Title of Invention

IMMUNOSUPPRESSANT COMPOUNDS AND A PHARMACEUTICAL COMPOSITION COMPRISING THE SAME

Abstract The present invention relates to immunosuppressant, process for their production, their uses and pharmaceutical compositions containing them. The invention provides a novel class of compounds useful in the treatment or prevention of diseases or disorders mediated by lymphocyte interactions, particularly, particularly diseases associated with EDG receptor mediated signal transduction.
Full Text


IMMUNOSUPPRESSANT COMPOUNDS AND COMPOSITIONS
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of priority to U.S. Provisional Patent Application Number 60/471,931 (filed 19 May 2003) and U.S. Provisional Patent Application Number 60/562,183 (filed 14 April 2004). The full disclosures of these applications are incorporated herein by reference in their entirety and for all purposes.
BACKGROUND OF THE INVENTION
Field of the Invention
The invention provides a novel class of immunosuppressant compounds useful in the treatment or prevention of diseases or disorders mediated by lymphocyte interactions, particularly diseases associated with EDG receptor mediated signal transduction.
Background
EDG receptors belong to a family of closely related, lipid activated G-protein coupled receptors. EDG-1, EDG-3, EDG-5, EDG-6, and EDG-8 (also respectively termed S1P1, S1P3, S1P2, S1P4, and S1P5) are identified as receptors specific for sphingosine-1-phosphate (SIP). EDG2, EDG4, and EDG7 (also termed LPA1, LPA2, and LP A3, respectively) are receptors specific for lysophosphatidic (LPA). Among the SIP receptor isotypes, EDG-1, EDG-3 and EDG-5 are widely expressed in various tissues, whereas the expression of EDG-6 is confined largely to lymphoid tissues and platelets, and that of EDG-8 to the central nervous system. EDG receptors are responsible for signal transduction and are thought to play an important role in cell processes involving cell development, proliferation, maintenance, migration, differentiation, plasticity and apoptosis. Certain EDG receptors are associated with diseases mediated by lymphocyte interactions, for example, in transplantation rejection, autoimmune diseases, inflammatory diseases, infectious diseases and cancer. An alteration in EDG receptor activity contributes to the pathology and/or

symptomology of these diseases. Accordingly, molecules that themselves alter the activity of EDG receptors are useful as therapeutic agents in the treatment of such diseases.
SUMMARY OF THE INVENTION
This invention relates to compounds of Formula I:
in which:
n is 1 or 2;
A is chosen from -C(0)OR9, - OP(0)(OR9)2, - P(0)(OR9)2, -S(0)2OR9, -
P(0)(R9)OR9 and lif-tetrazol-5-yl; and R9 is chosen from hydrogen and C1-4alkyl;
X is a bond or is chosen from Ci-4alkylene, -X1OX2-, -XiNRioX2-, -
X,C(0)NR,oX2- -X,NR10C(O)X2- -X,S(0)X2-, -X,S(0)2X2-, -X{SXr- and C2. 9heteroarylene; wherein Xi and X2 are independently chosen from a bond and C1-4alkylene; Rio is chosen from hydrogen and Ci-6alkyl; and any heteroarylene of X is optionally substituted by a member of the group chosen from halo and Ci.6alkyl;
Y is a fused 5,6 or 6,6 hetero bicyclic ring system consisting of at least one
aromatic ring, wherein said fused bicyclic ring system of Y can be optionally substituted with 1 to 3 radicals chosen from halo, hydroxy, cyano, nitro, C1-4alkyl, C1-4alkoxy, halo-substituted C1-4alkyl and halo-substituted C1-4alkoxy;
Ri is chosen from Ce-ioaryl and C2-9heteroaryl; wherein any aryl or heteroaryl
of Ri is optionally substituted by a radical chosen from C6-ioarylCo-4alkyl, C2.9heteroarylCo-4alkyl, C3-8cycloalkylCo-4alkyl, C3-8heterocycloalkylCo^alkyl or Ci-6alkyl; wherein any aryl, heteroaryl, cycloalkyl or heterocycloalkyl group of Ri can be optionally substituted by 1 to 3 radicals chosen from halo, C^alkyl, Ci-ealkoxy, halo-substituted-Ci-6alkyl and halo-substituted-C1-4alkoxy; and any alkyl group of Ri can optionally have a methylene replaced by an atom or group chosen from -S-, -S(O)-, -S(0)2-, -NR10- and -O-; wherein Rio is chosen from hydrogen or Chalky!;

R2, R3, R5, R R4 is chosen from hydrogen and C1-4alkyl; or R7 and either R2, R4 or R5 together with the atoms to which R2, R4, R5 and R7 are attached forms a 4 to 7 member ring; wherein said 4 to 7 member ring is saturated or partially unsaturated; and the N-oxide derivatives, prodrug derivatives, protected derivatives, individual isomers and mixtures of isomers thereof; and the pharmaceutically acceptable salts and solvates (e.g. hydrates) of such compounds.
A second aspect of the invention is a pharmaceutical composition which contains a
compound of Formula I or an N-oxide derivative, individual isomer or mixture of isomers
thereof, or a pharmaceutically acceptable salt thereof, in admixture with one or more suitable
excipients.
A third aspect of the invention is a method for treating a disease in an animal in
which alteration of EDG receptor mediated signal transduction can prevent, inhibit or
ameliorate the pathology and/or symptomology of the disease, which method comprises
administering to the animal a therapeutically effective amount of a compound of Formula I
or a N-oxide derivative, individual isomer or mixture of isomers thereof; or a
pharmaceutically acceptable salt thereof.
A fourth aspect of the invention is the use of a compound of Formula I in the manufacture of a medicament for treating a disease in an animal in which alteration of EDG receptor mediated signal transduction contributes to the pathology and/or symptomology of the disease.
A fifth aspect of the invention is a process for preparing compounds of Formula I and the N-oxide derivatives, prodrug derivatives, protected derivatives, individual isomers and mixtures of isomers thereof; and the pharmaceutically acceptable salts thereof.
DESCRIPTION OF THE PREFERRED EMBODIMENTS
The invention provides compounds that are useful in the treatment and/or prevention of diseases or disorders mediated by lymphocyte interactions. Also provided are methods for treating such diseases or disorders.

Definitions
In this specification, unless otherwise defined:
"Alkyl" as a group and as a structural element of other groups, for example halo-substituted-alkyl, alkoxy, acyl, alkylthio, alkylsulfonyl and alkylsulfinyl, can be either straight-chained or branched. "Alkenyl" as a group and as a structural element of other groups contains one or more carbon-carbon double bonds, and can be either straight-chain, or branched. Any double bonds can be in the cis- or trans- configuration. "Alkynyl" as a group and as structural element of other groups and compounds contains at least one C ^C triple bond and can also contain one or more C=C double bonds, and can, so far as possible, be either straight-chain or branched. Any cycloalkyl group, alone or as a structural element of other groups can contain from 3 to 8 carbon atoms, preferably from 3 to 6 carbon atoms. "Alkylene" and "alkenylene" are divalent radicals derived from "alkyl" and "alkenyl" groups, respectively. In this application, any alkyl group of Rl can be optionally interrupted by a member of the group selected from -S-, -S(O)-, -S(0)2-, -NR20- and -0- (wherein R20 is hydrogen or Ci.6alkyl). These groups include -CH2-O-CH2-, -CH2-S(0)2-CH2-, -(CH2)2-NR20-CH2- -CH2-0-(CH2)2-, and the like.
"Aryl" means a monocyclic or fused bicyclic aromatic ring assembly containing six to ten ring carbon atoms. For example, C6-i2aryl can be phenyl, biphenyl or naphthyl, preferably phenyl. A fused bicyclic ring can be partially saturated, for example, 1,2,3,4-tetrahydro-naphthalene, and the like. "Arylene" means a divalent radical derived from an aryl group. For example, arylene as used in this application can be phenylene, biphenylene, naphthylene and the like.
"Halo" or "halogen" means F, CI, Br or I, preferably F or CI. Halo-substituted alkyl groups and compounds can be partially halogenated or perhalogenated, whereby in the case of multiple halogenation, the halogen substituents can be identical or different. A preferred perhalogenated alkyl group is for example trifluoromethyl or trifluoromethoxy.
"Heteroaryl" means aryl, as defined in this application, with the addition of at least one heteroatom moiety selected from N, O or S, and each ring is comprised of 5 to 6 ring atoms, unless otherwise stated. For example, C2heteroaryl includes oxadiazole, triazole, and the like. Cgheteroaryl includes quinoline, 1,2,3,4-tetrahydro-quinoline, and the like. C2. 9heteroaryl as used in this application includes thienyl, pyridinyl, furanyl, isoxazolyl,

benzoxazolyl or benzo[l,3]dioxolyl, preferably thienyl, furanyl or pyridinyl. "Heteroarylene" means heteroaryl, as defined in this application, provided that the ring assembly comprises a divalent radical. A fused bicyclic heteroaryl ring system can be partially saturated, for example, 2,3-dihydro-lH-isoindole, 1,2,3,4-tetrahydro-quinoline, and the like.
As used in the present invention, an EDG-1 selective compound (agent or modulator) has a specificity that is selective for EDG-1 over EDG-3 and over one or more of EDG-5, EDG-6, and EDG-8. As used herein, selectivity for one EDG receptor (a "selective receptor") over another EDG receptor (a "non-selective receptor") means that the compound has a much higher potency in inducing activities mediated by the selective EDG receptor (e.g., EDG-1) than that for the non-selective SIP-specific EDG receptor. If measured in a GTP-yS binding assay (as described in the Example below), an EDG-1 selective compound typically has an EC50 (effective concentration that causes 50% of the maximum response) for a selective receptor (EDG-1) that is at least 5,10,25, 50,100, 500, or 1000 fold lower than its EC50 for a non-selective receptor (e.g., one or more of EDG-3, EDG-5, EDG-6, and EDG-8).
Detailed Description of the Invention
The invention provides compounds that are useful for treating or preventing diseases or disorders that are mediated by lymphocyte interactions. In one embodiment, for compounds of Formula I, Ri is phenyl, naphthyl, furanyl or thienyl optionally substituted by Ce-ioarylCo-4alkyl, C2-9heteroarylC0-4aIkyl, C3-gcycloalkylCo-4alkyl, C3-8heterocycloalkylCo-4alkyl or d_ $alkyl; wherein any aryl, heteroaryl, cycloalkyl or heterocycloalkyl group of Ri can be optionally substituted by one to five radicals chosen from halo, Ci-ealkyl, Ci-ealkoxy, halo-substituted-Ci-ealkyl and halo-substituted-Ci-6alkoxy; and any alkyl group of Ri can optionally have a methylene replaced by an atom or group chosen from -S-, -S(O)-, -S(0)2-, -NRio- and -0-; wherein Rio is hydrogen or Ci-ealkyl. In another embodiment, Y is chosen from:



















