WO2019152982A1 - Composés hétérobicycliques en tant qu'antagonistes ep4 - Google Patents

Composés hétérobicycliques en tant qu'antagonistes ep4 Download PDF

Info

Publication number
WO2019152982A1
WO2019152982A1 PCT/US2019/016637 US2019016637W WO2019152982A1 WO 2019152982 A1 WO2019152982 A1 WO 2019152982A1 US 2019016637 W US2019016637 W US 2019016637W WO 2019152982 A1 WO2019152982 A1 WO 2019152982A1
Authority
WO
WIPO (PCT)
Prior art keywords
trifluoromethyl
phenoxy
carboxamido
benzoic acid
alkyl
Prior art date
Application number
PCT/US2019/016637
Other languages
English (en)
Inventor
Yongkui Sun
Gang Zhou
Weiguo QUAN
Zhaoyin Wang
Original Assignee
Shenzhen Ionova Life Science Co., Ltd.
Foshan Ionova Biotherapeutics Co., Ltd.
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 Shenzhen Ionova Life Science Co., Ltd., Foshan Ionova Biotherapeutics Co., Ltd. filed Critical Shenzhen Ionova Life Science Co., Ltd.
Priority to CN201980010302.8A priority Critical patent/CN111936138B/zh
Publication of WO2019152982A1 publication Critical patent/WO2019152982A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/18Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D209/24Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with an alkyl or cycloalkyl radical attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/08Indoles; Hydrogenated indoles with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/12Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/18Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals

