WO2011005290A1 - Disubstituted oxadiazole derivatives useful in the treatment of autoimmune and inflammatory disorders - Google Patents

Disubstituted oxadiazole derivatives useful in the treatment of autoimmune and inflammatory disorders Download PDF

Info

Publication number
WO2011005290A1
WO2011005290A1 PCT/US2010/001787 US2010001787W WO2011005290A1 WO 2011005290 A1 WO2011005290 A1 WO 2011005290A1 US 2010001787 W US2010001787 W US 2010001787W WO 2011005290 A1 WO2011005290 A1 WO 2011005290A1
Authority
WO
WIPO (PCT)
Prior art keywords
cyano
compound according
chloro
dihydro
compounds
Prior art date
Application number
PCT/US2010/001787
Other languages
French (fr)
Inventor
Robert M. Jones
Daniel J. Buzard
Sangdon Han
Juerg Lehmann
Luis A. Lopez
Brett Ullman
Andrew M. Kawasaki
Lars Thoresen
Xiuwen Zhu
Original Assignee
Arena Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Arena Pharmaceuticals, Inc. filed Critical Arena Pharmaceuticals, Inc.
Publication of WO2011005290A1 publication Critical patent/WO2011005290A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

The present invention relates to certain disubstituted oxadiazole derivatives of Formula (Ia) and pharmaceutically acceptable salts, solvates, and hydrates thereof, which exhibit useful pharmacological properties, for example, as agonists of the S1P1 receptor. Also provided by the present invention are pharmaceutical compositions containing compounds of the invention, and methods of using the compounds and compositions of the invention in the treatment of S1P1-associated disorders, for example, psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy, glomerulosclerosis, gastritis, polymyositis, thyroiditis, vitiligo, hepatitis, biliary cirrhosis, microbial infections and associated diseases, viral infections and associated diseases, diseases and disorders mediated by lymphocytes, auto immune diseases, inflammatory diseases, and cancer.