carboxylic acid, l»[2^2-Trifluoromethyl-biphenyl-4-yl)-lH-benzoimidazol-5-ylmethyl]-azetidine-3-carboxylic acid, 3- {[3-Methyl-2-(2-trifluoromethyl-biphenyl-4-yl)-3H-benzoimidazol-5-ylmethyl]-amino} -propionic acid, 3- {[ 1 -Methyl-2-(2-trifluoromethyl-biphenyl-4-yl)-lH-benzoimidazol-5-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-ylmethyl)-2,3-dihydro-lH-isoindol-5-ylmethyl]-amino}-propionic acid, 3- {[2-(2-Trifluoromethyl-biphenyl-4-ylamino)-benzooxazol-5-ylmethyl]-amino}-propionic acid, 3-({2-[Methyl-(2-trifluoromethyl-biphenyl-4-yl)-amino]-benzooxazol-5-ylmethyl}-amino)-propionic acid, 3- {[4-Oxo-2-(2-trifluoromethyl-biphenyl-4-yl)-4H-chromen-7-ylmethyl]-amino}-propionic acid, 3-{[4-Oxo-2-(2-trifluoromethyl-biphenyl-4-yl)-4H-chromen-6-ylmethyl]-amino}-propionic acid and l-[4-Oxo-2-(2-trifluoromethyl-biphenyl-4-yl)-4H-chromen-6-ylmethyl]-azetidine-3-carboxylicacid( Further preferred compounds are also shown in the examples and table 1, infra.
The invention provides forms of the compound that have the hydroxyl or amine group present in a protected form; these function as prodrugs. Prodrugs are compounds that are converted into an active drug form after administration, through one or more chemical or biochemical transformations. Forms of the compounds of the present invention that are readily converted into the claimed compound under physiological conditions are prodrugs of the claimed compounds and are within the scope of the present invention. Examples of prodrugs include forms where a hydroxyl group is acylated to form a relatively labile ester such as an acetate ester, and forms where an amine group is acylated with the carboxylate group of glycine or an L-amino acid such as serine, forming an amide bond that is particularly susceptible to hydrolysis by common metabolic enzymes.
Compounds of Formula I can exist in free form or in salt form, e.g. addition salts with inorganic or organic acids. Where hydroxyl groups are present, these groups can also be present in salt form, e.g. an ammonium salt or salts with metals such as lithium, sodium, potassium, calcium, zinc or magnesium, or a mixture thereof. Compounds of Formula I and their salts in hydrate or solvate form are also part of the invention.
When the compounds of Formula I have asymmetric centers in the molecule, various optical isomers are obtained. The present invention also encompasses enantiomers, racemates, diastereoisomers and mixtures thereof. Moreover, when the compounds of Formula I include geometric isomers, the present invention embraces cis-compounds, trans-

compounds and mixtures thereof. Similar considerations apply in relation to starting materials exhibiting asymmetric carbon atoms or unsaturated bonds as mentioned above.
Methods and Pharmaceutical Compositions for Treating Immunomodulatory Conditions
The compounds of Formula I in free form or in pharmaceutical^ acceptable salt form, exhibit valuable pharmacological properties, e.g. lymphocyte recirculation modulating properties, for example, as indicated by the in vitro and in vivo tests of Example 6 and are therefore indicated for therapy. Compounds of Formula I preferably show an EC50 in the range of 1 x 10"n to 1 x 10"5 M, preferably less than 50nM. The compounds exhibit selectivity for one or more EDG/S1P receptors, preferably EDG-1/S1P-1. EDG-1/S1P-1 selective modulators of the present invention can be identified by assaying a compound's binding to EDG-1/S1P-1 and one or more of the other EDG/S1P receptors (e.g., EDG-3/S1P-3, EDG-5/S1P-2, EDG-6/S1P-4, and EDG-8/S1P-5). An EDG-1/S1P-1 selective modulator usually has an EC50 for the EDG-1/S1P-1 receptor in the range of 1 x lO4^11 to 1 x 10"5 M, preferably less than 50 nM, more preferably less than 5 nM. It also has an EC50 for one or more of the other EDG/S1P receptors that is at least 5,10, 25, 50,100, 500, or 1000 fold higher than its EC50 for EDG-1/S1P-1. Thus, some of the EDG-1/S1P-1 modulatory compounds will have an EC50 for EDG-1/S1P-1 that is less than 5 nM while their EC50 for one or more of the other EDG/S1P receptors are at least 100 nM or higher. Other than assaying binding activity to the EDG/S1P receptors, EDG-1/S1P-1 selective agents can also be identified by examining a test agent's ability to modify a cellular process or activity mediated by an EDG/S1P receptor.
The compounds of formula I are, therefore, useful in the treatment and/or prevention of diseases or disorders mediated by lymphocytes interactions, for example in transplantation, such as acute or chronic rejection of cell, tissue or organ alio- or xenografts or delayed graft function, graft versus host disease, autoimmune diseases, e.g. rheumatoid arthritis, systemic lupus erythematosus, hashimoto's thyroidis, multiple sclerosis, myasthenia gravis, diabetes type I or II and the disorders associated therewith, vasculitis, pernicious anemia, Sjoegren syndrome, uveitis, psoriasis, Graves ophthalmopathy, alopecia areata and others, allergic diseases, e.g. allergic asthma, atopic dermatitis, allergic rhinitis/conjunctivitis, allergic contact dermatitis, inflammatory diseases optionally with

underlying aberrant reactions, e.g. inflammatory bowel disease, Crohn's disease or ulcerative colitis, intrinsic asthma, inflammatory lung injury, inflammatory liver injury, inflammatory glomerular injury, atherosclerosis, osteoarthritis, irritant contact dermatitis and further eczematous dermatitises, seborrhoeic dermatitis, cutaneous manifestations of immunologically-mediated disorders, inflammatory eye disease, keratoconjunctivitis, myocarditis or hepatitis, ischemia/reperfusion injury, e.g. myocardial infarction, stroke, gut ischemia, renal failure or hemorrhage shock, traumatic shock, T cell lymphomas or T cell leukemias, infectious diseases, e.g. toxic shock (e.g. superantigen induced), septic shock, adult respiratory distress syndrome or viral infections, e.g. AIDS, viral hepatitis, chronic bacterial infection, or senile dementia. Examples of cell, tissue or solid organ transplants include e.g. pancreatic islets, stem cells, bone marrow, corneal tissue, neuronal tissue, heart, lung, combined heart-lung, kidney, liver, bowel, pancreas, trachea or oesophagus. For the above uses the required dosage will of course vary depending on the mode of administration, the particular condition to be treated and the effect desired.
Furthermore, the compounds of formula I are useful in cancer chemotherapy, particularly for cancer chemotherapy of solid tumors, e.g. breast cancer, or as an anti-angiogenic agent.
The required dosage will of course vary depending on the mode of administration, the particular condition to be treated and the effect desired. In general, satisfactory results are indicated to be obtained systemically at daily dosages of from about 0.03 to 2.5 mg/kg per body weight. An indicated daily dosage in the larger mammal, e.g. humans, is in the range from about 0.5 mg to about 100 mg, conveniently administered, for example, in divided doses up to four times a day or in retard form. Suitable unit dosage forms for oral administration comprise from ca. 1 to 50 mg active ingredient.
The compounds of Formula I can be administered by any conventional route, in particular enterally, for example, orally, e.g. in the form of tablets or capsules, or parenterally, for example, in the form of injectable solutions or suspensions, topically, e.g. in the form of lotions, gels, ointments or creams, or in a nasal or a suppository form. Pharmaceutical compositions comprising a compound of Formula I in free form or in pharmaceutically acceptable salt form in association with at least one pharmaceutical

acceptable carrier or diluent can be manufactured in conventional manner by mixing with a
pharmaceutically acceptable carrier or diluent.
The compounds of Formula I can be administered in free form or in
pharmaceutically acceptable salt form, for example, as indicated above. Such salts can be
prepared in a conventional manner and exhibit the same order of activity as the free
compounds.
In accordance with the foregoing the present invention further provides: LI A method for preventing or treating disorders or diseases mediated by
lymphocytes, e.g. such as indicated above, in a subject in need of such treatment, which
method comprises administering to said subject an effective amount of a compound of
formula I or a pharmaceutically acceptable salt thereof;
A method for preventing or treating acute or chronic transplant rejection or T-cell mediated inflammatory or autoimmune diseases, e.g. as indicated above, in a subject in need of such treatment, which method comprises administering to said subject an effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof;
A method for inhibiting or controlling deregulated angiogenesis, e.g. sphingosine-1-phosphate (SIP) mediated angiogenesis, in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof
A method for preventing or treating diseases mediated by a neo-angiogenesis process or associated with deregulated angiogenesis in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof.

A compound of formula I, in free form or in a pharmaceutically acceptable salt form for use as a pharmaceutical, e.g. in any of the methods as indicated under 1.1 to 1.4 above.
A pharmaceutical composition, e.g. for use in any of the methods as in 1.1 to 1.4 above comprising a compound of formula I in free form or pharmaceutically acceptable salt form in association with a pharmaceutically acceptable diluent or carrier therefor.

4. A compound of formula I or a pharmaceutically acceptable salt thereof for use in the preparation of a pharmaceutical composition for use in any of the method as in 1.1 to 1.4 above.
The compounds of formula I may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to, other drugs e.g. immunosuppressive or immunomodulating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of alio- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders, or a chemotherapeutic agent, e.g. a malignant cell antiproliferative agent. For example the compounds of formula I may be used in combination with a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a mTOR inhibitor, e.g. rapamycin, 40-O-(2-hydroxyethyl)-rapamycin, CCI779, ABT578 or AP23573; an ascomycin having immunosuppressive properties, e.g. ABT-281, ASM981, etc.; corticosteroids; cyclophosphamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenolic acid; mycophenolate mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; immunosuppressive monoclonal antibodies, e.g. monoclonal antibodies to leukocyte receptors, e.g. MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40. CD45, CD58, CD80, CD86 or their ligands; other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA-1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent.
By the term "chemotherapeutic agent" is meant any chemotherapeutic agent and it includes but is not limited to,
i. an aromatase inhibitor,
ii. an anti-estrogen, an anti-androgen (especially in the case of prostate cancer) or a gonadorelin agonist,
iii. a topoisomerase I inhibitor or a topoisomerase II inhibitor,
iv. a microtubule active agent, an alkylating agent, an antineoplastic antimetabolite or a platin compound,

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,
vi. a bradykinin 1 receptor or an angiotensin II antagonist,
vii. a cyclooxygenase inhibitor, a bisphosphonate, a histone deacetylase inhibitor, a heparanase inhibitor (prevents heparan sulphate degradation), e.g. PI-88, a biological response modifier, preferably a lymphokine or interferons, e.g. interferon D, an ubiquitination inhibitor, or an inhibitor which blocks anti-apoptotic pathways,
viii. 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 orDK8G557,
ix. a telomerase inhibitor, e.g. telomestatin,
x. 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, and/or
xi. a mTOR inhibitor.
The term "aromatase inhibitor" as used herein relates to a compound which inhibits the estrogen production, i.e. the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to steroids, especially atamestane, exemestane and formestane and, in particular, non-steroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole and letrozole. A combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, e.g. breast tumors.
The term "anti-estrogen" as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level. The term includes, but is not limited to tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride. A combination of the invention comprising a chemotherapeutic agent which is an anti-estrogen is particularly useful for the treatment of estrogen receptor positive tumors, e.g. breast tumors.
The term "anti-androgen" as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide.

