EtOAc. The organic fractions were combined, washed with brine, dried
(MgS04), filtered and concentrated in vacuo to afford tert-butyl 4-(l -ethoxy-l-oxobutan-2- ylidene)piperi dine- 1 -carboxy late a clear oil (1.64 g; 100% yield) which was used without further purification. XH NMR (400MHz, DMSO-d6) δ 4.13 (q, J=7.1 Hz, 2H), 3.39 - 3.35 (m, 2H), 3.35 - 3.31 (m, 2H), 2.45 - 2.39 (m, 2H), 2.31 - 2.22 (m, 4H), 1.40 (s, 9H), 1.21 (t, J=7.2 Hz, 3H), 0.93 (t, J=7.5 Hz, 3H).
20B. (±)-tert-Butyl 4-(l-ethoxy-l-oxobutan-2-yl)piperidine-l-carboxylate
[00193] To a flask charged with tert-butyl 4-(l -ethoxy-1 -oxobutan-2- ylidene)piperi dine- 1 -carboxy late (1.64 g, 5.52 mmol) and under nitrogen atmosphere, was added platinum(IV) oxide (0.07 g, 0.31 mmol) followed by careful addition of EtOH (5 mL). The nitrogen line was then replaced with a hydrogen balloon and the mixture was stirred at ambient temperature. After 15 hours, the reaction mixture was purged with nitrogen before being filtered through a pad of Celite. The pad was thoroughly rinsed with EtOAc before the combined filtrates were concentrated in vacuo to afford racemic tert-butyl 4-(l-ethoxy-l - oxobutan-2-yl)piperidine-l -carboxylate as a clear oil (1.65 g; 100% yield) which was used without further purification. XH NMR (400MHz, DMSO-d6) δ 4.08 (q, J=7.1 Hz, 2H), 3.98 - 3.83 (m, 2H), 2.78 - 2.52 (m, 2H), 2.13 - 2.03 (m, 1H), 1.69 - 1.62 (m, 1H), 1.52 - 1.43 (m, 2H), 1.38 (s, 9H), 1.28 - 1.13 (m, 5H), 1.10 - 0.98 (m, 2H), 0.80 (t, J=7.3 Hz, 3H).
20C. (±)-Ethyl 2-(piperidin-4-yl)butanoate
[00194] To a homogeneous mixture of tert-butyl 4-(l -ethoxy-1 -oxobutan-2- yl)piperi dine- 1 -carboxy late (1.65 g, 5.52 mmol) in anhydrous dioxane (5 mL), under nitrogen atmosphere, was added HC1 (4N in dioxane, 10 mL, 40.0 mmol). The mixture was stirred at ambient temperature for two hours before being concentrated in vacuo to remove volatiles. The resultant oil was treated with saturated aqueous NaHCCb solution until pH5 and then IN NaOH (aq) until pH 8, before the mixture was thoroughly extracted with EtOAc. The layers were separated and the aqueous layer was lyophilized to afford the HC1 salt of the title compound as a pale yellow solid (1.20g; 92% yield) which was used in the next step without further purification. MS (ES): m/z = 200 [M+H]+. tR = 0.57 min (Method A). XH NMR (500MHz, DMSO-d6) δ 4.07 (q, J=7.2 Hz, 2H), 3.01 - 2.78 (m, 2H), 2.48 - 2.29 (m, 3H), 2.07 - 1.98 (m, 1H), 1.60 - 1.35 (m, 5H), 1.18 (t, J=7.1 Hz, 3H), 1.1 1 - 0.88 (m, 2H), 0.80 (t, J=7.4 Hz, 3H).
20D. (±)-Ethyl 2-(l-((6-(trifluoromethyl)pyridin-2-yl)methyl)piperidin-4-yl)butanoate [00195] To a homogeneous mixture of ethyl 2-(piperidin-4-yl)butanoate, HCl (20C, 187.0 mg, 0.8 mmol) in anhydrous DMF (3 mL) was added K2C03 (351.0 mg, 2.5 mmol). The mixture was stirred at ambient temperature for five minutes before 2-(chloromethyl)-6- (trifluoromethyl)pyridine (164.0 mg, 0.8 mmol) was added, the vial was sealed and the mixture stirred at 40 °C. After stirring 63 hours, the mixture was allowed to cool to room temperature, before being partitioned between water and EtOAc. The layers were separated and the aqueous layer was extracted twice more with EtOAc. The organic layers were combined, washed with brine, dried (Na2S04), filtered and concentrated in vacuo to afford the title compound as a gold oil (265.4 mg; 83% yield) which was used without further purification. MS (ES): m/z = 359 [M+H]+. tR = 0.72 min (Method A). lH NMR (400MHz, DMSO-d6) d 8.05 (t, J=7.8 Hz, 1H), 7.79 - 7.70 (m, 2H), 4.14 - 4.03 (m, 2H), 3.63 (s, 2H), 2.86 - 2.74 (m, 2H), 2.12 - 1.93 (m, 3H), 1.71 - 1.63 (m, 1H), 1.59 - 1.40 (m, 4H), 1.29 - 1.14 (m, 5H), 0.80 (t, J=7.4 Hz, 3H).
20E. (±)-2-(l-((6-(Trifluoromethyl)pyridin-2-yl)methyl)piperidin-4-yl)butanoic acid
[00196] To a homogeneous mixture of ethyl 2-(l-((6-(trifluoromethyl)pyridin-2- yl)methyl)piperidin-4-yl)butanoate (265.4 mg, 0.7 mmol) in EtOH (3 mL), under nitrogen atmosphere, was added NaOH (8M aqueous solution, 0.75 mL, 6.0 mmol). The mixture was stirred at 40°C for 48 hours and then at room temperature for another 69 hours before the reaction was quenched with 4N HCl in dioxane (2.0 mL, 8.0 mmol), stirred for 10 minutes at room temperature then concentrated in vacuo to afford the HCl salt of the crude product as a tan solid, which was used in the next step without further purification. MS (ES): m/z = 331 [M+H]+. tR = 0.60 min (Method A).
20F. (±)-N-(Adamantan-2-yl)-2-(l-((6-(trifluoromethyl)pyridin-2-yl)methyl)piperidin-4- yl)butanamide
[00197] To a mixture of the HCl salt of (±)-2-(l-((6-(trifluoromethyl)pyridin-2- yl)methyl)piperidin-4-yl)butanoic acid (20E, 30.0 mg, 0.07 mmol) in anhydrous DMF was added PyBOP (57.6 mg, 0.1 mmol) followed by DIPEA (0.1 mL, 0.6 mmol). The resulting mixture was stirred at ambient temperature for 10 minutes before 2-adamantanamine hydrochloride (18.0 mg, 0.1 mmol) was added. The vial was sealed and the reaction was stirred at ambient temperature for 24 hours, before being diluted with DMF, passed through a syringe filter, then purified via preparative HPLC/MS to afford the title compound as a racemate (17.0 mg; 48% yield). MS (ES): m/z = 464 [M+H]+. tR = 1.76 min (Method B). Example 20-1. N-(Adamantan-2-yl)-2-(l-((6-(trifluoromethyl)pyridin-2- yl)methyl)piperidin-4-yl)butanamide (Enantiomer 1, absolute stereochemistry not assigned)
and
Example 20-2. N-(Adamantan-2-yl)-2-(l-((6-(trifluoromethyl)pyridin-2- yl)methyl)piperidin-4-yl)butanamide (Enantiomer 2, absolute stereochemistry not assigned)
[00198] Racemic Compound 20F (14.8 mg) was purified by chiral SFC (81/19 C02/MeOH with 0.1 % DEA mobile phase, Chiral AD 25 X 3 cm, 5μιη column, 85 ml/min, detector wavelength = 220 nm). Concentration of the appropriate (earlier eluting) fractions afforded Example 20-1 (7.2 mg) assigned as N-(adamantan-2-yl)-2-(l -((6- (trifluoromethyl)pyridin-2-yl)methyl)piperidin-4-yl)butanamide (Enantiomer 1). MS (ES): m/z = 464 [M+H]+. Tr = 1.78 min (Method B). XH NMR (500MHz, DMSO-d6) δ 8.05 (t, J=7.7 Hz, 1H), 7.76 (d, J=7.6 Hz, 1H), 7.72 (d, J=7.8 Hz, 1H), 7.65 (d, J=7.5 Hz, 1H), 3.91 - 3.40 (m, 2H), 2.91 - 2.67 (m, 2H), 2.11 - 1.88 (m, 5H), 1.81 - 1.64 (m, 11H), 1.48 - 1.10 (m, 8H), 0.75 (t, J=7.2 Hz, 3H).
[00199] Concentration of the later eluting fractions afforded Example 20-2 (5.8 mg) assigned as N-(adamantan-2-yl)-2-(l-((6-(trifluoromethyl)pyridin-2-yl)methyl)piperidin-4- yl)butanamide (Enantiomer 2). MS (ES): m/z = 464 [M+H]+. Tr = 1.74 min (Method B). l NMR (500MHz, DMSO-d6) δ 8.05 (t, J=7.8 Hz, 1H), 7.76 (d, J=7.6 Hz, 1H), 7.72 (d, J=7.6 Hz, 1H), 7.62 (d, J=7.6 Hz, 1H), 3.91 - 3.57 (m, 2H), 2.88 - 2.70 (m, 2H), 2.13 - 1.89 (m, 5H), 1.82 - 1.65 (m, 1 1H), 1.50 - 1.15 (m, 8H), 0.76 (t, J=7.2 Hz, 3H).
Example 21
(cis)-(2R)-N-(3,5-dimethyladamantan-l-yl)-2-(4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide
Figure imgf000060_0001