Definitions

  • This invention relates to heterocyclic amide derivatives, or their pharmaceutically acceptable salts, pharmaceutical acceptable prodrugs, pharmaceutical compositions containing them, and their medical uses.
  • the compounds of this invention have activity as prostaglandin E2 receptor antagonists, and these are useful in the treatment or alleviation of pain and inflammation and other inflammation-associated disorders, such as arthritis, treating or preventing disorders or medical conditions selected from pain, inflammatory diseases and the like.
  • Prostaglandins are mediators of pain, fever and other symptoms associated with inflammation. Especially prostaglandin E2 (PGE2) is the predominant eicosanoid detected in inflammation conditions. In addition, PGE2 has also been implicated as an important constituent in the immunosuppressive environment created by many solid tumors (Whiteside, Expert Opinion in Biological Therapy, 2010. 10, 1019-1035), and inhibition of EP4 signaling by antagonists were shown to reduce tumor growth (Terada et al. Cancer Res. 2010, 70, 1606-1615) and tumor metastasis in tumor animal models (Yang et al. Cancer Res. 2006, 66, 9665-9672).
  • PGE2 has also been shown to promote colorectal cancer stem cell expansion and metastasis in mice (Wang et al., Gastroentology, 2015, 1-12).
  • the anti-tumor activity EP4 antagonists was also shown to improve or synergize with the anti-tumor effect of antibodies against checkpoint proteins, such as anti-CTLA4, anti-PD-Ll or anti-PD-1 (Bao et al. Journal for ImmunoTherapy of Cancer 2015, 3(Suppl 2):P35).
  • This invention relates to a series of novel EP4 antagonists and methods for treating prostaglandin E2 mediated diseases, in particular for pain, inflammation and cancer immunotherapy, along with certain pharmaceutical compositions thereof.
  • the invention is directed to a series of novel heterobicyclic derivatives as EP4 receptor antagonists useful for the treatment of EP4 mediated diseases or conditions, such as acute and chronic pain, osteoarthritis, rheumatoid arthritis and cancer. Pharmaceutical compositions and methods of use are also included.
  • the present invention provides compounds of Formula I or a pharmaceutically acceptable salt or prodrug thereof,
  • each of A, B and C' is independently selected from N, CH and C(R);
  • each of R la and R 2a independently is H, Ci- 6 alkyl, Ci- 6 cyclolkyl, Ci- 6 fluorocycloalkyl, or Ci- 6 fluoroalkyl; or R 1 and R 2 , together with the carbon atom to which they are both attached to, form a three- to six-membered carbocyclic ring which is optionally substituted with R c , or form a three- to six-membered ring which contains one or two heteroatom(s) each independently being S, S(0) 2 , S(0)(NH), O, or NR e , wherein R e is H, Ci -6 alkyl, Ci- 6 cyclolkyl, Ci- 6 fluorocycloalkyl, Ci_ 6 fluoroalkyl, aryl, heteroaryl, C(O) Ci- 6 alkyl, C(0)aryl, S(0) 2 alkyl, or S(0) 2 aryl;
  • each R c independently is H, halogen, Ci -4 alkyl, Ci -4 fluoroalkyl, Ci -4 alkoxy, Ci -4 fluoroalkoxy, or acetyl;
  • each R independently is H, Ci -4 alkyl, Ci -4 fluoroalkyl, Ci -4 alkoxy, Ci -4 fluoroalkoxy, or acetyl;
  • Y is -CH 2 -, -C(0)-, -C(S)-, or -S(0) 2 -;
  • X is CH 2 , O, or S
  • W is NH or O; each of Ar 1 and Ar 2 , independently, is C3-6 cycloalkyl, aryl, heteroaryl, or heterocyclyl, or a fused analog of C3-6cycloalkyl, aryl, heteroaryl or heterocyclyl, and each of Ar 1 and Ar 2 , is optionally substituted with one to three R b groups;
  • each R b is independently H, halogen, Ci_ 6 alkyl, Ci- 6 alkoxyl, or Ci- 6 fluoroalkyl;
  • M is a pharmaceutically acceptable salt or an ester prodrug group.
  • the compounds of Formula I are of Formula la
  • each of A, B and C' independently is N, CH, or C(R);
  • each R b is independently H, halogen, Ci- 6 alkyl, Ci- 6 alkoxyl, or Ci- 6 fluoroalkyl;
  • each of R la and R 2a is independently H, Ci -6 alkyl, Ci- 6 cyclolkyl, Ci- 6 fluorocycloalkyl, or Ci- 6 fluoroalkyl, or
  • each of R 1 and R 2 is H, halogen, Ci -6 alkyl, Ci -6 alkoxyl, Ci- 6 cyclolkyl, Ci- 6 fluorocycloalkyl, Ci- 6 fluoroalkyl, or SF5; or R 1 and R 2 , together with the carbon atom to which they are both attached to, form a three- to six-membered carbocyclic ring which is optionally substituted with Rc or a three- to six-membered ring which contains one or two heteroatom(s) each independently being S, S(0) 2 , S(0)(NH), O, or NR e , wherein R e is H, Ci- 6 alkyl, Ci- 6 cyclolkyl, Ci_ 6 fluorocycloalkyl, Ci- 6 fluoroalkyl, aryl, heteroaryl, C(0)Ci- 6 alkyl, C(0)aryl, S(0) 2 alkyl, or S(0) 2 aryl; each R c is H, Ci
  • X is CH 2 , O, or S
  • each R b is independently H, halogen, Cl-6alkyl, Cl-6alkoxyl, or Cl-6fluoroalkyl;
  • n 0,1, 2, or 3.
  • the compounds are of Formula lb:
  • each of R1 and R2 is independently!-!, halogen, Cl-6alkyl, Cl-6alkoxyl, Cl-6cyclolkyl, Cl- 6fluorocycloalkyl, Cl-6fluoroalkyl, or SF5;
  • each Rb independently is H, halogen, Cl-6alkyl, Cl-6alkoxyl, or Cl-6fluoroalkyl;
  • each of Rla and R2a independently is H, Cl-6alkyl, Cl-6cyclolkyl, Cl-6fluorocycloalkyl, or Cl-6fluoroalkyl; or R1 and R2, together with the carbon atom to which they are both attached to, form a three- to six-membered carbocyclic ring which is optionally substituted with Rc or a three- to six-membered ring which contains one or two heteroatom(s) each independently being S, O or NRe, wherein Re is H or Cl-6alkyl;
  • each Rc independently is H, Cl-4alkyl, Cl-4fluoroalkyl, Cl-4alkoxy, Cl-4fluoroalkoxy or acetyl;
  • each R independently is H, halogen, Cl-4alkyl, Cl-4fluoroalkyl, Cl-4alkoxy, Cl- 4fluoroalkoxy or acetyl;
  • each Rb independently is H, halogen, Cl-6alkyl, Cl-6alkoxyl, or Cl-6fluoroalkyl; and n is 0,1, 2, or 3.
  • the fused ring moeity compounds of Formula I is selected from the following heterocyclic moieties:
  • Examples of the compounds of the present invention include
  • the invention also encompasses salts or prodrugs of compounds of Formula I.
  • a prodrug can be a choice of an ester or amide of the compound of Formula I.