The term "gonadorelin agonist" as used herein includes, but is not limited to abarelix, goserelin and goserelin acetate.
The term "topoisomerase I inhibitor" as used herein includes, but is not limited to topotecan, irinotecan, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148 (compound Al in WO99/17804).
The term "topoisomerase II inhibitor" as used herein includes, but is not limited to the anthracyclines such as doxorubicin, daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxines etoposide and teniposide.
The term "microtubule active agent" relates to microtubule stabilizing and microtubule destabilizing agents including, but not limited tp taxanes, e.g. paclitaxel and docetaxel, vinca alkaloids, e.g., vinblastine, especially vinblastine sulfate, vincristine especially vincristine sulfate, and vinorelbine, discodermolides and epothilones and derivatives thereof, e.g. epothilone B or a derivative thereof.
The term "alkylating agent" as used herein includes, but is not limited to busulfan, chlorambucil, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel™).
The term "antineoplastic antimetabolite" includes, but is not limited to 5-fluorouracil, capecitabine, gemcitabine, cytarabine, fludarabine, thioguanine, methotrexate and edatrexate.
The term "platin compound" as used herein includes, but is not limited to carboplatin, cis-platin and oxaliplatin.
The term "compounds targeting/decreasing a protein or lipid kinase activity or further anti-angiogenic compounds" as used herein includes, but is not limited to protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, e.g. compounds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR, ErbB2, ErbB3, ErbB4 as homo- or heterodimers), the vascular endothelial growth factor family of receptor tyrosine kinases (VEGFR), the platelet-derived growth factor-receptors (PDGFR), the fibroblast growth factor-receptors (FGFR), the insulin-like growth factor receptor 1 (IGF-1R), the Trk receptor tyrosine kinase family, the Axl receptor tyrosine kinase family, the Ret receptor tyrosine

kinase, the Kit/SCFR receptor tyrosine kinase, members of the c-Abl family and their gene-fuion products (e.g. BCR-Abl), members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK, FAX, PDK or PI(3) kinase family, or of the PI(3)-kinase-related kinase family, and/or members of the cyclin-dependent kinase family (CDK) and anti-angiogenic compounds having another mechanism for their activity, e.g. unrelated to protein or lipid kinase inhibition.
Compounds which target, decrease or inhibit the activity of VEGFR are especially compounds, proteins or antibodies which inhibit the VEGF receptor tyrosine kinase, inhibit a VEGF receptor or bind to VEGF, and are in particular those compounds, proteins or monoclonal antibodies generically and specifically disclosed in WO 98/35958, e.g. l-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutical^ acceptable salt thereof, e.g. the succinate, in WO 00/27820, e.g. a N-aryl(thio) anthranilic acid amide derivative e.g. 2-[(4-pyridyl)methyl]amino-N-[3-methoxy-5-(trifluoromethyl)phenyl]benzamide or 2-[(l -oxido-4-pyridyl)methyl]amino-N-[3-trifluoromethylphenyl]benzamide, or in WO 00/09495, WO 00/59509, WO 98/11223, WO 00/27819 and EP 0 769 947; those as described by M. Prewett et al in Cancer Research 59 (1999) 5209-5218, by F. Yuan et al in Proc. Natl Acad. Sci. USA, vol. 93, pp. 14765-14770, Dec. 1996, by Z. Zhu et al in Cancer Res. 58, 1998, 3209-3214, and by J. Mordenti et al in Toxicologic Pathology, Vol. 27, no. 1, pp 14-21, 1999; in WO 00/37502 and WO 94/10202; Angiostatin™, described by M. S. O'Reilly et al, Cell 79, 1994, 315-328; Endostatin™, described by M. S. O'Reilly et al, Cell 88,1997, 277-285; anthranilic acid amides; ZD4190; ZD6474; SU5416; SU6668; or anti-VEGF antibodies or anti-VEGF receptor antibodies,e.g. RhuMab.
By antibody is meant intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
Compounds which target, decrease or inhibit the activity of the epidermal growth factor receptor family are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, e.g. EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, or which have a dual inhibiting effect on the ErbB and VEGF receptor kinase and are in particular those compounds, proteins or monoclonal antibodies generically and specifically disclosed in WO 97/02266, e.g. the

compound of ex. 39, or in EP 0 564 409, WO 99/03854, EP 0520722, EP 0 566 226, EP 0 787 722, EP 0 837 063, US 5,747,498, WO 98/10767, WO 97/30034, WO 97/49688, WO 97/38983 and, especially, WO 96/30347 (e.g. compound known as CP 358774), WO 96/33980 (e.g. compound ZD 1839) and WO 95/03283 (e.g. compound ZM105180) or PCT/EP02/08780; e.g. trastuzumab (HerpetinR), cetuximab, Iressa, OSI-774, CI-1033, EKB-569, GW-2016, ELI, E2.4, E2.5, E6.2, E6.4, E2.ll, E6.3 or E7.6.3.
Compounds which target, decrease or inhibit the activity of PDGFR are especially compounds which inhibit the PDGF receptor, e.g. a N-phenyl-2-pyrimidine-amine derivative, e.g. imatinib.
Compounds which target, decrease or inhibit the activity of c-Abl family members and their gene fusion products are, e.g. a N-phenyl-2-pyrimidine-amine derivative, e.g. imatinib; PD180970; AG957; or NSC 680410.
Compounds which target, decrease or inhibit the activity of protein kinase C, Raf, MEK, SRC, JAK, FAK and PDK family members, or PI(3) kinase or PI(3) kinase-related family members, and/or members of the cyclin-dependent kinase family (CDK) are especially those staurosporine derivatives disclosed in EP 0 296 110, e.g. midostaurin; examples of further compounds include e.g. UCN-01, safingol, BAY 43-9006, Bryostatin 1, Perifosine; Ilmofosine; RO 318220 and RO 320432; GO 6976; Isis 3521; or LY333531/LY379196.
Further anti-angiogenic compounds are e.g. thalidomide (THALOMED) and TNP-470.
Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are, e.g. inhibitors of phosphatase 1, phosphatase 2A, PTEN or CDC25, e.g. okadaic acid or a derivative thereof.
Compounds which induce cell differentiation processes are, e.g. retinoic acid, a-, y- or 5-tocopherol or a-, y: or 8-tocotrienol.
The term cyclooxygenase inhibitor as used herein includes, but is not limited to, e.g. celecoxib (CelebrexR), rofecoxib (VioxxR), etoricoxib, valdecoxib or a 5-alkyl-2-arylaminophenylacetic acid, e.g. 5-methyl-2-(2'-chloro-6,-fluoroanilino)phenyl acetic acid.
The term "histone deacetylase inhibitor" as used herein includes, but is not limited to MS-27-275, SAHA, pyroxamide, FR-901228 or valproic acid.

The term 'Insphosphonates" as used herein includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid.
The term "matrix metalloproteinase inhibitor" as used herein includes, but is not limited to collagen peptidomimetic and non-petidomimetic inhibitors, tetracycline derivatives, e.g. hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat, prinomastat, BMS-279251, BAY 12-9566, TAA211 or AAJ996.
The term "mTOR inhibitor" as used herein includes, but is not limited to rapamycin (sirolimus) or a derivative thereof, e.g. 32-deoxorapamycin, 16-pent-2-ynyloxy-32-deoxorapamycin, 16-pent-2-ynyloxy-32(S)-dihydro-rapamycin, 16-pent-2-ynyloxy-32(S)-dihydro-40-O-(2-hydroxyethyl)-rapamycin and, more preferably, 40-0-(2-hydroxy-ethyl)-rapamycin. Further examples of rapamycin derivatives include e.g. CCI779 or 40- [3-hydroxy-2-(hydroxymethyl)-2-methylpropanoate]-rapamycin or a pharmaceutically acceptable salt thereof, as disclosed in USP 5,362,718, ABT578 or 40-(tetrazolyl)-rapamycin, particularly 40-epi-(tetrazolyl)-rapamycin, e.g. as disclosed in WO 99/15530, or rapalogs as disclosed e.g. in WO 98/02441 and WO01/14387, e.g. AP23573.
Where the compounds of formula I are administered in conjunction with other immunosuppressive / immunomodulatory, anti-inflammatory or chemotherapeutic therapy, dosages of the co-administered immunosuppressant, immunomodulatory, anti-inflammatory or chemotherapeutic compound will of course vary depending on the type of co-drug employed, e.g. whether it is a steroid or a calcineurin inhibitor, on the specific drug employed, on the condition being treated and so forth.
In accordance with the foregoing the present invention provides in a yet further aspect:
A method as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective non-toxic amount of a compound of formula I and at least a second drug substance, e.g. an immunosuppressant, immunomodulatory, antiinflammatory or chemotherapeutic drug, e.g. as indicated above.
A pharmaceutical combination, e.g. a kit, comprising a) a first agent which is a compound of formula I as disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one co-agent, e.g. an immunosuppressant, immunomodulatory, anti-

inflammatory or chemotherapeutic drug, e.g. as disclosed above. The kit may comprise instructions for its administration.
The terms "co-administration" or "combined administration" or the like as utilized herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
The term "pharmaceutical combination" as used herein means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term "fixed combination" means that the active ingredients, e.g. a compound of formula I and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term "non-fixed combination" means that the active ingredients, e.g. a compound of formula I and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the 2 compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of 3 or more active ingredients.
Methods for Preparing Compounds of the Invention
The present invention also includes processes for the preparation of immunomodulatory compounds of the invention. In the reactions described, it can be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups can be used in accordance with standard practice, for example, see T.W. Greene and P. G. M. Wuts in "Protective Groups in Organic Chemistry", John Wiley and Sons, 1991.
Compounds of Formula I can be prepared by proceeding as in the following reaction schemes:
Reaction Scheme 1


wherein n, R4, R9, R12 and R13 are as defined in the Summary of the Invention and W is a halogen, trifluromethanesulfonate, or the like. The reaction initially proceeds in the presence of a catalyst (e.g., palladium acetate, palladium chloride, palladium bromide, palladium cyanide, palladium acetylacetonate, palladium bis(benzonitrile) dichloride, tris(dibenzylideneacetone)-dipalladium, and the like) and a ligand (e.g., phosphorous ligands, such as triphenyl phosphine, tri-t-butyl phosphine, 2-(di-t-butylphosphino)biphenyl, dicyclohexylphosphinobiphenyl, and the like) in a solvent (e.g., tetrahydrofuran, 1,4-dioxane, benzene, toluene, xylene, N,N-dimethylformide, N-methyl-pyrrolidinone, and the like) at a temperature of about 20 to about 140 °C and can take up to about 48 hours to complete. The bromination reaction is carried out in the presence of a brominating agent (e.g., N-bromosuccinimide, bromine, and the like) and a radical initiator (e.g., 2,2'-azobisisobutyronitrile, benzoyl peroxide, and the like). The animation with amino carboxylic esters proceeds in the presence of a base (e.g., sodium hydride, triethylamine, diisopropylethylamine, potassium carbonate, sodium carbonate, and the like). The subsequent hydrolysis of esters proceeds in the presence of an acid (e.g. trifluoroacetic acid, hydrochloric acid, and the like) or a base (e.g., lithium hydroxide, sodium hydroxide, potassium hydroxide, sodium carbonate, and the like).
Reaction Scheme 2