Description

DISUBSTITUTED OXADIAZOLE DERIVATIVES USEFUL IN THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISORDERS
FIELD OF THE INVENTION
The present invention relates to certain disubstituted oxadiazole derivatives of Formula
(Ia) and pharmaceutically acceptable salts, solvates, and hydrates thereof, which exhibit useful pharmacological properties, for example, as agonists of the SlPl receptor.
Also provided by the present invention are pharmaceutical compositions containing compounds of the invention, and methods of using the compounds and compositions of the invention in the treatment of S 1 P 1 -associated disorders, for example, psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy, glomerulosclerosis, gastritis, polymyositis, thyroiditis, vitiligo, hepatitis, biliary cirrhosis, microbial infections and associated diseases, viral infections and associated diseases, diseases and disorders mediated by lymphocytes, auto immune diseases, inflammatory diseases, and cancer.
BACKGROUND OF THE INVENTION
The present invention relates to compounds that are SlPl receptor agonists having at least immunosuppressive, anti-inflammatory, and/or hemostatic activities, e.g. by virtue of modulating leukocyte trafficking, sequestering lymphocytes in secondary lymphoid tissues, and/or enhancing vascular integrity.
The present application is in part focused on addressing an unmet need for
immunosuppressive agents such as may be orally available which have therapeutic efficacy for at least autoimmune diseases and disorders, inflammatory diseases and disorders (e.g., acute and chronic inflammatory conditions), transplant rejection, cancer, and/or conditions that have an underlying defect in vascular integrity or that are associated with angiogenesis such as may be pathologic (e.g., as may occur in inflammation, tumor development, and atherosclerosis) with fewer side effects such as the impairment of immune responses to systemic infection.
The sphingosine-1 -phosphate (SlP) receptors 1-5 constitute a family of G protein- coupled receptors with a seven-transmembrane domain. These receptors, referred to as SlPl to S1P5 (formerly termed endothelial differentiation gene (EDG) receptor- 1, -5, -3, -6, and -8, respectively; Chun et al., Pharmacological Reviews, 54:265-269, 2002), are activated via binding by sphingosine-1 -phosphate, which is produced by the sphingosine kinase-catalyzed phosphorylation of sphingosine. SlPl, S1P4, and S1P5 receptors activate Gi but not Gq, whereas S1P2 and S1P3 receptors activate both Gi and Gq. The S1P3 receptor, but not the SlPl receptor, responds to an agonist with an increase in intracellular calcium. SlP receptor agonists having agonist activity on the SlPl receptor have been shown to rapidly and reversibly induce lymphopenia (also referred to as peripheral lymphocyte lowering (PLL); Hale et al, Bioorg. Med. Chem. Lett., 14:3351-3355, 2004). This is attended by clinically useful immunosuppression by virtue of sequestering T- and B-cells in secondary lymphoid tissue (lymph nodes and Peyer's patches) and thus apart from sites of inflammation and organ grafts (Rosen et al, Immunol. Rev., 195:160-177, 2003; Schwab et al., Nature Immunol., 8:1295-1301, 2007). This lymphocyte sequestration, for example in lymph nodes, is thought to be a consequence of concurrent agonist-driven functional antagonism of the SlPl receptor on T-cells (whereby the ability of SlP to mobilize T-cell egress from lymph nodes is reduced) and persistent agonism of the SlPl receptor on lymph node endothelium (such that barrier function opposing transmigration of lymphocytes is increased) (Matloubian et al, Nature, 427:355-360, 2004; Baumruker et al, Expert Opin. Investig. Drugs, 16:283-289, 2007). It has been reported that agonism of the SlPl receptor alone is sufficient to achieve lymphocyte sequestration (Sanna et al, J Biol Chem., 279:13839-13848, 2004) and that this occurs without impairment of immune responses to systemic infection (Brinkmann et al, Transplantation, 72:764-769, 2001; Brinkmann et al, Transplant Pr oc, 33:530-531, 2001).
That agonism of endothelial SlPl receptors has a broader role in promoting vascular integrity is supported by work implicating the SlPl receptor in capillary integrity in mouse skin and lung (Sanna et al, Nat Chem Biol, 2:434-441, 2006). Vascular integrity can be
compromised by inflammatory processes, for example as may derive from sepsis, major trauma and surgery so as to lead to acute lung injury or respiratory distress syndrome (Johan
Groeneveld, Vascul. Pharmacol, 39:247-256, 2003).