  • the present invention provides pharmaceutical compositions each comprising an above-described compound and a pharmaceutically or physiologically acceptable carrier.
  • the present invention also relates to a method for treating a subject suffering from a condition mediated by the action of PGE2 at EP4 receptors.
  • the method includes administering to the subject in need thereof an effective amount of an above-described compound or pharmaceutical composition.
  • the condition is an inflammatory disease or cancer.
  • the inflammatory disease include arthritis, acne vulgaris, asthma, autoimmune diseases, autoinflammatory diseases, Celiac disease, chronic prostatitis, colitis, diverticulitis, glomerulonephritis, hidradenitis suppurativa, hypersensitivities, inflammatory bowel diseases, interstitial cystitis, Mast Cell Activation Syndrome, mastocytosis, otitis, pelvic inflammatory disease, reperfusion injury, rheumatic fever, rheumatoid arthritis, rhinitis, sarcoidosis, and vasculitis.
  • the condition is cancer and the method comprises administering to the subject in need thereof an effective amount of an above-described compound or composition, optionally in combination with a second therapeutic agent which is an antibody, a kinase inhibitor, IDO inhibitor, TDO inhibitor, STING activator, HDAC inhibitor, or an chemotherapeutic agent.
  • a second therapeutic agent which is an antibody, a kinase inhibitor, IDO inhibitor, TDO inhibitor, STING activator, HDAC inhibitor, or an chemotherapeutic agent.
  • a suitable antibody include those against CTCLA4, PDL1 and PD1.
  • alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight (i.e. unbranched) or branched chain, or cyclic hydrocarbon radical, or combination thereof, which may be fully saturated, mono-or polyunsaturated and can include di-and multivalent radicals, having the number of carbon atoms designated (i.e. C1-C10 means one to ten carbons).
  • saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • An unsaturated alkyl group is one having one or more double bonds or triple bonds.
  • unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(l,4-pentadienyl), ethynyl, 1-and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
  • Alkyl groups which are limited to hydrocarbon groups are termed "homoalkyl".
  • fluoroalkyl means alkyl as defined above wherein one or more hydrogen atoms have been replaced by fluoro atoms.
  • an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention.
  • a “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
  • alkynyl means carbon chains which contain at least one carbon-carbon triple bond, and which may be linear or branched or combinations thereof.
  • alkynyl include ethynyl, propargyl, 3-methyl-l-pentynyl, 2-heptynyl and the like.
  • cycloalkyl means mono- or bicyclic saturated carbocyclic rings, each of which having from 3 to 10 carbon atoms.
  • a "fused analog" of cycloalkyl means a monocyclic rings fused to an aryl or heteroaryl group in which the point of attachment is on the non-aromatic portion. Examples of cycloalkyl and fused analogs thereof include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, tetrahydronaphthyl, decahydronaphthyl, indanyl, and the like.
  • alkoxy means alkoxy groups of a straight or branched having the indicated number of carbon atoms.
  • Ci- 6 alkoxy for example, includes methoxy, ethoxy, propoxy, isopropoxy, and the like.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of at least one carbon atoms and at least one heteroatom selected from the group consisting of O, N, P, Si and S, and wherein the nitrogen, phosphorus, and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N, P and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which alkyl group is attached to the remainder of the molecule.
  • heteroalkylene by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified, but not limited by, -CH2 -CH2- S -CH2-CH2- and -CH2-S-CH2-CH2-NH-CH2-.
  • heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxo, alkylenedioxo, alkyleneamino, alkylenediamino, and the like).
  • no orientation of the linking group is implied by the direction in which the formula of the linking group is written.
  • heteroalkyl groups include those groups that are attached to the remainder of the molecule through a heteroatom, such as -C(0)R', -C(0)NR', -NR'R", -OR', - SR', and/or -S02R'.
  • heteroalkyl is recited, followed by recitations of specific heteroalkyl groups, such as -NR'R" or the like, it will be understood that the terms heteroalkyl and--NR'R" are not redundant or mutually exclusive. Rather, the specific heteroalkyl groups are recited to add clarity. Thus, the term “heteroalkyl” should not be interpreted herein as excluding specific heteroalkyl groups, such as -NR'R” or the like.
  • cycloalkoxy means cycloalkyl as defined above bonded to an oxygen atom, such as cyclopropyloxy.
  • fluoroalkoxy means alkoxy as defined above wherein one or more hydrogen atoms have been replaced by fluoro atoms.
  • aryl means mono- or bicyclic aromatic rings containing only carbon atoms.
  • a "fused analog" of aryl means an aryl group fused to a monocyclic cycloalkyl or monocyclic heterocyclyl group in which the point of attachment is on the aromatic portion. Examples of aryl and fused analogs thereof include phenyl, naphthyl, indanyl, indenyl, tetrahydronaphthyl, 2,3-dihydrobenzofuranyl, dihydrobenzopyranyl, 1,4-benzodioxanyl, and the like.
  • heteroaryl means a mono- or bicyclic aromatic ring containing at least one heteroatom selected from N, O and S, with each ring containing 5 to 6 atoms.
  • a "fused analog" of heteroaryl means a heteroaryl group fused to a monocyclic cycloalkyl or monocyclic heterocyclyl group in which the point of attachment is on the aromatic portion.
  • heteroaryl examples include pyrrolyl, isoxazolyl, isothiazolyl, pyrazolyl, pyridyl, oxazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, triazinyl, thienyl, pyrimidyl, pyridazinyl, pyrazinyl, benzoxazolyl, benzothiazolyl, benzimidazolyl, benzofuranyl, benzothiophenyl, furo(2,3-b)pyridyl, quinolyl, indolyl, isoquinolyl, and the like.