Prodrug derivatives of the compounds of the invention can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et aL, (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985). For example, appropriate prodrugs can be prepared by reacting a non-derivatized compound of the invention with a suitable carbamylating agent (e.g., 1,1-acyloxyalkylcarbanochloridate, para-nitrophenyl carbonate, or the like).
Protected derivatives of the compounds of the invention can be made by means known to those of ordinary skill in the art. A detailed description of techniques applicable to the creation of protecting groups and their removal can be found in T W. Greene, "Protecting Groups in Organic Chemistry", 3rd edition, John Wiley and Sons, Inc., 1999.
Compounds of the present invention can be conveniently prepared, or formed during the process of the invention, as solvates (e.g., hydrates). Hydrates of compounds of the present invention can be conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
Compounds of the invention can be prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to forma pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomers. While resolution of enantiomers can be carried out using covalent diastereomeric derivatives of the compounds of the invention, dissociable complexes are preferred (e.g., crystalline diastereomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities. The diastereomers can be separated by chromatography, or preferable, by separation/resolution techniques based upon differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization. A more detailed description of the techniques applicable to the resolution of stereoisomers of compounds from the their racemic mixture can be found in Jean Jacques, Andre Collet, Samuel H. Wilen, "Enantiomers, Racemates and Resolutions", John Wiley And Sons, Inc., 1981,

In summary, the compounds of Formula I can be made by a process, which involves:
reaction schemes 1,2, 3,4, 5 or 6; and
optionally converting a compound of the invention into a pharmaceutical^ acceptable salt;
optionally converting a salt form of a compound of the invention to a non-salt form;
optionally converting an unoxidized form of a compound of the invention into a pharmaceutically acceptable N-oxide;
optionally converting an N-oxide form of a compound of the invention to its unoxidized form;
optionally resolving an individual isomer of a compound of the invention from a mixture of isomers;
optionally converting a non-derivatized compound of the invention into a pharmaceutically acceptable prodrug derivative; and
(h) optionally converting a prodrug derivative of a compound of the invention to its non-derivatized form.
Insofar as the production of the starting materials is not particularly described, the compounds are known or can be prepared analogously to methods known in the art or as disclosed in the Examples hereinafter.
One of skill in the art will appreciate that the above transformations are only representative of methods for preparation of the compounds of the present invention, and that other well known methods can similarly be used.
EXAMPLES
The following examples provide detailed descriptions of the preparation of representative compounds and are offered to illustrate, but not to limit the present invention.

Example 1
3- ([2-(2-Trifluoromethvl-biphenvl-4-vlVbenzor6]thiophen-5-vimethvl1-aminol -propionic
acid

To a solution of 5-methylbenzo[fc]thiophene (1.0 g, 6.75 mmol) in anhydrous ether (17 mL) at -78 °C is added w-BuLi (5,1 mL of a 1.52 M solution in hexanes, 7.75 mmol). The reaction flask is then moved to a 0 °C bath and stirred for 2.5 hours. The mixture is cooled back to -78 °C, and trimethyl borate (1.51 mL, 13.5 mmol) is added neat. The mixture is allowed to warm up to room temperature overnight, and then treated with 2 N HC1 (10 mL). After 2 hours, the mixture is extracted with ether (5 x), and the combined organic solution is dried (MgSC>4) and concentrated. The crude product, 5-methylbenzo[6]-thiophenylboric acid, is used without further purification.
To a solution of 5-methylbenzo[6]thiophenylboric acid (0.84 g, 4.4 mmol) in ethanol (2 mL) and toluene (8 mL) is added 2-chloro-5-bromobenzenetrifluoride (1.14 g, 4.4 mmol) and tetrakis(triphenylphosphine)palladium(0) (0.254 g, 0.22 mmol), followed by the addition of a solution of sodium carbonate (1.86 g, 17.6 mmol) in water (8 mL). The mixture is stirred vigorously at 80°C for 4 hours, and then filtered through a pad of Celite, which is rinsed with hexanes. The filtrate is concentrated and purified by column chromatography (100% hexanes) to give 1.15 g (80%) of 2-(4-chloro-3-trifluoromehtylphenyl)-5-methylbenzo[6]thiophene as a white solid.
To a solution of 2-(4-chloro-3-trifluoromehtylphenyl)-5-methylbenzo[6]-thiophene (0.746 g, 2.28 mmol) in carbon tetrachloride (23 mL) is added Af-bromosuccinimide (0.447 g, 2.51 mmol) and 2, 2,-azobisisobuyronitrile (AIBN, 0.075 g, 0.46 mmol). The mixture is stirred at 90°C overnight, and then concentrated. The residue is passed through a pad of silica gel, which is further rinsed with hexanes. The combined organic solution is concentrated in vacuo. The resulting crude product, 5-bromomethyl-2-(4-chloro-3-

trifluromehtylphenyl)benzo[6]thiophene, is dissolved in DMF (2 mL) and added to apre-stirred suspension of p-alanine /-butyl ester hydrochloride (0.828 g, 4,56 mmol) and sodium hydride (0.365 g, 60% dispersion in mineral oil, 9.12 mmol) in DMF (4 mL). The mixture is stirred at room temperature for 4 hours, quenched with water (1 mL) and concentrated in vacuo. The resulting residue is purified by column chromatography (50% to 70% EtOAc/hexanes) to afford 0.79 g (73%) of 3-{[2-(4-chloro-3-
trifluoromethylphenyl)benzo[6]thiophene-5-ylmetyl]amino}propionic acid /-butyl ester as a yellow solid.
To a solution of the above chloride (0.79 g, 1.68 mmol) in THF (20 mL) is added phenyl boric acid (0.41 g, 3.36 mmol), potassium fluoride (0.39 g, 6.72 mmol), 2-(dicyclohexylphosphino)biphenyl (59 mg, 0.17 mmol) and palladium (II) acetate (19 mg, 0.084 mmol). The mixture is stirred at 60 °C under argon for 24 hours. After concentration, the residue is purified by column chromatography (50% to 70% EtOAc/hexanes) to afford 3-{[2-(2-trifluoromethyl-biphenyi-4-yl)benzo[6]thiophen-5-ylmethyl]amino}propionicacidr-butyl ester. The ester is hydrolyzed in TFA-CH2C12 (1:2 v%, 20 mL). The crude product is purified by the preparative LCMS to afford 0.55 g (67%) of 3-{[2-(2-trifluoromethyl-biphenyl-4-yl)benzo/Z)]thiophen-5-ylmethyl]amino}propionic acid, which is converted to the corresponding HC1 salt: lH NMR (400 MHz, CD3OD) 5 8.03 (d, 1 H), 7.94 (dd, 2 H), 7.92 (s, 1 H), 7.42-7.30 (m, 5 H), 7.29-7.21 (m, 2 H), 4.29 (s, 2 H), 3.25 (t, 2 H), 2.70 (t, 2 H); MS (ES) 456.1 (M+H+).
Example 2
3-(r2-(4-PiDeridin-l-vl-3-trifluoromethyl-phenyl)enzor6]thiophen--5-vlmethvl1-aminoi-
propionic acid

Piperidine (30 /iL, 2.0 mmol), Pd2dba3 (2.7 mg, 0.003 mmol), potassium f-butoxide (59 mg, 0.53 mmol) and l,3-bis-(2,6-diisopropyIphenyl)-4,5-dihydroimidazol-2-ylidene HC1

salt (2.6 mg, 0.006 mmol) are added sequentially to a solution of 3-{[2-(4-chloro-3-trifluoromethyl-phenyl)-benzo[6]thiophen-5-ylmethyl]-amino}-propionic acid tert-bntyl ester (72 mg, 0.15 mmol) in 1,4-dioxane (0.8 mL). The mixture is irradiated in a microwave at 100 °C for 1 hour. The reaction is quenched with water and the mixture is concentrated in vacuo. The residue is dissolved in THF and aqueous 2N NaOH (4 mL, 1:1 v/v), and stirred at 60 °C for 3 hours. It is concentrated and purified with preparative LCMS to afford 20 mg of 3-{[2-(4-piperidin-l-yl-3-trifluoromethyl-phenyl)-benzo[6]thiophen-5-ylmethyl]-amino}-propionic acid, which is converted to HC1 salt: *H NMR (400 MHz, CD3OD) 8 8.15-8.00 (m, 4 H), 7.87 (s, 1 H), 7.63 (d, 1 H), 7.57 (d, 1 H), 4.48 (s, 2 H), 3.44 (t, 2 H), 3.03 (t, 4 H), 2.89 (t, 2 H), 1.90-1.80 (m, 4 H), 1.75-1.65 (m, 2 H); MS (ES) 463.4 (M+H+).
Example 3
3-lf2-r2-Trifluoromethvl-biphenvl-4-vl)-thienof23-61pvridin-5-vlmethvll-amino>-propionic
acid

To a solution of 2-(2-trifluoromethyl-biphenyl-4-yl)-thieno[2,3-6]pyridine-5-carbaldehyde (32 mg, 0.083 mmol) in MeOH (2 mL) is added p-alanine HC1 salt (37 mg, 0.42 mmol) and triethylamine (23 uL, 0.16 mmol). The mixture is stirred at 50 °C for 20 minutes. Sodium borohydride (30 mg, 0.8 mmol) is added at room temperature and it is stirred for 10 minutes. The mixture is purified with preparative LCMS to give 22 mg of 3-{[2-(2-trifluoromethyl-biphenyl-4-yl).
acid, which is converted to HC1 salt: lH NMR (400 MHz, CD3OD) 6 8.56 (d, 1 H), 8.30 (d, 1 H), 8.08 (d, 1 H), 7.98 (dd, 1 H), 7.84 (s, 1 H), 7.42 (d, 1 H), 7.40-7.34 (m, 3 H), 7.30-7.24 (m, 2 H); MS (ES) 457.1 (M+H+).