[00200] A solution of (cis)-(R)-2-(4-(6-fluoroquinolin-4-yl)cyclohexyl)propanoic acid (Preparation K, 0.030 g, 0.1 mmol) and 3,5-dimethyladamantan-l-amine, HCl (0.024 g, 0.110 mmol) in DMF (0.4 mL) was treated with triethylamine (0.049 mL, 0.350 mmol) followed by HATU (0.046 g, 0.120 mmol). The resulting solution was stirred lh at RT then quenched with 1 drop of water and purified by prep. HPLC. Concentration of the appropriate fractions afforded (cis)-(2R)-N-(3,5-dimethyladamantan-l-yl)-2-(4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide (0.042 g, 91 % yield). MS (ES): m/z = 463 [M + H]+. tR = 0.97 min (Method A). XH NMR (400MHz, DMSO-de) δ 8.85 (d, 1H, J = 4.4 Hz), 8.09 (dd, 1H, J = 9.0, 5.8 Hz), 7.96 (br. d, 1H, J = 9.1 Hz), 7.63-7.69 (m, 1H), 7.56 (d, 1H, J = 4.2 Hz), 7.42 (s, 1H), 3.30-3.37 (m, 1H), 2.58-2.65 (m, 1H), 2.05 (s, 1H), 1.87-1.96 (m, 1H), 1.52-1.83 (m, 14H), 1.26 (ABq, 4H, JAB = 11.8 Hz, Δν = 37.3 Hz), 1.08 (s, 2H), 0.96 (d, 3H, J = 6.5 Hz), 0.81 (s, 6H).
Example 22
N-((3s,5s,7s)-Adamantan-l-yl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4-
Figure imgf000061_0001
22A. 2-(l-(6-Fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)acetic acid
[00201] To a mixture of 4-chloro-6-fluoroquinoline (200.0 mg, 1.1 mmol) in anhydrous NMP (4 mL), in a sealable vial, was added 2-(4-methylpiperidin-4-yl)acetic acid, HCl (320.0 mg, 1.7 mmol) followed by DIPEA (0.8 mL, 4.6 mmol). The vial was sealed and the mixture was stirred at ambient temperature for 2 hours, then at 120 °C for 65 hours. After cooling to room temperature, the reaction mixture was partitioned between water and EtOAc. The layers were separated and the aqueous layer was extracted twice more with EtOAc. The product was determined to be mostly in aqueous layer, which was subsequently acidified with 6M HCl (aq) (0.76 mL, 4.6 mmol) then lyophilized to afford the crude product as an oil. Purification by Isco chromatography followed by preparative HPLC afforded the HCl salt of the title compound as a clear residue (23.3mg; 7% yield). MS (ES): m/z = 303 [M+H]+. tR = 0.58 min (Method A). XH NMR (400MHz, DMSO-d6) δ 13.84 (br. s., 1H), 8.48 (d, J=7.1 Hz, 1H), 8.32 (d, J=10.4 Hz, 1H), 8.03 - 7.85 (m, 2H), 6.77 (d, J=7.2 Hz, 1H), -3.60 (m, integration, exact chemical shift range obscured by water peak), 2.40 - 2.29 (m, 2H), 2.00 - 1.82 (m, 2H), 1.80 - 1.66 (m, 2H), 1.08 (s, 3H).
Example 22. N-((3s,5s,7s)-Adamantan-l-yl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin- 4-yl)acetamide
[00202] To a mixture of 2-(l -(6-fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)acetic acid (22A, 21.0 mg, 0.07 mmol) and (3s,5s,7s)-adamantan-l -amine, HC1 (15.7 mg, 0.08 mmol) in anhydrous DMF (1 mL), under nitrogen atmosphere, was added DIPEA (0.04 mL, 0.2 mmol) followed by PyBOP (36.1 mg, 0.07 mmol). After stirring at ambient temperature for 7.5 days, the mixture was diluted with DMF, filtered through a syringe filter, then purified via preparative HPLC/MS to afford the title compound (17.9 mg; 59% yield). MS (ES): m/z = 436 [M+H]+. tR = 1.67 min (Method B). XH NMR (500MHz, DMSO-d6) δ 8.34 (d, J=5.5 Hz, 1H), 7.94 (d, J=9.4 Hz, 1H), 7.86 (dd, J=9.1, 6.1 Hz, 1H), 7.58 - 7.49 (m, 1H), 7.37 (s, 1H), 6.53 (d, J=5.5 Hz, 1H), 3.66 - 3.32 (m, 2H), 2.54 (s, 2H), 2.09 (t, J=7.7 Hz, 2H), 1.98 - 1.93 (m, 3H), 1.90 - 1.85 (m, 7H), 1.84 - 1.71 (m, 2H), 1.64 (t, J=7.0 Hz, 2H), 1.59 - 1.55 (m, 5H), 1.02 (s, 3H).
Example 23
2-(l-(6-Fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)-N-((lr,3s,5R,7S)-3- hydroxyadamantan-l-yl)acetamide
Figure imgf000062_0001
[00203] To a mixture of 2-(l -(6-fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)acetic acid (22A, 26.5 mg, 0.09 mmol) in anhydrous DMF (1 mL) was added PyBOP (45.6 mg, 0.09 mmol) followed by DIPEA (0.06 mL, 0.3 mmol). The resulting mixture was stirred for 15 minutes before (ls,3r,5R,7S)-3-aminoadamantan-l-ol (17.6 mg, 0.1 mmol) was added. After stirring at ambient temperature for 21 hours, the mixture was diluted with DMF, filtered through a syringe filter, then purified via preparative HPLC/MS to afford the title compound (13.4 mg; 27% yield). MS (ES): m/z = 452 [M+H]+. tR = 1.18 min (Method B). XH NMR (500MHz, DMSO-d6) δ 8.35 (d, J=5.4 Hz, 1H), 8.00 - 7.92 (m, 1H), 7.86 (dd, J=9.0, 6.1 Hz, 1H), 7.57 - 7.49 (m, 1H), 7.43 (s, 1H), 6.53 (d, J=5.4 Hz, 1H), 3.82 - 3.47 (m, 2H), 2.54 (s, 2H), 2.10 - 2.08 (m, 2H), 1.92 - 1.88 (m, 3H), 1.83 - 1.71 (m, 8H), 1.65 (t, J=7.7 Hz, 2H), 1.53 - 1.36 (m, 6H), 1.02 (s, 3H).
Example 24
N-((3s,5s,7s)-Adamantan-l-yl)-2-(4-methyl-l-(2-(trifluoromethyl)pyridin-4-yl)piperidin-4-
Figure imgf000063_0001
24A. Methyl 2-(4-methylpiperidin-4-yl)acetate
[00204] To a flask charged with MeOH (7.5 mL), at 0°C under nitrogen atmosphere, was slowly added acetyl chloride (1.1 mL, 15.2 mmol). After the addition was complete, the mixture was stirred at 0°C for 5 minutes before a homogeneous mixture of 2-(4-methylpiperidin- 4-yl)acetic acid, HCl (675.0 mg, 3.5 mmol) in MeOH (1.5 mL) was added slowly dropwise. The resultant homogeneous mixture was stirred at 0°C for 5 minutes then at 60 °C for 8 hours, before being concentrated in vacuo to afford the HCl salt of the title compound as a white solid (718.0 mg; 99% yield) which was used without further purification. MS (ES): m/z = 172
[M+H]+. tR = 0.46 min (Method A). XH NMR (400MHz, DMSO-d6) δ 9.41 - 9.12 (m, 1H), 3.60 (s, 3H), 3.25 - 3.15 (m, 2H), 2.93 - 2.82 (m, 2H), 2.39 - 2.30 (m, 2H), 1.74 - 1.64 (m, 4H), 1.02 (s, 3H).
24B. Methyl 2-(4-methyl- l-(2-(trifluoromethyl)pyridin-4-yl)piperidin-4-yl)acetate
[00205] To a homogeneous mixture of 4-chloro-2-(trifluoromethyl)pyridine (300.0 mg,
1.7 mmol) in anhydrous NMP (4 mL), in a sealable vial, was added the HCl salt of methyl 2-(4- methylpiperidin-4-yl)acetate (24A, 412.0 mg, 2.0 mmol) followed by DIPEA (1.3 mL, 7.4 mmol). The vial was sealed and and the mixture was stirred at 120 °C. After 25 hours, the reaction mixture was cooled to room temperature then partitioned between water and EtOAc.
The layers were separated and the aqueous layer was extracted once more with EtOAc. The organic layers were combined, washed with brine, then concentrated in vacuo to afford the crude product. Purification by Isco chromatography afforded methyl 2-(4-methyl-l-(2- (trifluoromethyl)pyridin-4-yl)piperidin-4-yl)acetate as an oil (461.1 mg; 88% yield). MS (ES): m/z = 317 [M+H]+. tR = 0.65 min (Method A). lH NMR (400MHz, CHLOROFORM-d) δ 8.27 (d, J=5.9 Hz, 1H), 6.70 (d, J=2.4 Hz, 1H), 6.43 (dd, J=5.9, 2.4 Hz, 1H), 3.69 (s, 3H), 3.48 - 3.41 (m, 2H), 3.20 - 3.08 (m, 2H), 2.44 - 2.31 (m, 2H), 1.86 - 1.82 (m, 2H), 1.58 (s, 2H), 1.11 (s, 3H). 24C. 2-(4-Methyl- l-(2-(trifluoromethyl)pyridin-4-yl)piperidin-4-yl)acetic acid