  • aryl groups and heteroaryl groups referred to in the definitions of Arl and Ar2 are unsubstituted or are substituted by at least one substituent selected from the group consisting of substituents 0.
  • the substituents are selected from the group consisting of halogen atoms, alkyl groups having from 1 to 4 carbon atoms, alkoxy groups having from 1 to 4 carbon atoms, haloalkyl groups having from 1 to 4 carbon atoms, haloalkoxy groups having from I to 4 carbon atoms, cyano groups, alkynyl groups having from 2 to 6 carbon atoms, alkanoyl groups having from 1 to 5 carbon atoms, cycloalkyl groups having from 3 to 7 ring atoms, heteroaryl groups, aryl groups, aralkoxy groups having from 7 to 10 carbon atoms, arylcarbonyl groups, two adjacent-x groups are optionally joined together to form an alkylene or an alkenylene chain having 3 or 4 carbon atoms, aminocarbonyl groups, alkenyl groups having from 2 to 5 carbon atoms, alkylthio groups having from 1 to 4 carbon atoms, aminosulfinyl groups, aminosul
  • heterocyclyl means mono- or bicyclic saturated rings containing at least one heteroatom selected from N, S and O, each of said ring having from 3 to 10 atoms in which the point of attachment may be carbon or nitrogen.
  • a "fused analog" of heterocyclyl means a monocyclic heterocycle fused to an aryl or heteroaryl group in which the point of attachment is on the non-aromatic portion.
  • heterocyclyl and fused analogs thereof include pyrrolidinyl, piperidinyl, piperazinyl, imidazolidinyl, 2,3-dihydrofuro(2,3- b)pyridyl, benzoxazinyl, tetrahydrohydroquinolinyl, tetrahydroisoquinolinyl, dihydroindolyl, and the like.
  • the term also includes partially unsaturated monocyclic rings that are not aromatic, such as 2- or 4-pyridones attached through the nitrogen or N-substituted-(lH,3H)-pyrimidine- 2,4-diones (N-substituted uracils).
  • halo or halogen
  • substituents mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom.
  • terms such as “haloalkyl,” are meant to include monohaloalkyl and polyhaloalkyl.
  • halo(Cl-C4)alkyl is mean to include, but not be limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
  • prodrug refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrugs may also have improved solubility in pharmaceutical compositions over the parent drug.
  • An example, without limitation, of a prodrug would be a compound of any of Formula I, which is administered as an ester (the "prodrug") to facilitate transmittal across a cell membrane where water solubility is detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the active entity, once inside the cell where water-solubility is beneficial.
  • a further example of a prodrug might be a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety.
  • Compounds of Formula I contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. The present invention is meant to comprehend all such isomeric forms of the compounds of Formula I.
  • tautomers Some of the compounds described herein may exist with different points of attachment of hydrogen, referred to as tautomers. Such an example may be a ketone and its enol form known as keto-enol tautomers. The individual tautomers as well as mixture thereof are encompassed with compounds of Formula I to Formula Ig.
  • Compounds of the Formula I may be separated into diastereoisomeric pairs of enantiomers by, for example, fractional crystallization from a suitable solvent, for example MeOH or EtOAc or a mixture thereof.
  • a suitable solvent for example MeOH or EtOAc or a mixture thereof.
  • the pair of enantiomers thus obtained may be separated into individual stereoisomers by conventional means, for example by the use of an optically active amine as a resolving agent or on a chiral HPLC column.
  • any enantiomer of a compound of the general Formula I may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known configuration.
  • salts refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids.
  • Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylenediamine, diethylamine, 2-diethyl- aminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
  • basic ion exchange resins such as
  • salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p- toluenesulfonic acid, and the like.
  • Particularly preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.
  • the compounds of the invention are antagonists of the EP4 receptor and are therefore useful in treating a prostaglandin E2 mediated disease or condition.
  • the invention also encompasses a method of treating cancer with an effective amount of a compound of the present invention or using a combination of an effective amount of a compound of the present invention with an effective amount of radiation; antibodies to cytotoxic t-lymphocyte antigen 4 (anti-CTLA4); antibodies to programmed death ligand 1 (anti-PDLI); antibodies to programmed cell death protein 1 (anti-PDl); indoleamine-2, 3-dioxygenase (IDO) inhibitors; tryptophan-2, 3-dioxygenase (TDO) inhibitors; and antimetabolites.
  • anti-CTLA4 cytotoxic t-lymphocyte antigen 4
  • anti-PDLI antibodies to programmed death ligand 1
  • anti-PDl antibodies to programmed cell death protein 1
  • tryptophan-2 3-dioxygenase (TDO) inhibitors
  • antimetabolites antimetabolites.
  • These antibodies can be selected from, but not limited to, MDX-010 (ipilimumab, Bristol-Myers Squibb), CP- 675,206 (tremelimumab, Pfizer), MPDL3280A (Roche), MDX-1106 (nivolumab, Bristol- Myers Squibb), labrolizumab (Merck), and pembrolizumab (KEYTRUDA ® , Merck).
  • the cancer treated is selected from the group consisting of breast cancers, cervical cancers, colorectal cancers, endometrial cancers, glioblastomas, head and neck cancers, kidney cancers, liver cancers, lung cancers, medulloblastomas, ovarian cancers, pancreatic cancers, prostate cancers, skin cancers and urinary tract cancers.
  • IB treating cancer and/or generating a memory immune response comprising administering a compound of Formula I.