Example 4
34r2-(2-Trifluoromethyl-biphenyl

Bromoacetaldehyde dimethyl acetal (1.6 mL, 0.01 mol) is added dropwise to a mixture of m-methylbenzenethiol (1.5 mL, 0.01 mol) and K2CO3 (1.66mg, 0.01 mol) in 20 mL acetone at room temperature. The reaction mixture is stirred for 16 hours and then filtered. The solid is washed with acetone, and the combined filtrate and washes are concentrated in vacuo. The residue is diluted with water and extracted with ether. The ether layer is washed with 0.5 M KOH, water, and brine, dried, filtered and concentrated in vacuo to give 2 g yellow oil.
A solution of above yellow oil in CH2CI2 (20 mL) is added dropwise to a solution of BF3 in ether (0.7 mL, 0.005 mol) in CH2CI2 (100 mL) at room temperature. The reaction mixture is stirred for 3 hours, treated with aqueous NaHC03 solution and stirred until both phase are clear. The CH2CI2 layer is separated, dried, filtered and concentrated in vacuo to give 0.45 g of an approximately 1:3 mixture of 4- and 6-methylJ>enzo[b]thiophene as a dark brown oil. Major isomer: !H NMR (DMSO-de): 6 7.78 (d, 1H), 7.76 (d, 1H), 7.64 (d, 1H), 7.39 (m, 1H), 7.17 (m, 1H), 2.43 (s, 3H).
To a solution of 6-methyl_benzo[b]thiophene (0.16 g, 0.001 mol) in 10 mL of anhydrous THF at -60 °C is added n-BuLi (0.8 mL, 0.0012 mol) dropwise via syringe. After stirring for 30 minutes triisopropyl borate (0.3 mL, 0.0012 mol) is added dropwise. The reaction mixture is allowed to warm to 0°C and then partitioned between 1.0N HC1 and EtOAc. The organic layer is separated, dried, filtered and concentrated to produce a white solid that is triturated from ether/hexane. Filtration provides 0.17 g of 6-Methyl-benzo[b]thiophene-2-boronic acid as white solid. MS m/z 193 [M+l]+.
To a slurry of 6-methyl-benzo[b]thiophene-2-boronic acid (0.19 g, 0.001 mol) in 10 mL of benzene is added 4-bromo-2-trifluoromethyl-biphenyl ( 0.3g, 0.001 mol). The




Example 5
3-(f2-(2-Trifluoromethvl-biDhcnvl^vn-23-dihvdro-lH-isoindol-5-vlmethvl1-
amino}-propionic acid

3,4-Bis-bromomethyl-benzoic acid methyl ester (0.32 g, 0.001 mol) and 4-bromo-3-trifluoromethyl-phenylamine (0.72 mL, 0.003 mol) are dissolved in anhydrous 30 mL of EtOH. The reaction mixture is heated to reflux for 3 hours, then cooled down to room temperature. After filtering, a white solid product is obtained. MS m/z 400 [M+l]+.
2-(4-Bromo-3-trifluoromethyl-phenyl)-2,3-dihydro-1 H-isoindole-5-carboxylic acid methyl ester (0.2 g, 0.0005 mol) is dissolved in 20 mL of anhydrous toluene, followed by adding 40 mg of tetrakis(triphenylphosphine-palladium(0). After bubbling N2 through the solution for 3 minutes, tributyl-phenyl-stannane (0.22 g, 0.0006 mol) is added to the solution. The reaction mixture is heated to reflux for 16 hours. After column (9:1 Hexane: EtOAc), a white solid product (0.18g) is obtained. MS m/z 398 [M+l]+.
2-(2-Trifluoromethvl-biphenvl-4"VlV2 To a solution of 2-(2-trifluoromethyl-biphenyl-4-yl)-2,3-dihydro-lH-isoindole-5-carbaldehyde (19 mg, 0.05 mmol) in 3 mL MeOH, 0-alanine (8.7mg, 0.1 mmol) and catalytic amount EtaN are added. The suspension is heated to 50 °C for 0.5 hours, followed by adding 2 mg of NaBH4. After pre-LC-MS, 8 mg of 3-{[2-(2-trifluoromethyl-biphenyl-4-yl)-2,3-dihydro-lH-isoindol-5-ylmethyl]-amino}-propionic acid is obtained as a white solid.

USm/z 441[M+1]+. 'H NMR (DMSO-C1-4: 6 7.08-7.26 (m, 9H), 6.80 (brs, 2H), 4.53 (s, 4H), 3.68 (s, 2H), 2.62 (t, 2H), 2.16 (t, 2H).
Example 6
3"{[2-(2-Trifluoromethvl-biphenvl-4-vl)-benzooxa2ol-5-vl-methvll-amino)-propionic acid

A solution of 2-amino-4-methylphenol (1 eq) and 4-chloro-3-trifluoromethyl-benzaldehyde (1 eq) in methanol (0.1 M) is heated at 50°C for 30 minutes. After concentration, the residue is dissolved in CH2CI2 (0.1 M) and treated with DDQ (1,05 eq). The resulting mixture is stirred at room temperature for 10 minutes. It is then diluted with CH2CI2 and washed with NaHCC>3 and brine. The organic layer is dried over Na2S04. After concentration, the desired product is purified by column chromatography (5% EtOAc/hexane) to give a white solid. MS: (ES*): 312.0 (M+l)+.
A mixture of 2-(4-chloro-3-trifluoromethyl-phenyl)-5-methyl-benzooxazole (1 eq), phenyl boronic acid (1.5 eq), Pd(OAc)2 (0.03 eq), phosphine ligand (0.06 eq) and KF (3 eq) in dry THF (0.5 M) is heated at 100 °C in microwave for 30 minutes. The resulting mixture is diluted with EtOAc and washed with brine. The organic layer is dried over Na2SC>4. After concentration, the residue is purified by column chromatography (5% EtOAc in hexane) to give the desired product as a white solid. MS: (ES4): 354.1 (M+l)+.
A mixture of 5-methyl-2-(2-trifiuoromethyl-biphenyl-4-yl)-benzooxazole (1 eq), NBS (1 eq) and AIBN (0.1 eq) in CCI4 (0.1 M) is refluxed for 5 hours. After concentration, the desired product is purified by column chromatography (10% EtOAc/hexane). MS: (ES+): 432.0 (M+l)+.
To a solution of p-alanine methyl ester hydrochloride salt (2 eq) in dry DMF (0.5 M) is added NaH (3.5 eq). After stirring at room temperature for 10 minutes, a solution of 5-bromomethyl-2-(2-trifluoromethyl-biphenyl-4-yl)-benzooxazole (1 eq) in dry DMF (1 M) is

added. The resulting mixture is stirred at room temperature for 2 hours. It is diluted with H20 and extracted with EtOAc. The organic solution is washed with brine and dried over Na2S04. After concentration, the residue is dissolve in MeOH (0.2 M) and treated with 2N LiOH solution (3 eq) for 10 hours. The final compound is purified by preparative LCMS to give3-{[2-(2-trifluoromethyl-biphenyl-4-yl)-benzooxazol-5-ylmethyl]-amino}-propionic acid; !H NMR (400 MHz, CD3OD) 5 2.74 (t, 7= 6.7 Hz, 2H), 3.30 (t, J= 6.8 Hz, 2H), 4.39 (s, 2H), 7.34 (m, 2H), 7.43 (m, 3H), 7.57 (m, 2H), 7.78 (d, /= 8.3 Hz, 1H), 7.94 (s, 1H), 8.43 (d,7=8.0Hz, lH),8.59(s, 1H). MS: (ES+): 441.3 (M+l)+.
Example 7
l-("2-f4-Isobutvl-3-trifluoromethvl-phenvl)-benzooxazol-6"Vlmethvl1-azetidine-3-carboxvlic
acid

A mixture of [2-(4-chloro-3-trifluoromethyl-phenyl)-benzooxazol-6-yl]-methanol (1 eq) and Pd(PBu'3)2 (0.05 eq) is treated with isobutyl zinc bromide in THF (0.5 M, 3 eq). The resulting mixture is heated at 100 °C in microwave for 30 minutes. The reaction mixture is diluted with aqueous HC1 (5%) and extracted with EtOAc. The organic solution is washed with brine and dried over Na2S04. After concentration, the residue is purified by flash column chromatography (30% EtOAc in hexane) to give the desired intermediate [2-(4-isobutyl-3-trifluoromethyl-phenyl)-benzooxazol-6-yl]-methanol. MS: (ES*): 350.1 (M+l)+.
To a solution of [2-(4-isobutyl-3-trifluoromethyl-phenyl)-benzooxazol-6-yl]-methanol (1 eq) in dioxane (0.2 M) is treated with MnC>2 (10 eq). The resulting mixture is refluxed for 20 minutes and filtered through celite. After concentration, the residue is redissolved in MeOH (0.2 M) and is added azetidine-3-carboxylic acid (2 eq) and Et3N (1.8 eq). The resulting mixture is heated at 50°C for 1 hour. After cooling to room temperature, NaBHaCN (3 eq) is added in portions. The final compound is purified by preparative LCMS. lH NMR (400 MHz, CD3OD) 5 8.49 (s, 1H), 8.38 (d, J- 8.4 Hz, 1H), 7.88 (s, 1H),