[00206] To a homogeneous mixture of methyl 2-(4-methyl-l-(2- (trifluoromethyl)pyridin-4-yl)piperidin-4-yl)acetate (461.1 mg, 1.5 mmol) in MeOH (5 mL), under nitrogen atmosphere, was added dropwise 2M NaOH aqueous solution (1.6 mL, 3.2 mmol). The reaction was then stirred at ambient temperature for 13 hours before being treated with IN HCl (aq) until pH 3-4 to pH test strips. The mixture was then partitioned between water and EtOAc, the layers were separated and the aqueous layer was twice extracted with EtOAc. The organic layer from the extraction was concentrated in vacuo to afford the HCl salt of the crude product as a white solid (362.4 mg, 64% yield) which was used without further purification. MS (ES): m/z = 303 [M+H]+. tR = 0.56 min (Method A). XH NMR (400MHz, DMSO-de) δ 12.03 (br. s, 1H), 8.19 (d, J=5.7 Hz, 1H), 6.80 (d, J=2.3 Hz, 1H), 6.63 (dd, J=5.9, 2.3 Hz, 1H), 3.43 - 3.37 (m, 2H), 3.18 - 3.09 (m, 2H), 2.35 - 2.23 (m, 2H), 1.87 - 1.72 (m, 2H), 1.71 - 1.63 (m, 2H), 1.02 (s, 3H).
Example 24. N-((3s,5s,7s)-Adamantan- l-yl)-2-(4-methyl- l-(2-(trifluoromethyl)pyridin-4- yl)piperidin-4-yl)acetamide
[00207] To a mixture of the HCl salt of 2-(4-methyl-l-(2-(trifluoromethyl)pyridin-4- yl)piperidin-4-yl)acetic acid (24C, 25.6 mg, 0.09 mmol) in anhydrous DMF (1 mL) was added PyBOP (44.1 mg, 0.09 mmol) followed by DIPEA (0.06 mL, 0.3 mmol)). The resulting mixture was stirred for 15 minutes before (3s,5s,7s)-adamantan-l -amine (16.0 mg, 0.1 mmol) was added. After stirring at ambient temperature for 15 hours, the mixture was diluted with DMF, filtered through a syringe filter, then purified via preparative HPLC/MS to afford the title compound (30.2 mg; 64% yield). MS (ES): m/z = 436 [M+H]+. tR = 1.71 min (Method B). lH NMR (500MHz, DMSO-de) δ 8.19 (d, J=5.9 Hz, 1H), 7.34 (s, 1H), 6.80 (s, 1H), 6.69 - 6.59 (m, 1H), 3.19 - 3.06 (m, 2H), 2.54 (s, 2H), 2.08 (t, J=7.7 Hz, 2H), 2.01 - 1.94 (m, 3H), 1.92 - 1.87 (m, 6H), 1.84 - 1.71 (m, 2H), 1.64 - 1.55 (m, 8H), 1.01 (s, 3H). Materials and Methods
[00208] The following general materials and methods were used, where indicated, or may be used in the Examples below:
[00209] Standard methods in molecular biology are described in the scientific literature (see, e.g., Sambrook et al, Molecular Cloning, Third Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY (2001); and Ausubel et al, Current Protocols in Molecular Biology, Vols. 1-4, John Wiley and Sons, Inc. New York, NY (2001), which describes cloning in bacterial cells and DNA mutagenesis (Vol. 1), cloning in mammalian cells and yeast (Vol. 2), glycoconjugates and protein expression (Vol. 3), and bioinformatics (Vol. 4)).
[00210] The scientific literature describes methods for protein purification, including immunoprecipitation, chromatography, electrophoresis, centrifugation, and crystallization, as well as chemical analysis, chemical modification, post-translational modification, production of fusion proteins, and glycosylation of proteins (see, e.g., Coligan et al, Current Protocols in Protein Science, Vols. 1-2, John Wiley and Sons, Inc., NY (2000)).
[00211] Software packages and databases for determining, e.g., antigenic fragments, leader sequences, protein folding, functional domains, glycosylation sites, and sequence alignments, are available (see, e.g., GCG® Wisconsin Package (Accelrys, Inc., San Diego, CA); and DECYPHER® (TimeLogic Corp., Crystal Bay, NV).
[00212] The literature is replete with assays and other experimental techniques that can serve as a basis for evaluation of the compounds described herein.
[00213] An IDO enzyme assay and cellular production of kynurenine (KYN) is described in Sarkar, S.A. et al, Diabetes, 56:72-79 (2007). Briefly, all chemicals can be purchased from Sigma- Aldrich (St. Louis, MO) unless specified otherwise. Groups of 1,000 human islets can be cultured for 24 h in 1 mL medium with cytokines, recovered by
centrifugation for 5 min at 800 x g and sonicated in 150 PBS containing a protease inhibitor cocktail (Set 2; Calbiochem, EMD Biosciences, San Diego, CA). The sonicate can be centrifuged for 10 min at 10,000 x g, and the supernatant can be assayed in triplicate by incubating a 40 μΐ sample with an equal volume of 100 mmol/L potassium phosphate buffer, pH 6.5, containing 40 mmol/L ascorbic acid (neutralized to pH 7.0), 100 μιηοΙ/L methylene blue, 200 μg/mL catalase, and 400 μιτιοι/1 L-Trp for 30 min at 37 °C. The assay can be terminated by the addition of 16 μΐ. 30% (w/v) trichloroacetic acid (TCA) and further incubated at 60 °C for 15 min to hydrolyze N-formylkynurenine to ΚΥΝ. The mixture can then be centrifuged at 12,000 rpm for 15 min, and ΚΥΝ can be quantified by mixing equal volume of supernatant with 2% (w/v) Ehrlich's reagent in glacial acetic acid in 96-well microtiter plate and reading the absorbance at 480 nm using L-KYN as standard. Protein in the islet samples can be quantified by Bio-Rad Protein assay at 595 nm. For the detection of L-KYN in the islet culture
supernatants, proteins can be precipitated with 5% (w/v) TCA and centrifuged at 12,000 rpm for 15 min, and determination of KYN in the supernatant with Ehrlich's reagent can be determined as described above. IL-4 (10 μg/mL; 500-2,000 units/mL) and 1-a-methyl Trp (1-MT; 40 μιηοΙ/L) can be added to the incubation media as indicated. This assay can also form the basis of a cell-based assay, and may be quantified via LCMS/MS as an alternative to UV/Vis detection.
[00214] Western Blot Analyses. Groups of 1,000-1,200 islets incubated for 24 h in Miami medium in the presence of cytokines can be harvested and sonicated in PBS as above, and 50 μg protein samples can be electrophoresed on 10% SDS-PAGE gels. COS7 cells (0.6 χ 106 cells/60 mm3 petri dish) transfected with human-IDO plasmid (3 μg) or empty vector cells can be used as positive and negative controls, respectively. Proteins can be transferred
electrophoretically onto polyvinylidine fluoride membranes by semidry method and blocked for 1 h with 5% (w/v) nonfat dry milk in Tris-buffered saline and 0.1% Tween and then incubated overnight with anti-human mouse IDO antibody (1 :500; Chemicon, Temecula, CA), phospho- STATic p91, and STATia p91 (1 :500; Zymed, San Francisco, CA). Immunoreactive proteins can be visualized with ECL PLUS® Westem blotting detection reagent (Amersham Biosciences, Buckinghamshire, U.K.) after incubation for 1 h with anti-mouse horseradish peroxidase- conjugated secondary antibody (Jackson Immunolabs, West Grove, PA).
[00215] Immunohistochemical Detection of IDO. Islets can be fixed in 4%
paraformaldehyde in PBS (Invitrogen) for 1 h, immobilized in molten 10% porcine skin gelatin blocks (37 °C), and embedded in optimal cutting temperature compound. Immunofluorescent staining on islet tissue can be performed on 7 μιτι sections that were stained with antibodies raised against pancreatic duodenal homeobox 1 (PDX1) and IDO. Antigen retrieval can be performed in a water bath for 30 min in a buffer containing 10 mmol/1 Tris and 1 mmol/1 EDTA (pH 9.0) at 97 °C. The sections can be blocked for 1 h with 5% normal goat serum in PBS. The tissues can then be reacted with mouse monoclonal anti-human IDO antibody (1 :20; Chemicon) and goat polyclonal anti-human PDX1 antibody (1 :2,000; which may be requested from Dr. Chris Wright, School of Medicine, Vanderbilt, TN) overnight at room temperature in a humid chamber. Secondary antibodies anti-goat (labeled with Cy3) and anti-mouse (labeled with Cy2) can be purchased from Jackson Immunolabs and can be used at a concentration of 1 :200. The nuclei can be stained with Hoechst 33258 (Molecular Probes, Eugene, OR). Images can be acquired by Intelligent Imaging System software from an Olympus 1X81 inverted motorized microscope equipped with Olympus DSU (spinning disk confocal) and Hamamatsu ORCA HER monochromatic CCD camera.