  • the term "treating a prostaglandin E2 mediated disease or condition” means treating or preventing any chronic disease or condition that is advantageously treated or prevented selective EP4 antagonists.
  • the term includes the relief of pain, fever and inflammation of a variety of conditions including rheumatic fever, symptoms associated with influenza or other viral infections, common cold, low back pain, neck pain, dysmenorrhea, headache, migraine, toothache, sprains and strains, myositis, neuralgia, synovitis, arthritis, including rheumatoid arthritis, degenerative joint diseases (osteoarthritis), gout, ankylosing spondylitis, bursitis, burns, injuries, and pain and inflammation following surgical procedures.
  • such a compound may inhibit cellular neoplastic transformations and metastatic tumor growth and hence can be used in the treatment and/or prevention of cancer.
  • the term "Effective amount” or “treatment-effective amount” refers to an amount that is effective for treating a prostaglandin E2 mediated disease or condition or a cancer as noted through clinical testing and evaluation, patient observation, and/or the like.
  • An “effective amount” can further designate an amount that causes a detectable change in biological or chemical activity. The detectable changes may be detected and/or further quantified by one skilled in the art for the relevant mechanism or process.
  • an “effective amount” can designate an amount that maintains a desired physiological state, i.e., reduces or prevents significant decline and/or promotes improvement in the condition.
  • An “effective amount” can further refer to a therapeutically effective amount.
  • cancer may include cancers that are the result of genetically inherited mutations.
  • examples of such cancers include, but are not limited to, breast cancers, cancers which can be related to Li-Fraumeni syndrome, for example, childhood sarcomas, leukemias and brain cancers, cancers which can be related to Lynch syndrome, for example, colon cancers, bile duct cancers, brain cancers, endometrial cancers, kidney cancers, ovarian cancers, pancreatic cancers, small intestinal cancers, stomach cancers and ureter cancers, lung cancers, melanomas, prostate cancers, retinoblastomas, thyroid cancers and uterine cancers.
  • cancer can be the result of acquired mutations, for example, mutations resulting from diet, environment and/or lifestyle, or somatic mutations.
  • cancers may include, but are not limited to, adrenal cancer, adrenal cortex cancer, bladder cancer, brain cancer, primary brain cancer, glioma, glioblastoma, breast cancer, cervical cancer, colon cancer (non-limiting examples include colorectal carcinomas such as colon adenocarcinoma and colon adoma), endometrial cancer, epidermal cancer, esophageal cancer, gall bladder cancer, genitourinary cancer, head or neck cancer, kidney cancer, liver cancer, lung cancer (non-limiting examples include adenocarcinoma, small cell lung cancer and non-small cell lung cancer), lymphomas (non-limiting examples include B-cell lymphoma, T-cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma), melanoma, malignant melanoma,
  • Compounds of Formula I can also be used in combination with one or more chemotherapeutic agents such as:
  • an antiestrogen ii. an antiestrogen, an anti-androgen (especially in the case of prostate cancer) or a gonadorelin agonist
  • a topoisomerase I inhibitor or a topoisomerase II inhibitor iv. a microtubule active agent, an alkylating agent, an antineoplastic antimetabolite or a platin compound,
  • a compound targeting/decreasing a protein or lipid kinase activity or a protein or lipid phosphatase activity, a further anti-angiogenic compound or a compound which induces cell differentiation processes v. a compound targeting/decreasing a protein or lipid kinase activity or a protein or lipid phosphatase activity, a further anti-angiogenic compound or a compound which induces cell differentiation processes
  • bradykinin I receptor or an angiotensin II antagonist
  • a cyclooxygenase inhibitor a bisphosphonate, a rapamycin derivative such as everolimus, a heparanase inhibitor (prevents heparan sulphate degradation), e.g. PI 88, a biological response modifier, preferably a lymphokine or interferons, e.g. interferon if, an ubiquitination inhibitor, or an inhibitor which blocks anti-apoptotic pathways,
  • an inhibitor of Ras oncogenic isoforms e. g. H-Ras, K-Ras or N-Ras, or a farnesyl transferase inhibitor, e.g. L-744, 832 or DK8G557,
  • telomere inhibitor e.g. telomestatin
  • a protease inhibitor a matrix metalloproteinase inhibitor, a methionine aminopeptidase inhibitor, e.g. bengamide or a derivative thereof, or a proteosome inhibitor, e.g. PS 341,
  • xi. histone deacetylase inhibitors e.g. Vorinostat, MG0103 or MS275, and
  • compositions of the present invention comprise a compound of Formula I as an active ingredient or a pharmaceutically acceptable salt, thereof, and may also contain a pharmaceutically acceptable carrier and optionally other therapeutic ingredients.
  • pharmaceutically acceptable salts refers to salts prepared from pharmaceutically acceptable non-toxic bases including inorganic bases and organic bases. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N- dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
  • basic ion exchange resins such as argin
  • the EP4 antagonist, antibody and/or anti-metabolite can be administered to subjects by any suitable route, including orally (inclusive of administration via the oral cavity and further including administration via an orogastric feeding tube), intraperitoneally, parenterally, by inhalation spray, topically (i.e., both skin and mucosal surfaces, including airway surfaces), transdermally, rectally, nasally (including a nasogastric feeding tube), sublingually, buccally, vaginally or via an implanted reservoir.
  • suitable route including orally (inclusive of administration via the oral cavity and further including administration via an orogastric feeding tube), intraperitoneally, parenterally, by inhalation spray, topically (i.e., both skin and mucosal surfaces, including airway surfaces), transdermally, rectally, nasally (including a nasogastric feeding tube), sublingually, buccally, vaginally or via an implanted reservoir.