7.84 (d, y= 8.4 Hz, 1H), 7.66 (d, J= 8.4 Hz, 1H), 7,52 (d, /= 8.4 Hz, 1H), 4.57 (s, 2H), 4.34 (m, 4H), 3.70 (m, 1H), 2.76 (d, 7= 6.8 Hz, 2H), 2,03 (m, 1H), 0.95 (d, J = 7.2 Hz, 6H). MS:
(ES4): 433.2 (M+l)+.
5 Example 8
3- ([2-(2-Trifluoromethvl-biphenvl-4-vl)-benzofuran-5-vlmethvl1-amino) -propionic acid
To a solution oi 4-nyaroxy-j-ioaoDenzaiaenyae (l eq; ana i-cftloro-4-ethynyl-2-trifluoromehtylbenzene (1 eq) in DMF (0.2 M) is added copper(I) iodide (0.1 eq), dichlorobis(triphenylphosphine)palladium(II) (0.1 eq) and diisopropylethylamine (3 eq). The mixture is irradiated with microwave at 80 °C for 10 minutes. The product, 2-(4-chloro-3-trifluoromethyl-phenyl)-benzofuran-5-carbaldehyde, is purified with column chromatography.
To a solution of 2-(4-chloro-3-trifluoromethyl-phenyl)-benzofuran-5-carbaldehyde (1 eq) in CH3OH (0.2 M) is added p-alanine /-butyl ester (2.5 eq) and triethylamine (2 eq). The mixture is stirred at 50 °C for 30 minutes. Sodium borohydride (5 eq) is then added at room temperature and the mixture is stirred for 10 minutes. The product, 3-{[2-(4-chloro-3-trifluoromethyl-phenyl)-benzofuran-5-ylmethyl]-amino}-propionic acid /er/-butyl ester, is purified with column chromatography.
To a solution of 3-{[2-(4-chloro-3-trifluoromethyl-phenyl)-benzofuran-5-ylmethyl]-amino}-propionic acid /erf-butyl ester (1 eq) and phenyl boric acid (1.5 eq) in THF (0.5 M) is added palladium(II) acetate (0.1 eq), 2-(dicyclohexylphosphino)biphenyl (0.2 eq) and potassium fluoride (4.0 eq). The mixture is irradiated with microwave at 120 °C for 45 minutes. The product, 3-{[2-(2-trifluoromethyl-biphenyl-4-yl)-benzofuran-5-ylmethyl]-amino}-propionic acid fer/-butyl ester, is purified with column chromatography. The ester is hydrolyzed with TFA in CH2CI2 (1:2, v/v) at room temperature. It is purified with preparative LCMS to afford 3-{[2-(2-trifluoromethyl-biphenyl-4-yl)-benzofuran-5-

ylmethyl]-amino}-propionic acid, which is converted to HC1 salt: *H NMR (400 MHz, CD3OD) 8 8.29 (d, 1 H), 8.18 (dd, 1 H), 7.82 (d, 1 H), 7.71 (d, 1 H), 7.55-7.30 (m, 8 H), 4.36 (s, 2 H), 3.32 (t, 2 H), 2.77 (t, 2 H); MS (ES) 440.2 (M+H+).
Example 9
3-{[2-(2-Trifluoromethvl-biphenvl-4-vl)-benzothiazol-6-vljiiethvl1-amino}-
propionic acid

To a solution of/?-toluidine (0.44 g, 4.1 mmol) in 10 ml of CH2CI2 (pre-cooled to 0°C) are added Et3N (1.14 ml, 2 eq.) and 4-chloro-3-trifluoromethyl-berizoyl chloride (1 g, 4.1 mmol) in 5 ml of CH2CI2. The mixture is slowly warmed to room temperature and continued to stir at room temperature for an hour. The mixture is diluted with 50 ml of CH2CI2, washed with IN HC1 solution, and brine. The organic layer is separated, dried over MgS(>4, filtered, and concentrated. The residue is purified by column chromatography (EtOAc/Hexanes, 2:3) to give 1.25 g (97%) of 4-chloro-N-/?-tolyl-3-trifluoromethyl-benzamide.
4-chloro-N-/?-tolyl-3-trifluoromethyl-benzamide (1.0 g, 3.19 mmol), Lawesson's reagent (774 mg, 0.6 eq.) and toluene (2.5 mL) is mixed in a microwave vial. The mixture is heated to 120 °C for 1000 seconds using microwave irradiation. The mixture turned into clear solution. Ether (50 ml) is added to dilute the reaction mixture. The solution is then washed with brine, dried over MgSC>4, filtered and concentrated. The mixture is purified by column chromatography (EtOAc/Hexanes=5/95) to afford 970 mg (92%) of 4-chloro-N-p-tolyl-3-trifluoromethyl-thiobenzamide as a yellow solid.
To 2M aqueous solution K3Fe(CN)6 (4 mL, 8 mmol) (pre-heated to 90°C) is added dropwise to a suspension of 4-chlorO'N-p-tolyl-3-trifluoromethyl-thiobenzamide (660 mg, 2 mmol) in 2M NaOH (9 ml) and EtOH (3 ml). The mixture is heated at 90°C overnight. The







chromatography (EtOAc/Hexane, gradient) to give 43 mg of 3-{[3-chIoro-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[6]thiophen-5-ylmethyl]-amino}-propionic acid ten-butyl ester in 76% yield.
3-{[3-Chloro-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[6]thiophen-5-ylmethyl]-amino}-propionic acid /erf-butyl ester (43 mg, 0.079 mmol) is dissolved in TFA/CH2CI2 (1/1) (1 mL). The solution is stirred at room temperature for an hour. The mixture is concentrated and purified by reversed phase preparative LC/MS to give 23 mg of 3-{[3-
chloro-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[6]thiophen-5-ylmethyl]-amino}-propionic acid: lH NMR (CD3OD, 400 MHz) 8 8.10 (s, 1 H), 7.96-8.02 (m, 3 H), 7.54 (d, 1 H), 7.46 (d, 1 H), 7.35-7.37 (m, 3 H), 7.28-7.29 (m, 2 H), 4.37 (s, 2 H), 3.26(m, 2 H), 2.71 (m, 2 H), MS (ES+) 490.3 (M+H*).
By repeating the procedure described in the above examples, using appropriate starting materials, the following compounds of Formula I are obtained as identified in Table 1.






































Example 11 Compounds of Formula I Exhibit Biological Activity
A. In vitro: GPCR activation assay measuring GTP fy-35S1 binding to membranes prepared from CHO cells expressing human EDG receptors
EDG-1 (SIPi) GTP [y-35S] binding assay: Homogenized membranes are prepared from CHO cell clones stably expressing a human EDG-1 N-terminal c-myc tag. Cells are grown in suspension in two 850 cm2 roller bottles for three or fours days before harvesting. The cells are centrifuged down, washed once with cold PBS, and resuspended in ^20 ml of Buffer A (20 mM HEPES, pH 7.4,10 mM EDTA, EDTA-free complete protease inhibitor cocktail [1 tablet/25 ml]). The cell suspension is homogenized on ice, using a Polytron homogenizer at 30000 rpm at three intervals of 15 seconds each. The homogenate is first centrifuged at 2000 rpm on a tabletop low speed centrifuge for 10 minutes. The supernatant, after passing through a cell strainer, is then re-centrifuged at 50,000 x g for 25 minutes at 4°C. The pellet is resuspended into buffer B (15% glycerol, 20 mM HEPES, pH 7.4,0.1 mM EDTA, EDTA-free complete protease inhibitor cocktail [1 tablet/10 ml]). Protein concentration of the prep is determined using the BCA Protein Assay kit (Pierce) using BSA as standard. The membranes are aliquoted and kept frozen at -80°C.
Solutions of test compounds ranging from 10mM to O.OlnM are prepared in DMSO. SIP is diluted in 4% BSA solution as positive controls. The desired amount of membrane prep is diluted with ice-cold assay buffer (20 mM HEPES, pH 7.4,100 mM

NaCl, 10 mM MgCl2, 0.1% Fatty acid-free BSA, 5 \M GDP) and vortexed well. 2 jxl or less of compound is distributed into each well of a round-bottom 96-well polystyrene assay plate, followed by addition of 100 jal of diluted membranes (3-10 jig/well) and kept on ice until the addition of hot GTPyS. [35S]-GTPyS is diluted 1:1000 (v/v) with cold assay buffer and 100 \il is added into each well. The reaction is carried out at room temperature for 90 minutes before the membranes are harvested onto Perkin-Elmer Unifilter® GF/B-96 filter plate using a Packard Filtermate Harvester. After several washes with wash buffer (20 mM HEPES, pH 7.4, 100 mM NaCl, 10 mM MgCl2)) and a rinse with 95% ethanol, the filter is dried in a 37°C oven for 30 minutes. MicroScint-20 is added and the plate sealed for scintillation counting on TopCount. EC50 values are obtained by fitting the GTP [y-35S] binding curves (raw data) with the dose response curve-fitting tool of GraphPad Prism. Six or twelve different concentrations are used to generate a concentration response curve (using three data points per concentration).
EDG-3,-5,-6 and -8 GTP [y-35S] binding assays are carried out in a comparable manner to the EDG-1 GTP [y-35S] binding assay using membranes from CHO cells stably expressing c-terminal c-myc tagged or untagged receptors. For each membrane preparation, titration experiments are first run with SIP control to determine the optimal amount of membranes to be added per assay well. Compounds of the invention were tested according to the above assay and were observed to exhibit selectivity for the EDG-1 receptor. For example, 3-{[2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[ft]thiophen-5-ylmethyl]-amino}-propionic acid (example 1) has an EC50 of 0.6 nM in the above assay and is at least 1000 fold selective for EDG-1 compared to one or more of the other receptors including EDG-3, EDG-5, EDG-6 and EDG-8.
B. In vitro: FLIPR calcium flux assay
Compounds of the invention are tested for agonist activity on EDG-1, EDG-3, EDG-5, and EDG-6 with a FLIPR calcium flux assay. Briefly, CHO cells expressing an EDG receptor are maintained in F-12K medium (ATCC), containing 5% FBS, with 500ug/ml of G418. Prior to the assay, the cells are plated in 384 black clear bottom plates at the density of 10,000 cells/well/25^1 in the medium of F-12K containing 1% FBS. The second day, the cells are washed three times (25 jal/each) with washing buffer. About 25 jal of dye are added

to each well and incubated for 1 hour at 37°C and 5% CO2. The cells are then washed four times with washing buffer (25 jxl/each). The calcium flux is assayed after adding 25 jil of SEQ2871 solution to each well of cells. The same assay is performed with cells expressing each of the different EDG receptors. Titration in the FLIPR calcium flux assay is recorded over a 3-minute interval, and quantitated as maximal peak height percentage response relative to EDG-1 activation.
C. In vivo: Screening Assays for measurement of blood lymphocyte depletion and assessment of heart effect
Measurement of circulating lymphocytes: Compounds are dissolved in DMSO and diluted to obtain a final concentration of 4% DMSO (v/v, final concentration) and then further diluted in a constant volume of Tween80 25%/H20, v/v. Tween80 25%/H20 (200 jil), 4% DMSO, and FTY720 (lOjxg) are included as negative and positive controls, respectively. Mice (C57bl/6 male, 6-10 week-old) are administered 250-300 ^L of compound solution orally by gavages under short isoflurane anesthesia.
Blood is collected from the retro-orbital sinus 6 and 24 hours after drug administration under short isoflurane anesthesia. Whole blood samples are subjected to hematology analysis. Peripheral lymphocyte counts are determined using an automated analyzer. Subpopulations of peripheral blood lymphocytes are stained by fluorochrome-conjugated specific antibodies and analyzed using a fluorescent activating cell sorter (Facscalibur). Two mice are used to assess the lymphocyte depletion activity of each compound screened. The result is an ED50, which is defined as the effective dose required displaying 50 % of blood lymphocyte depletion. Compounds of the invention were tested according to the above assay and were preferably found to exhibit an ED50 of less than lmg/kg, more preferably an ED50 of less than 0.5 mg/kg. For example, 3-{[2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[6]thiophen-5-ylmethyl]-amino}-propionicacid (example 1) exhibits an ED50 of 0.2 mg/kg.
Assessment of Heart Effect: The effects of compounds on cardiac function are monitored using the AnonyMOUSE ECG screening system. Electrocardiograms are recorded in conscious mice (C57bl/6 male, 6-10 week-old) before and after compound administration. ECG signals are then processed and analyzed using the e-MOUSE software.