[00216] Alternative means for evaluating the IDO inhibitors of the present invention are described in WO 2010/0233166 and are summarized hereafter.
[00217] Biochemical Assay. cDNA clones for both human and mouse IDO have been isolated and verified by sequencing and are commercially available. In order to prepare IDO for biochemical studies, C-terminal His-tagged IDO protein can be produced in E. coli using the IPTG-inducible pET5a vector system and isolated over a nickel column. The yield of the partially purified protein can be verified by gel electrophoresis and the concentration estimated by comparison to protein standards. To assay IDO enzymatic activity, a 96-well plate spectrophotometric assay for kynurenine production can be run following published procedures (see, e.g., Littlejohn, T.K. et al., Prot. Exp. Purif, 19:22-29 (2000)). To screen for IDO inhibitory activity, compounds can be evaluated at a single concentration of, for example, 200 μΜ against 50 ng of IDO enzyme in 100 reaction volumes with tryptophan added at increasing concentrations at, for example, 0, 2, 20, and 200 μΜ. Kynurenine production can be measured at 1 hour.
[00218] Cell-based Assay. COS-1 cells can be transiently transfected with a CMV promoter-driven plasmid expressing IDO cDNA using Lipofectamine 2000 (Invitrogen) as recommended by the manufacturer. A companion set of cells can be transiently transfected with TDO-expressing plasmid. Forty-eight hours post-transfection, the cells can be apportioned into a 96-well format at 6 χ 104 cells per well. The following day, the wells can be washed and new media (phenol red free) containing 20μg/mL tryptophan can be added together with inhibitor. The reaction can be stopped at 5 hours and the supernatant removed and spectrophotometrically- assayed for kynurenine as previously described for the enzyme assay. To obtain initial confirmation of IDO activity, compounds can be evaluated at a single concentration of, for example, 100 μΜ. More extensive dose-escalation profiles can be collected for select compounds.
[00219] Pharmacodynamic and Pharmacokinetic Evaluation. A pharmacodynamic assay can be based on measuring serum levels of both kynurenine and tryptophan, and calculating the kynurenine/tryptophan ratio provides an estimate of IDO activity that is independent of baseline tryptophan levels. Serum tryptophan and kynurenine levels can be determined by HPLC analysis, and serum compound levels can optionally also be determined in the same HPLC run.
[00220] Compounds can be initially evaluated by challenging mice with LPS and then subsequently administering a bolus dose of compound at the time that the serum kynurenine level plateaus. As the kynurenine pool is rapidly turned over with a half-life in serum of less than 10 minutes, pre-existing kynurenine is not expected to unduly mask the impact that an IDO inhibitor has on kynurenine production. Each experiment can include non-LPS-exposed mice (to determine baseline kynurenine levels against which to compare the other mice) and a set of LPS- exposed mice dosed with vehicle alone (to provide a positive control for IDO activation). Each compound can initially be evaluated in mice at a single high i.p. bolus dose in the range of at least 100 mg/kg. Blood can be collected at defined time intervals (for example, 50 sample at 5, 15, 30 min., 1, 2, 4, 6, 8, and 24 hr. following compound administration) for HPLC analysis of kynurenine and tryptophan levels (pharmacodynamic analysis) as well as for the level of compound (pharmacokinetic analysis). From the pharmacokinetic data the peak serum concentration of compound achieved can be determined as well as the estimated rate of clearance. By comparing the level of compound in serum relative to the kynurenine/tryptophan ratio at various time points, the effective IC50 for IDO inhibition in vivo can be roughly estimated. Compounds exhibiting efficacy can be evaluated to determine a maximum dose that achieves 100% IDO inhibition at the peak concentration.
EVALUATION OF BIOLOGICAL ACTIVITY
[00221] Exemplary compounds were tested for inhibition of IDO activity.
Experimental procedures and results are provided below.
[00222] HEK293 cells were transfected with a pCDNA-based mammalian expression vector harboring human IDOl cDNA (NM 002164.2) by electroporation. They were cultured in medium (DMEM with 10% FBS) containing 1 mg/ml G418 for two weeks. Clones of HEK293 cells that stably expressed human IDOl protein were selected and expanded for IDO inhibition assay.
[00223] The human IDO 1/HEK293 cells were seeded at 10,000 cells per 50μί per well with RPMI/phenol red free media contains 10% FBS in a 384-well black wall clear bottom tissue culture plate (Matrix Technologies LLC) 100 nL of certain concentration of compound was then added to each well using ECHO liquid handling systems. The cells were incubated for 20 hours in 37 °C incubator with 5% C02. [00224] The compound treatments were stopped by adding trichloroacetic acid (Sigma- Aldrich) to a final concentration at 0.2%. The cell plate was further incubated at 50 °C for 30 minute. The equal volume supernatant (20μί) and 0.2% (w/v) Ehrlich reagent (4- dimethylaminobenzaldehyde, Sigma-Aldrich) in glacial acetic acid were mixed in a new clear bottom 384-well plate. This plate was then incubated at room temperature for 30 minute. The absorbance at 490 nm was measured on Envision plate reader.
[00225] Compound IC50 values were calculated using the counts of 500 nM of a reference standard treatment as one hundred percent inhibition, and counts of no compound but DMSO treatment as zero percent inhibition.
[00226] Assessment of inhibitor activity in HeLa cell-based indoleamine 2,3- di oxygenase (IDO) assay:
[00227] HeLa (ATCC® CCL-2) cells were obtained from the ATCC® and cultured in Dulbecco's Modified Eagle Medium supplemented with 4.5 g/L glucose, 4.5 g/L L-glutamine and 4.5 g/L sodium pyruvate (#10-013-CV, Corning), 2 mM L-alanyl-L-glutamine dipeptide (#35050-061, Gibco), lOOU/mL penicillin, 100μg/mL streptomycin (#SV30010, HyClone) and 10% fetal bovine serum (#SH30071.03 HyClone). Cells were maintained in a humidified incubator at 37 °C in 5% C02.