  • parenteral includes subcutaneous, intramuscular, intradermal, intravenous, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the EP4 antagonist, antibody and/or anti-metabolite is administered orally.
  • the EP4 antagonist, antibody and/or antimetabolite is administered intravenously.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the technique described in the U.S. Patent 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for control release.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredients is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • an oil medium for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethyl-cellulose, methylcellulose, hydroxypropylmethy-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl, p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • preservatives for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • flavoring agents such as sucrose, saccharin or aspartame.
  • sweetening agents such as sucrose, saccharin or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium EDTA
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium EDTA, sodium bicarbonate, sodium bicarbonate
  • the pharmaceutical compositions of the invention may also be in the form of an oil-in- water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavouring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • Suitable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the compounds of the invention can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an I atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1, 1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane.
  • the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
  • the pressurized container, pump, spray, atomizer, or nebuliser contains a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • the drug product Prior to use in a dry powder or suspension formulation, the drug product is micronized to a size suitable for delivery by inhalation (typically less than 5 microns).
  • This may be achieved by any appropriate comminuting method, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.
  • Capsules (made, for example, from gelatin or HPMC), blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound of the invention, a suitable powder base such as lactose or starch and a performance modifier such as l-leucine, mannitol, or magnesium stearate.
  • the lactose may be anhydrous or in the form of the monohydrate, preferably the latter.
  • Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose and trehalose.
  • a suitable solution formulation for use in an atomizer using electrohydrodynamics to produce a fine mist may contain from log to 20 mg of the compound of the invention per actuation and the actuation volume may vary from 11 to 1001.
  • a typical formulation may comprise a compound of formula (1), propylene glycol, sterile water, ethanol and sodium chloride.
  • Alternative solvents which may be used instead of propylene glycol include glycerol and polyethylene glycol.
  • Suitable flavors, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium may be added to those formulations of the invention intended for inhaled/intranasal administration.
  • Formulations for inhaled/intranasal administration may be formulated to be immediate and/or modified release using, for example, poly(DL-lactic-coglycolic acid (PGLA).
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • the dosage unit is determined by means of a valve which delivers a metered amount.
  • Units in accordance with the invention are typically arranged to administer a metered dose or "puff" containing from 1 fig to 10 mg of the compound of formula (I).
  • the overall daily dose will typically be in the range 1 lag to 10 mg which may be administered in a single dose or, more usually, as divided doses throughout the day.
  • Compounds of Formula I may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • creams, ointments, jellies, solutions or suspensions, etc., containing the compound of Formula I are employed.
  • topical application shall include mouth washes and gargles.
  • Dosage levels of the order of from about 0.01 mg to about 140 mg/kg of body weight per day are useful in the treatment of the above-indicated conditions, or alternatively about 0.5 mg to about 7 g per patient per day.
  • inflammation may be effectively treated by the administration of from about 0.01 to 50 mg of the compound per kilogram of body weight per day, or alternatively about 0.5 mg to about 3.5 g per patient per day, preferably 2.5 mg to 1 g per patient per day.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a formulation intended for the oral administration of humans may contain from 0.5 mg to 5 g of active agent compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95 percent of the total composition.
  • Dosage unit forms will generally contain between from about 1 mg to about 500 mg of an active ingredient, typically 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg, or 1000 mg.
  • the compounds of Formula I can be tested using the following assays to determine their prostanoid antagonist or agonist activity in vitro and in vivo and their selectivity.
  • the prostaglandin receptor activities demonstrated are DP, EP1, EP2, EP3, EP4, FP, IP and TP.
  • Prostanoid receptor cDNAs corresponding to full length coding sequences are subcloned into the appropriate sites of mammalian expression vectors and transfected into HEK 293(ebna) cells.
  • HEK 293(ebna) cells expressing the individual cDNAs are grown under selection and individual colonies are isolated after 2-3 weeks of growth using the cloning ring method and subsequently expanded into clonal cell lines.
  • Transfected HEK 293(ebna) cells are maintained in culture, harvested and membranes are prepared by differential centrifugation, following lysis of the cells in the presence of protease inhibitors, for use in receptor binding assays.