90 ^g of compound further diluted in 200^1 water, 15% DMSO are injected IP. Four mice are used to assess the heart effect of each compound.
D: In vivo: Anti-angiogenic Activity
Porous chambers containing (i) sphingosine-1-phosphate (5 nM/chamber) or (ii) human VEGF (1 jig/chamber) in 0.5 ml of 0.8% w/v agar (containing heparin, 20 U/ml) are implanted subcutaneously in the flank of mice. SIP or VEGF induces the growth of vascularized tissue around the chamber. This response is dose-dependent and can be quantified by measuring the weight and blood content of the tissue. Mice are treated once a day orally or intravenously with a compound of formula I starting 4-6 hours before implantation of the chambers and continuing for 4 days. The animals are sacrificed for measurement of the vascularized tissues 24 hours after the last dose. The weight and blood content of the vascularized tissues around the chamber is determined. Animals treated with a compound of formula I show reduced weight and/or blood content of the vascularized tissues compared to animals treated with vehicle alone. Compounds of Formula I are anti-angiogenic when administered at a dose of about 0.3 to about 3mg/kg.
E: In vitro: Antitumor Activity
A mouse breast cancer cell line originally isolated from mammary carcinomas is used, e.g. JygMC(A). The cell number is adjusted to 5xl05 for plating in fresh medium before the procedure. Cells are incubated with fresh medium containing 2.5mM of thymidine without FCS for 12 hours and then washed twice with PBS, followed by addition of fresh medium with 10% FCS and additionally incubated for another 12 hours. Thereafter the cells are incubated with fresh medium containing 2.5mM of thymidine without FCS for 12 hours. To release the cells from the block, the cells are washed twice with PBS and replated in fresh medium with 10% FCS. After synchronization, the cells are incubated with or without various concentrations of a compound of formula I for 3,6, 9,12,18 or 24 hours. The cells are harvested after treatment with 0.2% EDTA, fixed with ice-cold 70% ethanol solution, hydrolyzed with 250|ag/ml of RNaseA (type 1-A: Sigma Chem. Co.) at 37°C for 30 minutes and stained with propidium iodide at lOmg/ml for 20 minutes. After the incubation period, the number of cells is determined both by counting cells in a Coulter counter and by the SRB

colorimetric assay. Under these conditions compounds of formula I inhibit the proliferation of the tumor cells at concentrations ranging from 10-12 to 10-6 M.
It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and understanding of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference for all purposes.


WE CLAIM
1. A compound of Formula I:
in which:
n is 1 or 2;
A is chosen from -C(0)OR9, - OP(0)(OR9)2, - P(0)(OR9)2, -S(0)2OR9, -
P(0)(R9)OR9 and l/f-tetrazol-5-yl; and R9 is chosen from hydrogen and Ci^alkyl;
X is a bond or is chosen from Cualkylene, -XiOX2-, -XiNRioX2-, -
XiC(O)NR10X2- -X,NRi0C(O)X2- -X^O^-, -XiS(0)2X2-, -XiSX2- and C2. 9heteroarylene; wherein Xi and X2 are independently chosen from a bond and Ci^alkylene; Rio is chosen from hydrogen and d-ealkyl; and any heteroarylene of X is optionally substituted by a member of the group chosen from halo and Ci^alkyl;
Y is a fused 5,6 or 6,6 hetero bicyclic ring system consisting of at least one
aromatic ring, wherein said fused bicyclic ring system of Y can be optionally substituted with 1 to 3 radicals chosen from halo, hydroxy, cyano, nitro, Ci-ealkyi, C^alkoxy, halo-substituted Ci-ealkyl and halo-substituted C^alkoxy;
Ri is chosen from C6-ioaryl and C2.9heteroaryl; wherein any aryl or heteroaryl
of Rj is optionally substituted by a radical chosen from C6-ioarylCo^alkyl, C2.9heteroarylCo-4alkyl, C3-8cycloalkylCo-4alkyl, C3-8heterocycloalkylCo-4alkyl or Ci_6alkyl; wherein any aryl, heteroaryl, cycloalkyl or heterocycloalkyl group of Rj can be optionally substituted by 1 to 3 radicals chosen from halo, Ci-6alkyl, Ci^alkoxy, halo-substituted-Ci_6alkyl and halo-substituted-Ci-6alkoxy; and any alkyl group of Rj can optionally have a methylene replaced by an atom or group chosen from -S-, -S(O)-, -S(0)2-, -NRl0- and -O-; wherein Rio is chosen from hydrogen or Ci^alkyl;
R2, R3, Rs, R
R4 is chosen from hydrogen and Ci^alkyl; or R7 and either R2, R4 or R5 together with the atoms to which R2, R4, R5 and R7 are attached forms a 4 to 7 member ring; wherein said 4 to 7 member ring is saturated or partially unsaturated; and the pharmaceutical^ acceptable salts, hydrates, solvates, isomers and prodrugs thereof.
2. The compound of claim 1 in which R\ is phenyl, naphthyl furanyl, or thienyl optionally substituted by Ce-ioarylQMalkyl, C2-9heteroarylCo^alkyl, C3-8cycloalkylCo-4alkyl, C3-8heterocycloalkylCo-4alkyl or Ci^alkyl; wherein any aryl, heteroaryl, cycloalkyl or heterocycloalkyl group of Ri can be optionally substituted by one to five radicals chosen from halo, Cj^alkyl, C^alkoxy, halo-substituted-Ci^alkyl and halo-substituted-Ci^alkoxy; and any alkyl group of Ri can optionally have a methylene replaced by an atom or group chosen from -S-, -S(O)-, -S(0)2~, -NR10- and -0-; wherein Rio is hydrogen or Ci^alkyl.
3. The compound of claim 1 in which Y is chosen from:


wherein Rj j is hydrogen or Ci^alkyl; and the left and right asterisks of Y indicate the point of attachment between either -C(R.2)(R3)- and X of Formula I or between X and -C(R2)(R3>- of Formula I, respectively; and Y can be optionally substituted with 1 to 3 radicals chosen from halo, hydroxy, cyano, nitro, Ci^alkyl, Ci^alkoxy, halo-substituted C|. ealkyl and halo-substituted Ci-galkoxy.
4. The compound of claim 1 in which Rj is chosen from:

wherein the asterisk is the point of attachment of Rj with X; m is chosen from 1 and 2; R]2 is hydrogen, C6-ioarylC 5. The compound of claim 1 in which A is -C(0)OH; R2, R3, R5, R 6. The compound of claim 5 in which Y is chosen from:


wherein Rn is hydrogen or C^alkyl; and the left and right asterisks of Y indicate the point of attachment between either -C(R2)(R3>- and X of Formula I or between X and -C(R2)(R3)- of Formula I, respectively; and Y can be optionally substituted with 1 to 3 radicals chosen from chloro, fluoro, methyl, ethyl, cyano and bromo.
7. The compound of claim 6 in which X is chosen from a bond, -NH- and -
N(CH3)-; and Rj is chosen from:

wherein m is chosen from 1 and 2; R12 is hydrogen, phenyl, piperidinyl, 2-methyl-butyl, 3-methyl-butyl, cyclohexyl, cyclohexyl-oxy, cyclopentyl-oxy, sec-butoxy, tetrahydropyranyl, phenoxy, benzo[l,3]dioxolyl, naphthyl, 2,2-dimethyl-pentyl, butyl, benzo[b]furanyl, benzyl, phenethyl, phenyl-ethenyl, 1-phenyl-ethyl and cyclopropyl; wherein any aryl, heteroaryl, cycloalkyl or heterocycloalkyl group of R12 can be optionally substituted by one to three radicals chosen from fluoro, isobutyl, 2-methyl-butyl, trifluoromethyl, chloro, methyl, trifluoromethoxy and methoxy; and R13 is chosen from trifluoromethyl, trifluoromethoxy, methyl, fluoro, chloro and methoxy.

8. The compound of claim 7 chosen from: 3-{[2-(2-trifluoromethyl-biphenyl-
4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-piperidin-l-yl-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-yl)-thieno[2,3-b]pyridin-5-ylmethyI]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-6-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-yl)-2,3-dihydro-lH-isoindol-5-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-yl)-benzooxazol-5-ylmethyl]-amino}-propionic acid, 1 -[2-(4-Isobutyl-3-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-azetidine-3-carboxylic acid, 3- {[2-(2-Trifluoromethyl-biphenyl-4-yl)-benzofuran-5-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-yl)-benzothiazol-6-ylmethylj-amino}-propionic acid, 3-{[3-Chloro-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, l-[2-(2-Trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-azetidine-3-carboxylic acid, 3- {[2-(2'-Fluoro-2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3- {[2-(5-Fluoro-2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-6-ylmethyl]-amino}-propionic acid, 3-{[3-Fluoro-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Cyclohexyl-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino} -propionic acid, 3- {[4-Chloro-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, l-[2-(4-Cyclohexyl-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-azetidine-3-carboxylic acid, 3- {[6-Methoxy-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[6-Chloro-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Cyclopentyloxy-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-sec-Butoxy-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-sec-Butyl-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Isobutyl-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Cyclohexyloxy-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-({2-[4-(Tetrahydro-pyran-4-yl)-3-trifluoromethyl-phenyl]-benzo[b]thiophen-5-ylmethyl}-amino)-propionicacid, 3-{[3-Methyl-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[3-Cyano-2-(2-

trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3- {[3-Bromo-2-(2-trifluoromethyl-biphenyl-4-yl)-benzo[ty^
acid, 3»{[2-(3-Fluoro-5-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3- {[2-(2-Fluoro-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3- {[2-(2-Trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, l-[2-(2-Trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-4-ylmethyl]-azetidine-3-carboxylic acid, 3- {[2-(4-Chloro-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-4-ylmethyl]-amino}-propionic acid, 3-{[2-(2,5-Bis-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Methyl-5-trifluoromethyl-phenyl)-ben2o[b]thiophen-5-ylmethyl]-amino} -propionic acid, 3-[(2-Phenyl-benzo[b]thiophen-5-ylmethyl)-amino]-propionic acid, 3- {[2-(4-Methyl-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, l-[2-(3-Trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-azetidine-3-carboxylic acid, 3- {[2-(4-Fluoro-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino} -propionic acid, 2-Fluoro-3-{[2-(3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(3,5-Bis-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Trifluoromethoxy-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionicacid, 1-[2-(2-Fluoro-5-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-azetidine-3-carboxylic acid, 3-{[2-(2-Chloro-5-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(3-Trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, l-[2-(3-Trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-pyrrolidine-3-carboxylic acid, 3- {[2-(2-Fluoro-5-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Methoxy-3-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Methoxy-5-trifluoromethyl-phenyl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3- {[3-(2-Trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-5-ylmethyl]-amino}-propionic acid, 3- {[5-(2-Trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-2-ylmethyl]-amino}-propionic acid, 3-{[5-(4-Cyclohexyl-3-trifluoromethyl-phenyl)-benzo[b]thiophen-2-ylmethyl]-amino}-propionic acid, 3- {[3-Chloro-5-(2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-2-