[00228] IDO activity was assessed as a function of kynurenine production as follows: HeLa cells were seeded in a 96-well culture plate at a density of 5,000 cells/well and allowed to equilibrate overnight. After 24 hours, the media was aspirated and replaced with media containing IFNy (#285-IF/CF, R&D Systems) at a final concentration of 25 ng/mL. A serial dilution of each test compound was added to the cells in a total volume of 200 of culture medium. After a further 48 hour incubation, 170 μΐ. of supernatant was transferred from each well to a fresh 96-well plate. 12.1 μΐ. of 6. IN trichloroacetic acid (#T0699, Sigma-Aldrich) was added to each well and mixed, followed by incubation at 65 °C for 20 minutes to hydrolyze N- formylkynurenine, the product of indoleamine 2,3-dioxygenase, to kynurenine. The reaction mixture was then centrifuged for 10 mins at 500xg to sediment the precipitate. 100 μΐ. of the supernatant was transferred from each well to a fresh 96-well plate. 100 μΐ of 2% (w/v) p- dimethylaminobenzaldehyde (#15647-7, Sigma-Aldrich) in acetic acid (#A6283, Sigma-Aldrich) was added to each well mixed and incubated at room temperature for 20 mins. Kynurenine concentrations were determined by measuring absorbance at 480nm and calibrating against an L- kynurenine (#K8625, Sigma-Aldrich) standard curve using a SPECTRAMAX® M2e microplate reader (Molecular Devices). The percentage activity at each inhibitor concentration was determined and IC50 values assessed using nonlinear regression.
[00229] Activity for compounds described herein is provided below, wherein potency levels are provided as follows: (Potency: IDO IC50: A < 0.1 μΜ; B < 1 μΜ; C < 10 μΜ).
[00230] Results of the IDO assays are shown in the table below.
HEK Human IDO- 1
Figure imgf000070_0001
ASPECTS
Aspect 1. A compound of formula I or formula II
Figure imgf000071_0001
wherein
X is CH or N;
T is CH or N;
R1 is H, halo, or Ci-Cehaloalkyl;
R1A is H, halo, or Ci-C6haloalkyl;
Y is CH or N;
n is 0, 1 , 2, 3, or 4;
W is N or CR4 wherein R4 is H or (Ci-C6alkyl);
V is Ci-Cealkylene optionally substituted with one, two, or three substituents
independently selected from Ci-Cealkyl and C3-Cecycloalkyl;
R2 is H or Ci-C6alkyl;
Z is Ci-Cealkylene optionally substituted with one, two, or three substituents
independently selected from Ci-C6alkyl and C3-C6cycloalkyl;
B is aryl optionally substituted with 1 , 2, or 3 R substituents independently selected from -OH, Ci-C6alkyl, Ci-C6haloalkyl, halo, -OCi-C6alkyl, -OCi-C6haloalkyl, -
CN, aryl, or -Oaryl; or
C3-Ci2cycloalkyl optionally substituted with 1 , 2, or 3 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi- Cealkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl;
or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
Aspect 2. The compound of Aspect 1 that is a compound of formula I.
Aspect 3. The compound of cl Aspect aim 1 that is a compound of formula II. Aspect 4. The compound of any one of the preceding Aspects, wherein R1 is H, F, or -CF3. Aspect 5. The compound of any one of the preceding Aspects, wherein R is H.
Aspect 6. The compound of any one of the preceding Aspects, wherein Y is CH.
Aspect 7. The compound of any one of Aspects 1 to 3, wherein Y is N.
Aspect 8. The compound of any one of the preceding Aspects, wherein n is 2.
Aspect 9. The compound of any one of the preceding Aspects, wherein W is CR4.
Aspect 10. The compound of any one of Aspects 1 to 8, wherein W is N.
Aspect 11. The compound of any one of the preceding Aspects, wherein V is Cialkylene optionally substituted with one substituent that is Ci-Cealkyl and C3-C6cycloalkyl.
Aspect 12. The compound of any one of the preceding Aspects, wherein Z is Cialkylene optionally substituted with one or two substituents independently selected from Ci- Cealkyl and C3-C6cycloalkyl.
Aspect 13. The compound of any one of the preceding claims, wherein B is aryl optionally substituted with 1, 2, or 3 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi-C6alkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl.
Aspect 14. The compound of Aspects 13, wherein B is phenyl optionally substituted with 1 or 2 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, - OCi-Cealkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl.
Aspect 15. The compound of any one of Aspects 1 to 12, wherein B is C3-Ci2cycloalkyl optionally substituted with 1 , 2, or 3 R substituents independently selected from -OH, Ci- Cealkyl, Ci-C6haloalkyl, halo, -OCi-C6alkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl.
Aspect 16. The compound of Aspect 1 that is
N-(4-Chloro-2-methylbenzyl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide;
N-(l -(2,4-dichlorophenyl)cyclo-propyl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide;
-(2-(4-chlorophenyl)propan-2-yl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide; (S)-N-(l-(4-chlorophenyl)ethyl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide;
N-(4-chlorobenzyl)-2-(l -(6-fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)acetamide;
(S)-2-(4-Methyl-l-(2-(trifluoromethyl)pyridin-4-yl)piperidin-4-yl)-N-(l- phenylpropyl)acetamide;
N-(l -(2,4-dichlorophenyl)cy clo-propyl)-2-(4-methyl- 1 -(2-(trifluoro-methyl)pyridin-4- yl)piperidin-4-yl)acetamide;
N-(2-(4-chlorophenyl)propan-2-yl)-2-(4-methyl-l-(2-(trifluoromethyl)-pyridin-4- yl)piperidin-4-yl)acetamide;
(S)-N-(l -(4-chlorophenyl)ethyl)-2-(4-methyl-l -(2-(trifluoromethyl)-pyridin-4-yl)piperi yl)acetamide;
(i?)-N-(2-chlorobenzyl)-2-((cz ,)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide;
(i?)-N-(2-chlorobenzyl)-2-((cz ,)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide;
(R)-N-([\ , 1 '-bipheny 1] -4-y lmethy l)-2-((cw)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(i?)-2-((cw)-4-(6-fluoroquinolin-4-yl)cyclohexyl)-N-((^-l-phenylethyl)propanamide;
(i?)-N-((5 -l-(4-chlorophenyl)ethyl)-2-((cw)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(i?)-N-((5 -l-(4-chlorophenyl)ethyl)-2-((cw)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(i?)-N-(l-(2,4-dichlorophenyl)cyclopropyl)-2-((cw)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(i?)-N-((i?)-l -(4-chlorophenyl)ethyl)-2-((l s,4S)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(R)-N-benzyl-2-((l s,4S)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide;
N-((3R,5R R)-Adamantan-l-yl)-2-(4-(6-fluoroquinolin-4-yl)piperazin-l -yl)pentanamide; N-(Adamantan-2-yl)-2-(l-((6-(trifluoromethyl)pyridin-2-yl)methyl)piperidin-4- yl)butanamide;
(cis)-(2R)-N-(3,5-dimethyladamantan-l-yl)-2-(4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
N-((3s,5s,7s)-Adamantan-l-yl)-2-(l -(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide; 2-(l-(6-Fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)-N-((lr,3s,5R,7S)-3- hydroxyadamantan-l -yl)acetamide; or
N-((3s,5s,7s)-Adamantan-l -yl)-2-(4-methyl-l -(2-(trifluoromethyl)pyridin-4-yl)piperidin yl)acetamide;
or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
Aspect 17. A pharmaceutical composition comprising a compound of any one of Aspects 1 to 16 and a pharmaceutically acceptable carrier.
Aspect 18. The pharmaceutical composition of Aspect 17, further comprising YERVOY, OPDIVO, or KEYTRUDA, or a combination thereof.
Aspect 19. A method of treating cancer in a patient in need of such treatment comprising administering to the patient a therapeutically effective amount of a compound according to any one of Aspects 1 to 16.