  • Prostanoid receptor binding assays for DPI, DP2 (CRTH2), EP1, EP2, EP3-III, EP4, FP, IP, and TP
  • 10 mM MES/KOH pH 6.0
  • EPs, FP and TP 10 mM HEPES/KOH
  • DPs and IP containing 1 mM EDTA, 2.5-30 mM divalent cation and the appropriate radioligand.
  • Synthetic compounds are added in dimethylsulfoxide which is kept constant at 1 % (v/v) in all incubations. The reaction is initiated by addition of membrane protein. Non-specific binding is determined in the presence of 10 mM of the corresponding non-radioactive prostanoid . Incubations are conducted for 60-90 min at room temperature or 30 °C and terminated by rapid filtration. Specific binding is calculated by subtracting non specific binding from total binding. The residual specific binding at each ligand concentration is calculated and expressed as a function of ligand concentration in order to construct sigmoidal concentration-response curves.
  • EP4 receptor binding assays were performed at MSD Pharma Service in Taiwan under the following assay conditions:
  • Incubation Buffer 10 mM MES, pH 6.0, 1 mM EDTA, 10 mM MgCI.
  • Non-Specific Ligand 10 mM Prostaglandin E 2 (PGE 2 )
  • second messenger responses are expressed as a function of ligand concentration and both EC50 values and the maximum response as compared to a PGE2 standard are calculated.
  • the ability of a ligand to inhibit an agonist response is determined by carrying out dose-response curves in the presence of PGE2 agonist at a concentration corresponding to its EC70.
  • IC50 values are calculated as the concentration of ligand required to inhibit 50% of the PGE2-induced activity.
  • Female Lewis rats (body weight ⁇ 146-170 g) are weighed, ear marked, and assigned to groups (a negative control group in which arthritis was not induced, a vehicle control group, a positive control group administered indomethacin at a total daily dose of 1 mg/kg and four groups administered with a test compound at total daily doses of 0.10-3.0 mg/kg) such that the body weights were equivalent within each group.
  • Six groups of 10 rats each are injected into a hind paw with 0.5 mg of Mycobacterium butyricum in 0.1 mL of light mineral oil (adjuvant), and a negative control group of 10 rats was not injected with adjuvant.
  • Body weights, contralateral paw volumes (determined by mercury displacement plethysmography) and lateral radiographs (obtained under Ketamine and Xylazine anesthesia) are determined before (day -1) and 21 days following adjuvant injection, and primary paw volumes are determined before (day -1) and on days 4 and 21 following adjuvant injection.
  • the rats are anesthetized with an intramuscular injection of 0.03 - 0.1 mL of a combination of Ketamine (87 mg/kg) and Xylazine (13 mg/kg) for radiographs and injection of adjuvant.
  • the radiographs are made of both hind paws on day 0 and day 21 using the Faxitron (45 kVp, 30 seconds) and Kodak X-OMAT TL film, and are developed in an automatic processor. Radiographs are evaluated for changes in the soft and hard tissues by an investigator who was blinded to experimental treatment. The following radiographic changes are graded numerically according to severity: increased soft issue volume (0-4), narrowing or widening of joint spaces (0-5) subchondral erosion (0-3), periosteal reaction (0-4), osteolysis (0- 4) subluxation (0-3), and degenerative joint changes (0-3). Specific criteria are used to establish the numerical grade of severity for each radiographic change. The maximum possible score per foot was 26.
  • test compound at total daily doses of 0.1, 0.3, 1, and 3 mg/kg/day, indomethacin at a total daily dose of 1 mg/kg/day, or vehicle (0.5% methocel in sterile water) are administered per os b.i.d. beginning post injection of adjuvant and continuing for 21 days.
  • the compounds are prepared weekly, refrigerated in the dark until used, and vortex mixed immediately prior to administration.
  • the assays described in the article by Spranger et al can be used to evaluate the synergistic effects of the compounds of the present invention in combination with an effective amount of antibodies to cytotoxic t-lymphocyte antigen 4 (anti-CTLA4); antibodies to programmed death ligand 1 (anti-PDLI); antibodies to programmed cell death protein 1 (anti-PDl); indoleamine-2, 3-dioxygenase (IDO) inhibitors; tryptophan-2, 3-dioxygenase (TDO) inhibitors.
  • anti-CTLA4 cytotoxic t-lymphocyte antigen 4
  • anti-PDLI antibodies to programmed death ligand 1
  • anti-PDl antibodies to programmed cell death protein 1
  • IDO 3-dioxygenase
  • TDO 3-dioxygenase
  • Step 4 Methyl 4-(l-(7-(4-(trifluoromethyl)phenoxy)-lH-indole-l- carboxamido)cyclopropyl)benzoate
  • Step 5 4-(l-(7-(4-(trifluoromethyl)phenoxy)-lH-indole-l-carboxamido)cyclopropyl)benzoic acid
  • Step 2 methyl 4-(l-(7-(4-(trifluoromethyl)phenoxy)indoline-l-carboxamido) cyclopropyl) benzoate
  • Step 3 4-(l-(7-(4-(trifluoromethyl)phenoxy)indoline-l-carboxamido)cyclopropyl)benzoic acid
  • Step 1 4-nitrophenyl 7-(4-(trifluoromethyl)phenoxy)indoline-l-carboxylate
  • Step 2 methyl 4-((7-(4-(trifluoromethyl)phenoxy)indoline-l-carboxamido)methyl)benzoate
  • Step 3 4-((7-(4-(trifluoromethyl)phenoxy)indoline-l-carboxamido)methyl)benzoic acid
  • Step 2 l-methyl-4-(4-(trifluoromethyl)phenoxy)-lH-indole
  • Step 3 l-methyl-4-(4-(trifluoromethyl)phenoxy)-lH-indole-3-carboxylic acid
  • Step 4 methyl 4-(l-(l-methyl-4-(4-(trifluoromethyl)phenoxy)-lH-indole-3- carboxamido)cyclopropyl)benzoate
  • Step 5 4-(l-(l-methyl-4-(4-(trifluoromethyl)phenoxy)-lH-indole-3- carboxamido)cyclopropyl)benzoic acid
  • Step 6 methyl (S)-4-(l-(l-methyl-4-(4-(trifluoromethyl)phenoxy)-lH-indole-3- carboxamido)ethyl)benzoate
  • Step 7 (S)-4-(l-(l-methyl-4-(4-(trifluoromethyl)phenoxy)-lH-indole-3- carboxamido)ethyl)benzoic acid
  • Step 3 methyl 4-(l-(4-(4-(trifluoromethyl)phenoxy)-lH-indole-3- carboxamido)cyclopropyl)benzoate
  • Step 4 4-(l-(4-(4-(trifluoromethyl)phenoxy)-lH-indole-3-carboxamido)cyclopropyl)benzoic acid