ylmethylj-amino}-propionic acid, l-[5-(2-Trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-2-ylmethyl]-azetidine-3-carboxylic acid, 3- {[3-Bromo-5-(2-trifluoromethyl-biphenyl-4-yl)-benzo[b]thiophen-2-ylmethyl]-amino}-propionicacid, 3-{[2-(2,-Fluoro-2-trifluoromethyl-biphenyl-4-yl)-ben2ooxazol-5-ylmethyl]-amino}-propionicacid,3-{[2-(3,«Fluoro-2 trifluoromethyl-biphenyl-4-yl)-ben2ooxazol-5-ylmethyl]-amino}-propionic acid, 3- {[2-(T' Chloro-2-trifluoromethyl-biphenyl-4-yl)^
3-{[2-(4-Phenoxy-3-trifluoromethyl-phenyl)-benzooxazol-5-ylmethyl]-amino}-propionic acid, 3- {[2-(2'-Fluoro-2-trifluoromethyl-biphenyl-4-yl)-benzooxazol-6-ylmethyl]-amino} -propionic acid, 3-{[2-(4-Cyclohexyl-3-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-yl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(5,-Fluoro-2,-methyl-2-trifluoromethyl-biphenyl-4-yl)-benzooxazol-6-ylmethylj-amino}-propionic acid, 2-Fluoro-3-{[2-(2'-fluoro-2-trifluoromethyl-biphenyl-4-yl)-benzooxazol-6-ylmethyl]-amino}-propionicac Dichloro-2-(2-trifluoromethyl-biphenyl-4-yl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(3LChloro-2-trifluorome&^
propionic acid, 3- {[5-Chloro-2«(2-trifluoromethyl-biphenyl-4-yl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3- {[5-Bromo-2-(2-trifluoromethyl-biphenyl-4-yl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Isobutyl-3-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Benzo[l,3]dioxol-5-yl-3-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-amino} -propionic acid, 3- {[2-(4-Cyclohexyl-3-fluoro-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Fluoro-biphenyl-4-yl)-benzooxazol-6-yImethyl]-amino}-propionic acid, 3-{[2-(3,-Chloro-4f-fluoro-2-trifluoromethyl-biphenyl-4-yl)-ben20oxazol-6-ylmethyl]-amino}-propionic acid, 3- {[2-(4-sec-Butyl-3-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[5-Ethyl-2-(2-trifluoromethyl-biphenyl-4-yl^
3-{[2-(4-Naphthalen-2-yl-3-trifluoromethyI-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 1- {2-[4-(2,2-Dimethyl-propyl)-3-trifluoromethyl-phenyl]-benzooxazol-6-ylmethyl} -azetidine-3-carboxylic acid, 3- {[2-(4-Butyl-3-trifluoromethyl-phenyl)-benzooxazol~6-ylmethyl]-amino}-propionic acid, 3- {[2-(4-Benzofuran-2-yl-3-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-({2-[4-(2,6-Difluoro-benzyl)-3-trifluoromethyl-phenyl]-benzooxazol-6-ylm'ethyl}-amino)-pro

acid, 3- {[2-(4-PhenethylO-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyI]-amino}-propionic acid, 3-{[2-(4-Styryl-3-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-({2-[4-(l-Phenyl-ethyl)-3«trifluoromethyl-phenyl]-benzooxazol-6-ylmethyl} -amino)-propionic acid, 3- {[2-(5,-Fluoro-2f-methoxy-2-trifluoromethyl-biphenyl-4-yl)-benzooxazol-6-ylmethyl]-methyl-amino}-propionic acid, 3-{[2-(5f-Fluoro-2f-methoxy-2-trifluoromethyl-biphenyl-4-yl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(3-Trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(4-tert-Butyl-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, l-[2-(2-Fluoro-5-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-azetidine-3-carboxylic acid, 3- {[5-Chloro-2-(3-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, l-[2-(2-FIuoro-5«trifluoromethyl-phenyl)-benzooxazol-5-ylmethyl]-azetidine-3-carboxylicacid, 1-[2-(2-Fluoro-5-trifluoromethyl-phenyl)-bera^
3-{[2-(4-Chloro-3-trifluoromethyl-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionicaci 3-{[2-(4-Cyclopropyl-3-trifluoromethyl-phenyl)-ben2ooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Fluoro-phenyl)-benzooxazol«6-ylmethyl]-amino}-propionic acid, 3-{[2-(3-Fluoro-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Fluoro-phenyl)-benzooxazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(4-Cyclohexyl-3-trifluoromethyl-phenyl)-benzoftiran-5-ylmethyl]-amino}-propionicacid, 3«{[2-(4-Cyclohexyl-3-trifluoromethyl-phenyl)-2,3-dihydro-lH-isoindol-5-ylmethyl]-amino} -propionic acid, 3-{[2-(4-CyclohexylO-trifluoromethyl-phenyl)-2H-isoindol-5-ylmethyl]-amino}-propionicacid, 3- {[2-(2-Trifluoromethyl-biphenyl-4-yl)-benzothiazol-5-ylmethyl]-amino}-propionic acid, 3- {[2-(2-Trifluoromethyl-biphenyI-4-yl)-benzothiazol-7-ylmethyl]-amino} -propionic acid, 3- {[2-(3-Trifluoromethyl-phenyl)-benzothiazol-7-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-yl)-2H-indazol-6-ylmethyl]-amino}-propionic acid, 3-{[2-(5-Fluoro-2-trifluoromethyl-biphenyl-4-yl)-2H-indazol-6-ylmethyl]-amino} -propionic acid, 1 -[2-(5-Fluoro-2-trifluoromethyl-biphenyl-4-yl)-2H-indazol-6-ylmethyl]-azetidine-3-carboxylic acid, l-[2-(2-Trifluoromethyl-biphenyl-4-yl)-lH-benzoimidazol-5-ylmethyl]-azetidine-3-carboxylic acid, 3- {[3-Methyl-2-(2-trifluoromethyl-biphenyl-4-yl)-3H-benzoimidazol-5-ylmethyI]-amino}-propionic acid, 3-{[l-Methyl-2-(2-trifluoromethyl-biphenyl-4-yl)-lH-benzoimidazol-5-ylmethyl]-amino}-propionic acid, 3-{[2-(2-Trifluoromethyl-biphenyl-4-ylmethyl)-2,3-dihydro-lH-isoindol-5-ylmethyl]-amino}-

propionic acid, 3- {[2-(2-Trifluoromethyl-biphenyl-4-ylamino)-benzooxazol-5-ylmethyl]-amino}-propionic acid, 3-( {2-[Methyl-(2-trifluoromethyl-biphenyl-4-yl)-amino]-benzooxazol-5-ylmethyl} -amino)-propionic acid, 3- {[4-Oxo-2-(2-trifluoromethyl-biphenyl-4-yl)-4H-chromen-7-ylmethyl]-amino}-propionic acid, 3-{[4-Oxo-2-(2-trifluoromethyl-biphenyl-4-yl)-4H-chromen-6-ylmethyl]-amino}-propionic acid and 1-[4-Oxo-2-(2« trifluoromethyl-biphenyl-4-yl)-4H-chromen-6-ylmethyl]-azetidine-3-carboxylicacid.
9. A pharmaceutical composition comprising a therapeutically effective amount
of a compound of Claim 1 in combination with a pharmaceutically acceptable excipient.
10. A method for treating a disease in an animal in which alteration of
EDG/S1P receptor mediated signal transduction can prevent, inhibit or ameliorate the
pathology and/or symptomology of the disease, which method comprises administering to
the animal a therapeutically effective amount of a compound of Claim 1.
11. A method for preventing or treating disorders or diseases mediated by
lymphocytes, for preventing or treating acute or chronic transplant rejection or T-cell
mediated inflammatory or autoimmune diseases, for inhibiting or controlling deregulated
angiogenesis, or for preventing or treating diseases mediated by a neo-angiogenesis process
or associated with deregulated angiogenesis in a subject comprising administering to the
subject in need thereof an effective amount of a compound of claimsl, or a pharmaceutically
acceptable salt thereof.
12. The use of a compound of claim 1 in the manufacture of a medicament for
treating a disease in an animal in which alteration of EDG/S1P receptor mediated signal
transduction contributes to the pathology and/or symptomology of the disease.


Documents:

3049-chenp-2005 complete specification as granted.pdf

3049-CHENP-2005 CLAIMS GRANTED.pdf

3049-CHENP-2005 CORRESPONDENCE OTHERS.pdf

3049-CHENP-2005 CORRESPONDENCE PO.pdf

3049-CHENP-2005 FORM 1.pdf

3049-CHENP-2005 FORM 2.pdf

3049-CHENP-2005 FORM 3.pdf

3049-CHENP-2005 PETITIONS.pdf

3049-chenp-2005-abstract.pdf

3049-chenp-2005-assignement.pdf

3049-chenp-2005-claims.pdf

3049-chenp-2005-correspondnece-others.pdf

3049-chenp-2005-description(complete).pdf

3049-chenp-2005-form 1.pdf

3049-chenp-2005-form 18.pdf

3049-chenp-2005-form 26.pdf

3049-chenp-2005-form 3.pdf

3049-chenp-2005-form 5.pdf

3049-chenp-2005-pct.pdf


Patent Number 234776
Indian Patent Application Number 3049/CHENP/2005
PG Journal Number 29/2009
Publication Date 17-Jul-2009
Grant Date 15-Jun-2009
Date of Filing 18-Nov-2005
Name of Patentee IRM LLC
Applicant Address HURST HOLME, 12 TROTT ROAD, HAMILTON 11
Inventors:
# Inventor's Name Inventor's Address
1 MI, YUNA 11175 AFFINITY COURT, UNIT 45,SAN DIEGO, CA 92131
2 PAN, SHIFENG, 13880 KERRY LANE,SAN DIEGO, CA 92130, USA
3 GRAY, NATHANAEL, SCHIANDER 5652 LAMAS STREET, SAN DIEGO, CA 92122,USA
4 GAO, WENQI 7958 HARMARSH STREET, SAN DIEGO, CA 92123
5 FAN, YI 12228 PEPPER TREE LANE, POWAY, CA 92064
6 JIANG, TAO 10721 CALLE MAR DE MARIPOSA, SAN DIEGO, CA 92130
PCT International Classification Number C07D333/60
PCT International Application Number PCT/US04/015702
PCT International Filing date 2004-05-19
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/471,931 2003-05-19 U.S.A.
2 60/562,183 2004-04-14 U.S.A.