Claims

What is Claimed:
1. A compound of formula I or formula II
Figure imgf000075_0001
wherein
X is CH or N;
T is CH or N;
R1 is H, halo, or Ci-Cehaloalkyl;
R1A is H, halo, or Ci-C6haloalkyl;
Y is CH or N;
n is 0, 1, 2, 3, or 4;
W is N or CR4 wherein R4 is H or (Ci-C6alkyl);
V is Ci-Cealkylene optionally substituted with one, two, or three substituents
independently selected from Ci-Cealkyl and C3-Cecycloalkyl;
R2 is H or Ci-C6alkyl;
Z is a bond or Ci-Cealkylene optionally substituted with one, two, or three substituents independently selected from Ci-Cealkyl and C3-Cecycloalkyl;
B is aryl optionally substituted with 1, 2, or 3 R substituents independently selected from -OH, Ci-C6alkyl, Ci-C6haloalkyl, halo, -OCi-C6alkyl, -OCi-C6haloalkyl, -
CN, aryl, or -Oaryl; or
C3-Ci2cycloalkyl optionally substituted with 1, 2, or 3 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi- Cealkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl;
or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
2. The compound of claim 1 that is a compound of formula I.
3. The compound of claim 1 that is a compound of formula II.
4. The compound of any one of the preceding claims, wherein R is H, F, or -CF3.
5. The compound of any one of the preceding claims, wherein R is H.
6. The compound of any one of the preceding claims, wherein Y is CH.
7. The compound of any one of claims 1 to 3, wherein Y is N.
8. The compound of any one of the preceding claims, wherein n is 2.
9. The compound of any one of the preceding claims, wherein W is CR4.
10. The compound of any one of claims 1 to 8, wherein W is N.
11. The compound of any one of the preceding claims, wherein V is Cialkylene optionally substituted with one substituent that is Ci-C6alkyl and C3-C6cycloalkyl.
12. The compound of any one of the preceding claims, wherein Z is Cialkylene optionally substituted with one or two substituents independently selected from Ci-Cealkyl and C3- Cecycloalkyl.
13. The compound of any one of the preceding claims, wherein B is aryl optionally
substituted with 1, 2, or 3 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi-C6alkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl.
14. The compound of claim 13, wherein B is phenyl optionally substituted with 1 or 2 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi- Cealkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl.
15. The compound of any one of claims 1 to 12, wherein B is C3-Ci2cycloalkyl optionally substituted with 1, 2, or 3 R substituents independently selected from -OH, Ci-Cealkyl, Ci-Cehaloalkyl, halo, -OCi-C6alkyl, -OCi-C6haloalkyl, -CN, aryl, or -Oaryl.
16. The compound of claim 1 that is
N-(4-Chloro-2-methylbenzyl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide; N-(l -(2,4-dichlorophenyl)cyclo-propyl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide;
-(2-(4-chlorophenyl)propan-2-yl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide;
(S)-N-(l-(4-chlorophenyl)ethyl)-2-(l-(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide;
N-(4-chlorobenzyl)-2-(l -(6-fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)acetamide;
(S)-2-(4-Methyl-l-(2-(trifluoromethyl)pyridin-4-yl)piperidin-4-yl)-N-(l- phenylpropyl)acetamide;
N-(l -(2,4-dichlorophenyl)cy clo-propyl)-2-(4-methyl- 1 -(2-(trifluoro-methyl)pyridin-4- yl)piperidin-4-yl)acetamide;
N-(2-(4-chlorophenyl)propan-2-yl)-2-(4-methyl-l-(2-(trifluoromethyl)-pyridin-4- yl)piperidin-4-yl)acetamide;
(S)-N-(l -(4-chlorophenyl)ethyl)-2-(4-methyl-l -(2-(trifluoromethyl)-pyridin-4-yl)piperi yl)acetamide;
(i?)-N-(2-chlorobenzyl)-2-((cz ,)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide;
(i?)-N-(2-chlorobenzyl)-2-((cz ,)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide;
(R)-N-([\ , 1 '-bipheny 1] -4-y lmethy l)-2-((cw)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(i?)-2-((cw)-4-(6-fluoroquinolin-4-yl)cyclohexyl)-N-((^-l-phenylethyl)propanamide;
(i?)-N-((5 -l-(4-chlorophenyl)ethyl)-2-((cw)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(i?)-N-((5 -l-(4-chlorophenyl)ethyl)-2-((cw)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(i?)-N-(l-(2,4-dichlorophenyl)cyclopropyl)-2-((cw)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(i?)-N-((i?)-l -(4-chlorophenyl)ethyl)-2-((l s,4S)-4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
(R)-N-benzyl-2-((l s,4S)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide;
N-((3R,5R R)-Adamantan-l-yl)-2-(4-(6-fluoroquinolin-4-yl)piperazin-l -yl)pentanamide; N-(Adamantan-2-yl)-2-(l-((6-(trifluoromethyl)pyridin-2-yl)methyl)piperidin-4- yl)butanamide; (cis)-(2R)-N-(3,5-dimethyladamantan-l-yl)-2-(4-(6-fluoroquinolin-4- yl)cyclohexyl)propanamide;
N-((3s,5s,7s)-Adamantan-l-yl)-2-(l -(6-fluoroquinolin-4-yl)-4-methylpiperidin-4- yl)acetamide;
2-(l-(6-Fluoroquinolin-4-yl)-4-methylpiperidin-4-yl)-N-((lr,3s,5R,7S)-3- hydroxyadamantan-l -yl)acetamide; or
N-((3s,5s,7s)-Adamantan-l -yl)-2-(4-methyl-l -(2-(trifluoromethyl)pyridin-4-yl)piperidin yl)acetamide;
or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, a tautomer thereof, or a solvate thereof.
17. A pharmaceutical composition comprising a compound of any one of claims 1 to 16 and a pharmaceutically acceptable carrier.
18. The pharmaceutical composition of claim 17, further comprising YERVOY, OPDIVO, or KEYTRUDA, or a combination thereof.
19. A method of treating cancer in a patient in need of such treatment comprising
administering to the patient a therapeutically effective amount of a compound according to any one of claims 1 to 16.
PCT/US2017/030997 2016-05-04 2017-05-04 Inhibitors of indoleamine 2,3-dioxygenase and methods of their use WO2017192813A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2018557904A JP2019516687A (en) 2016-05-04 2017-05-04 Indoleamine 2,3-dioxygenase inhibitors and methods of use thereof
US16/098,245 US11066383B2 (en) 2016-05-04 2017-05-04 Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
EP17793316.5A EP3452451A4 (en) 2016-05-04 2017-05-04 Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
CN201780040617.8A CN109311816A (en) 2016-05-04 2017-05-04 The inhibitor and its application method of indole amine 2,3-dioxygenase
KR1020187034654A KR20190004742A (en) 2016-05-04 2017-05-04 Inhibitors of indoleamine 2,3-dioxygenase and methods of use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662331871P 2016-05-04 2016-05-04
US62/331,871 2016-05-04