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne de nouveaux composés antagonistes du récepteur EP4 représentés par la formule (I) ou des sels pharmaceutiquement acceptables de ceux-ci, qui peuvent être utiles pour le traitement du cancer ou de maladies inflammatoires.
PCT/US2019/016637 2018-02-05 2019-02-05 Composés hétérobicycliques en tant qu'antagonistes ep4 WO2019152982A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201980010302.8A CN111936138B (zh) 2018-02-05 2019-02-05 作为ep4受体拮抗剂的杂二环化合物

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862709882P 2018-02-05 2018-02-05
US62/709,882 2018-02-05

Publications (1)

Publication Number Publication Date
WO2019152982A1 true WO2019152982A1 (fr) 2019-08-08

Family

ID=67478959

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/016637 WO2019152982A1 (fr) 2018-02-05 2019-02-05 Composés hétérobicycliques en tant qu'antagonistes ep4

Country Status (2)

Country Link
CN (1) CN111936138B (fr)
WO (1) WO2019152982A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022102731A1 (fr) 2020-11-13 2022-05-19 小野薬品工業株式会社 Traitement du cancer par utilisation combinée d'un antagoniste d'ep4 et d'un inhibiteur de point de contrôle immunitaire

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170066773A1 (en) * 2015-08-26 2017-03-09 Blueprint Medicines Corporation Compounds and compositions useful for treating disorders related to ntrk

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102149384B (zh) * 2008-08-14 2014-08-20 南京奥昭生物科技有限公司 作为ep4受体拮抗剂的杂环酰胺衍生物
WO2010121382A1 (fr) * 2009-04-21 2010-10-28 Merck Frosst Canada Ltd. Procédé pour la fabrication de dérivés de cyclopropylamides de l'indole

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170066773A1 (en) * 2015-08-26 2017-03-09 Blueprint Medicines Corporation Compounds and compositions useful for treating disorders related to ntrk

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE PubChem Compound 17 March 2015 (2015-03-17), "4-(4-Cyclopropylbutyl)-3-methyl-1H-pyrrolo[3,4-c]pyridine | C15H20N2", XP055627883, retrieved from ncbi Database accession no. CID91363017 *
DATABASE PubChem Compound 17 March 2015 (2015-03-17), "4-Methoxy-2-methyl-1-[4-[2-(3-methyl-1H-pyrrolo[3,4-c]pyridin-4-yl)ethyl]phenyl]butan-1-one | C22H26N2O2", XP055627840, retrieved from ncbi Database accession no. CID91299494 *
DATABASE PubChem Compound 29 July 2006 (2006-07-29), "4-(4-Cyclopropylbutyl)-3-methyl-1H-pyrrolo[3,4-c]pyridine | C15H20N2", XP055627889, retrieved from ncbi Database accession no. CID7539974 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022102731A1 (fr) 2020-11-13 2022-05-19 小野薬品工業株式会社 Traitement du cancer par utilisation combinée d'un antagoniste d'ep4 et d'un inhibiteur de point de contrôle immunitaire

Also Published As

Publication number Publication date
CN111936138B (zh) 2024-03-08
CN111936138A (zh) 2020-11-13

Similar Documents

Publication Publication Date Title
CN112601750B (zh) Ptpn11(shp2)抑制剂
EP2320906B1 (fr) Dérivés d amides hétérocycliques en tant qu antagonistes du récepteur ep4
CA2822000C (fr) Indoles en tant qu'agents antiviraux contre le virus respiratoire syncytial
CA3161878A1 (fr) Agents de degradation d'irak et leurs utilisations
CN112823005A (zh) 吡咯并嘧啶itk抑制剂
EP2651933A1 (fr) Azabenzimidazoles en tant qu'agents antiviraux contre le virus respiratoire syncytial
BRPI0607915A2 (pt) composto e isÈmeros, sais, solvatos, formas quimicamente protegidas, e pró-drogas do mesmo, composição farmacêutica, e, uso de um composto ou sais farmaceuticamente aceitáveis do mesmo
JP2016504387A (ja) Wntシグナル伝達経路の3−(ベンゾイミダゾール−2−イル)−インダゾール阻害剤およびそれらの治療的使用
US20140024654A1 (en) Poly (adp-ribose) polymerase (parp) inhibitors
MXPA02008113A (es) Derivados de azaindol antiviral.
EP2864323B1 (fr) Dérivés de 1,3-dihydro-2h-benzimidazol-2-one substitués par des hétérocycles en tant qu'agents antiviraux du virus respiratoire syncytial
AU2019301679A1 (en) Dimeric immuno-modulatory compounds against cereblon-based mechanisms
JP2015518895A (ja) Egfr活性を調節するための化合物および組成物
KR20220161396A (ko) 치환된 피리디논-피리디닐 화합물의 제조, 조성물, 및 결정형
CA2973773C (fr) Derives de 2-phenyl-3h-imidazo[4,5-b]pyridine utiles comme inhibiteurs de l'activite de la tyrosine kinase mammifere ror11
AU2002211827B2 (en) Amino-substituted tetracyclic compounds useful as anti-inflammatory agents and pharmaceutical compositions comprising same
CA3106513A1 (fr) Composes heteroaromatiques utilises en tant qu'inhibiteurs de vanine
JP2022120130A (ja) ヘテロ二環式有機酸およびその塩類
CA3090485C (fr) Acides carboxyliques heterobicycliques pour traiter le cancer ou des maladies inflammatoires
WO2019152982A1 (fr) Composés hétérobicycliques en tant qu'antagonistes ep4
CA3170503A1 (fr) Agents de degradation de stat et leurs utilisations
WO2018048930A1 (fr) Inhibiteurs de la lysyl oxydase-like 2 et utilisations desdits inhibiteurs
CA3120036A1 (fr) Composes heteroaromatiques utilises en tant qu'inhibiteurs de vanine
EP2781508B9 (fr) Composés cycliques condensés contenant de l'azote pour utilisation en tant qu'antagonistes du crth2
CN101479263A (zh) 药用化合物

Legal Events

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

Ref document number: 19748426

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19748426

Country of ref document: EP

Kind code of ref document: A1