Publications (1)

Publication Number Publication Date
WO2017192813A1 true WO2017192813A1 (en) 2017-11-09

Family

ID=60203365

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/030997 WO2017192813A1 (en) 2016-05-04 2017-05-04 Inhibitors of indoleamine 2,3-dioxygenase and methods of their use

Country Status (6)

Country Link
US (1) US11066383B2 (en)
EP (1) EP3452451A4 (en)
JP (1) JP2019516687A (en)
KR (1) KR20190004742A (en)
CN (1) CN109311816A (en)
WO (1) WO2017192813A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109574988A (en) * 2017-12-25 2019-04-05 成都海博锐药业有限公司 A kind of compound and application thereof
WO2019074822A1 (en) * 2017-10-09 2019-04-18 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
WO2019099294A1 (en) 2017-11-14 2019-05-23 Merck Sharp & Dohme Corp. Novel substituted biaryl compounds as indoleamine 2,3-dioxygenase (ido) inhibitors
WO2019120198A1 (en) * 2017-12-18 2019-06-27 成都华健未来科技有限公司 Quinoline derivatives
WO2019120200A1 (en) * 2017-12-18 2019-06-27 成都华健未来科技有限公司 Quinoline derivative and uses thereof
WO2019120199A1 (en) * 2017-12-18 2019-06-27 成都华健未来科技有限公司 Quinoline derivatives and bioactivity thereof
WO2019129114A1 (en) * 2017-12-29 2019-07-04 杭州阿诺生物医药科技有限公司 Indoleamine 2,3-dioxygenase inhibitors and use of same in medicine
WO2019154380A1 (en) * 2018-02-11 2019-08-15 南京明德新药研发股份有限公司 Kynurenine pathway inhibitor
WO2019179369A1 (en) * 2018-03-19 2019-09-26 四川科伦博泰生物医药股份有限公司 Ring-containing compound, preparation method therefor, and medical application thereof
WO2020112581A1 (en) * 2018-11-28 2020-06-04 Merck Sharp & Dohme Corp. Novel substituted piperazine amide compounds as indoleamine 2, 3-dioxygenase (ido) inhibitors
CN111630048A (en) * 2018-03-19 2020-09-04 四川科伦博泰生物医药股份有限公司 Amidine and guanidine derivatives, preparation method and application thereof in medicines

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109843322A (en) * 2016-08-26 2019-06-04 百时美施贵宝公司 The inhibitor and its application method of indole amine 2,3-dioxygenase
US11839659B2 (en) 2020-07-02 2023-12-12 Northwestern University Proteolysis-targeting chimeric molecules (PROTACs) that induce degradation of indoleamine 2,3-dioxygenase (IDO) protein

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015006520A1 (en) * 2013-07-11 2015-01-15 Bristol-Myers Squibb Company Ido inhibitors
WO2015188085A1 (en) * 2014-06-06 2015-12-10 Flexus Biosciences, Inc. Immunoregulatory agents
US20160022619A1 (en) * 2013-03-15 2016-01-28 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase (ido)

Family Cites Families (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU1628599A (en) 1997-12-05 1999-06-28 Medical College Of Georgia Research Institute, Inc. Regulation of t cell-mediated immunity by tryptophan
JP4921965B2 (en) 2003-03-27 2012-04-25 ランケナー インスティテュート フォー メディカル リサーチ New cancer treatment system
TWI380996B (en) 2004-09-17 2013-01-01 Hoffmann La Roche Anti-ox40l antibodies
ES2432091T5 (en) 2005-03-25 2022-03-18 Gitr Inc GITR binding molecules and uses thereof
KR101386494B1 (en) 2005-05-10 2014-04-24 인사이트 코포레이션 Modulators of indoleamine 2,3-dioxygenase and methods of using the same
US20070004763A1 (en) 2005-06-10 2007-01-04 Nand Baindur Aminoquinoline and aminoquinazoline kinase modulators
CN104356236B (en) 2005-07-01 2020-07-03 E.R.施贵宝&圣斯有限责任公司 Human monoclonal antibodies to programmed death ligand 1(PD-L1)
JP5294874B2 (en) 2005-12-20 2013-09-18 インサイト・コーポレイション N-hydroxyamidino heterocycle as modulator of indoleamine 2,3-dioxygenase
CL2007002650A1 (en) 2006-09-19 2008-02-08 Incyte Corp COMPOUNDS DERIVED FROM HETEROCICLO N-HIDROXIAMINO; PHARMACEUTICAL COMPOSITION, USEFUL TO TREAT CANCER, VIRAL INFECTIONS AND NEURODEGENERATIVE DISORDERS BETWEEN OTHERS.
WO2008036642A2 (en) 2006-09-19 2008-03-27 Incyte Corporation N-hydroxyamidinoheterocycles as modulators of indoleamine 2,3-dioxygenase
EP1987839A1 (en) 2007-04-30 2008-11-05 I.N.S.E.R.M. Institut National de la Sante et de la Recherche Medicale Cytotoxic anti-LAG-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
ES2591281T3 (en) 2007-07-12 2016-11-25 Gitr, Inc. Combination therapies that employ GITR binding molecules
EP2044949A1 (en) 2007-10-05 2009-04-08 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
EP2227233B1 (en) 2007-11-30 2013-02-13 Newlink Genetics Ido inhibitors
AR072999A1 (en) 2008-08-11 2010-10-06 Medarex Inc HUMAN ANTIBODIES THAT JOIN GEN 3 OF LYMPHOCYTARY ACTIVATION (LAG-3) AND THE USES OF THESE
PE20120341A1 (en) 2008-12-09 2012-04-24 Genentech Inc ANTI-PD-L1 ANTIBODIES AND ITS USE TO IMPROVE T-CELL FUNCTION
US8709424B2 (en) 2009-09-03 2014-04-29 Merck Sharp & Dohme Corp. Anti-GITR antibodies
WO2011056652A1 (en) 2009-10-28 2011-05-12 Newlink Genetics Imidazole derivatives as ido inhibitors
ES2722300T3 (en) 2009-12-10 2019-08-09 Hoffmann La Roche Antibodies that preferentially bind to extracellular domain 4 of CSF1R and its use
CA2791658C (en) 2010-03-04 2019-10-01 Macrogenics, Inc. Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
US9169323B2 (en) 2010-03-05 2015-10-27 Hoffmann-La Roche Inc. Antibodies against human CSF-1R
RU2617966C2 (en) 2010-03-05 2017-04-28 Ф.Хоффманн-Ля Рош Аг Antibodies to human csf-1r and use thereof
WO2011140249A2 (en) 2010-05-04 2011-11-10 Five Prime Therapeutics, Inc. Antibodies that bind csf1r
SG10201506906VA (en) 2010-09-09 2015-10-29 Pfizer 4-1bb binding molecules
NO2694640T3 (en) 2011-04-15 2018-03-17
CA2833636A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
LT2785375T (en) 2011-11-28 2020-11-10 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
CN104159921B (en) 2011-12-15 2018-05-04 霍夫曼-拉罗奇有限公司 Antibody for people CSF-1R and application thereof
RU2014136332A (en) 2012-02-06 2016-03-27 Дженентек, Инк. COMPOSITIONS AND METHODS OF APPLICATION OF CSF1R INHIBITORS
AR090263A1 (en) 2012-03-08 2014-10-29 Hoffmann La Roche COMBINED ANTIBODY THERAPY AGAINST HUMAN CSF-1R AND USES OF THE SAME
RU2670743C9 (en) 2012-05-11 2018-12-19 Файв Прайм Терапьютикс, Инк. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
US9428460B2 (en) * 2012-06-26 2016-08-30 Bayer Pharma Aktiengesellschaft N-[4-(quinolin-4-yloxy)cyclohexyl(methyl)](hetero)arylcarboxamides as androgen receptor antagonists, production and use thereof as medicinal products
UY34887A (en) 2012-07-02 2013-12-31 Bristol Myers Squibb Company Una Corporacion Del Estado De Delaware OPTIMIZATION OF ANTIBODIES THAT FIX THE LYMPHOCYTE ACTIVATION GEN 3 (LAG-3) AND ITS USES
RU2718751C2 (en) 2012-08-31 2020-04-14 Файв Прайм Терапьютикс, Инк. Methods of treating pathological conditions with antibodies which bind to the colony-stimulating factor 1 receptor (csf1r)
US10002275B2 (en) * 2013-07-08 2018-06-19 Ramot At Tel-Aviv University Ltd. Optical element having two phase masks
GB201419579D0 (en) 2014-11-03 2014-12-17 Iomet Pharma Ltd Pharmaceutical compound
MX2017005462A (en) 2014-11-05 2017-07-28 Flexus Biosciences Inc Immunoregulatory agents.
UY36390A (en) * 2014-11-05 2016-06-01 Flexus Biosciences Inc MODULATING COMPOUNDS OF INDOLAMINE ENZYME 2,3-DIOXYGENASE (IDO), ITS SYNTHESIS METHODS AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
AR102537A1 (en) 2014-11-05 2017-03-08 Flexus Biosciences Inc IMMUNOMODULATING AGENTS
GB201511790D0 (en) 2015-07-06 2015-08-19 Iomet Pharma Ltd Pharmaceutical compound

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160022619A1 (en) * 2013-03-15 2016-01-28 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase (ido)
WO2015006520A1 (en) * 2013-07-11 2015-01-15 Bristol-Myers Squibb Company Ido inhibitors
WO2015188085A1 (en) * 2014-06-06 2015-12-10 Flexus Biosciences, Inc. Immunoregulatory agents

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11649212B2 (en) 2017-10-09 2023-05-16 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
WO2019074822A1 (en) * 2017-10-09 2019-04-18 Bristol-Myers Squibb Company Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
WO2019099294A1 (en) 2017-11-14 2019-05-23 Merck Sharp & Dohme Corp. Novel substituted biaryl compounds as indoleamine 2,3-dioxygenase (ido) inhibitors
EP3709986A4 (en) * 2017-11-14 2021-05-26 Merck Sharp & Dohme Corp. Novel substituted biaryl compounds as indoleamine 2,3-dioxygenase (ido) inhibitors
US11498904B2 (en) 2017-11-14 2022-11-15 Merck Sharp & Dohme Llc Substituted biaryl compounds as indoleamine 2,3-dioxygenase (IDO) inhibitors
WO2019120200A1 (en) * 2017-12-18 2019-06-27 成都华健未来科技有限公司 Quinoline derivative and uses thereof
CN111527069B (en) * 2017-12-18 2023-08-08 成都华健未来科技有限公司 Quinoline derivatives
WO2019120199A1 (en) * 2017-12-18 2019-06-27 成都华健未来科技有限公司 Quinoline derivatives and bioactivity thereof
CN111527069A (en) * 2017-12-18 2020-08-11 成都华健未来科技有限公司 Quinoline derivatives
WO2019120198A1 (en) * 2017-12-18 2019-06-27 成都华健未来科技有限公司 Quinoline derivatives
CN109574988A (en) * 2017-12-25 2019-04-05 成都海博锐药业有限公司 A kind of compound and application thereof
CN109574988B (en) * 2017-12-25 2022-01-25 成都海博锐药业有限公司 Compound and application thereof
WO2019129114A1 (en) * 2017-12-29 2019-07-04 杭州阿诺生物医药科技有限公司 Indoleamine 2,3-dioxygenase inhibitors and use of same in medicine
CN111741946B (en) * 2017-12-29 2022-09-23 厦门宝太生物科技股份有限公司 Indoleamine 2, 3-dioxygenase inhibitors and their use in medicine
CN111741946A (en) * 2017-12-29 2020-10-02 杭州阿诺生物医药科技有限公司 Indoleamine 2, 3-dioxygenase inhibitors and their use in medicine
US11111230B2 (en) 2017-12-29 2021-09-07 Adlai Nortye Biopharma Co., Ltd. Indoleamine 2,3-dioxygenase inhibitors and use of same in medicine
GB2583606A (en) * 2017-12-29 2020-11-04 Adlai Nortye Biopharma Co Ltd Indoleamine 2,3-Dioxygenase inhibitors and use of same in medicine
GB2583606B (en) * 2017-12-29 2021-04-21 Adlai Nortye Biopharma Co Ltd Indoleamine 2,3-Dioxygenase inhibitors and use of same in medicine
CN111868032B (en) * 2018-02-11 2021-09-03 基石药业(苏州)有限公司 Kynurenine pathway inhibitors
CN111868032A (en) * 2018-02-11 2020-10-30 基石药业(苏州)有限公司 Kynurenine pathway inhibitors
WO2019154380A1 (en) * 2018-02-11 2019-08-15 南京明德新药研发股份有限公司 Kynurenine pathway inhibitor
CN111630048A (en) * 2018-03-19 2020-09-04 四川科伦博泰生物医药股份有限公司 Amidine and guanidine derivatives, preparation method and application thereof in medicines
CN111630043B (en) * 2018-03-19 2022-02-15 四川科伦博泰生物医药股份有限公司 Compound containing ring, preparation method and application thereof in medicine
CN111630043A (en) * 2018-03-19 2020-09-04 四川科伦博泰生物医药股份有限公司 Compound containing ring, preparation method and application thereof in medicine
WO2019179369A1 (en) * 2018-03-19 2019-09-26 四川科伦博泰生物医药股份有限公司 Ring-containing compound, preparation method therefor, and medical application thereof
CN111630048B (en) * 2018-03-19 2023-09-29 四川科伦博泰生物医药股份有限公司 Amidine and guanidine derivatives, preparation method and medical application thereof
WO2020112581A1 (en) * 2018-11-28 2020-06-04 Merck Sharp & Dohme Corp. Novel substituted piperazine amide compounds as indoleamine 2, 3-dioxygenase (ido) inhibitors

Also Published As

Publication number Publication date
JP2019516687A (en) 2019-06-20
US11066383B2 (en) 2021-07-20
EP3452451A4 (en) 2019-11-13
KR20190004742A (en) 2019-01-14
EP3452451A1 (en) 2019-03-13
CN109311816A (en) 2019-02-05
US20190144417A1 (en) 2019-05-16

Similar Documents

Publication Publication Date Title
US11066383B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US10544099B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US10633342B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US11337970B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US10696648B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US11066392B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US10323004B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US11351164B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US11649212B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use
US11203592B2 (en) Inhibitors of indoleamine 2,3-dioxygenase and methods of their use

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2018557904

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 17793316

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 20187034654

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017793316

Country of ref document: EP

Effective date: 20181204