An exemplary SlP receptor agonist having agonist activity on the SlPl receptor is FTY720 (fingolimod), an immunosuppressive agent currently in clinical trials (Martini et al, Expert Opin. Investig. Drugs, 16:505-518, 2007). FTY720 acts as a prodrug which is phosphorylated in vivo; the phosphorylated derivative is an agonist for SlPl, S 1P3, S1P4, and S1P5 receptors (but not the S1P2 receptor) (Chiba, Pharmacology & Therapeutics, 108:308- 319, 2005). FTY720 has been shown to rapidly and reversibly induce lymphopenia (also referred to as peripheral lymphocyte lowering (PLL); Hale et al, Bioorg. Med. Chem. Lett., 14:3351-3355, 2004). This is attended by clinically useful immunosuppression by virtue of sequestering T- and B-cells in secondary lymphoid tissue (lymph nodes and Peyer's patches) and thus apart from sites of inflammation and organ grafts (Rosen et al, Immunol. Rev., 195:160-177, 2003; Schwab et al, Nature Immunol, 8:1295-1301, 2007).
In clinical trials, FTY720 elicited an adverse event {i.e., transient asymptomatic bradycardia) due to its agonism of the S1P3 receptor (Budde et al, J. Am. Soc. Nephrol,
13:1073-1083, 2002; Sanna et al, J. Biol Chem., 279:13839-13848, 2004; Ogawa et al, BBRC, 361:621-628, 2007). FTY720 has been reported to have therapeutic efficacy in at least: a rat model for autoimmune myocarditis and a mouse model for acute viral myocarditis (Kiyabayashi et al, J. Cardiovasc. Pharmacol, 35:410-416, 2000; Miyamoto et al, J. Am. Coll. Cardiol, 37:1713- 1718, 2001); mouse models for inflammatory bowel disease including colitis (Mizushima et al, Inflamm. BowelDis., 10: 182-192, 2004; Deguchi et al, Oncology Reports, 16:699-703, 2006; Fujii et al, Am. J. Physiol. Gastrointest. Liver Physiol, 291 :G267-G274, 2006; Daniel et al, J. Immunol, 178:2458-2468, 2007); a rat model for progressive mesangioproliferative glomerulonephritis (Martini et al, Am. J. Physiol. Renal Physiol, 292:F1761-F1770, 2007); a mouse model for asthma, suggested to be primarily through the SlPl receptor on the basis of work using the SlPl receptor agonist SEW2871 (Idzko et al, J. Clin. Invest., 116:2935-2944,
2006); a mouse model for airway inflammation and induction of bronchial hyperresponsiveness (Sawicka et al, J. Immunol, 171;6206-6214, 2003); a mouse model for atopic dermatitis (Kohno et al, Biol. Pharm. Bull, 27:1392-1396, 2004); a mouse model for ischemia-reperfusion injury (Kaudel et al, Transplant. Proc, 39:499-502, 2007); a mouse model for systemic lupus erythematosus (SLE) (Okazaki et al, J. Rheumatol, 29:707-716, 2002; Herzinger et al, Am. J. Clin. Dermatol., 8:329-336, 2007); rat models for rheumatoid arthritis (Matsuura et al, Int. J. Immunopharmacol, 22:323-331, 2000; Matsuura et al, Inflamm. Res., 49:404-410, 2000); a rat model for autoimmune uveitis (Kurose et al, Exp. Eye Res., 70:7-15, 2000); mouse models for type I diabetes (Fu et al, Transplantation, 73: 1425-1430, 2002; Maki et al, Transplantation, 74:1684-1686, 2002; Yang et al, Clinical Immunology, 107:30-35, 2003; Maki et al,
Transplantation, 79:1051-1055, 2005); mouse models for atherosclerosis (Nofer et al, Circulation, 115:501-508, 2007; Keul et al, Arterioscler. Thromb. Vase. Biol, 27:607-613, 2007); a rat model for brain inflammatory reaction following traumatic brain injury (TBI) (Zhang et al, J. Cell. MoI Med., 11:307-314, 2007); and mouse models for graft coronary artery disease and graft-versus-host disease (GVHD) (Hwang et al, Circulation, 100: 1322-1329, 1999; Taylor et al, Blood, 110:3480-3488, 2007). In vitro results suggest that FTY720 may have therapeutic efficacy for jS-amyloid-related inflammatory diseases including Alzheimer's disease (Kaneider et al, FASEB J., 18:309-311, 2004). KRP-203, an SlP receptor agonist having agonist activity on the SlPl receptor, has been reported to have therapeutic efficacy in a rat model for autoimmune myocarditis (Ogawa et al, BBRC, 361 :621-628, 2007). Using the
SlPl receptor agonist SEW2871, it has been shown that agonism of endothelial SlPl receptors prevents proinflammatory monocyte/endothelial interactions in type I diabetic vascular endothelium (Whetzel et al, Circ. Res., 99:731-739, 2006) and protects the vasculature against TNFα-mediated monocyte/endothelial interactions (Bolick et al, Arterioscler. Thromb. Vase. Biol, 25:976-981, 2005).
Additionally, FTY720 has been reported to have therapeutic efficacy in experimental autoimmune encephalomyelitis (EAE) in rats and mice, a model for human multiple sclerosis (Brinkmann et al, J. Biol. Chem., 277:21453-21457, 2002; Fujino et al, J. Pharmacol Exp. Ther., 305:70-77, 2003; Webb et al, J. Neuroimmunol., 153: 108-121, 2004; Rausch et al, J. Magn. Reson. Imaging, 20:16-24, 2004; Kataoka et al, Cellular & Molecular Immunology, 2:439-448, 2005; Brinkmann et al, Pharmacology & Therapeutics, 115:84-105, 2007;
Baumruker et al, Expert Opin. Investig. Drugs, 16:283-289, 2007; Balatoni et al, Brain
Research Bulletin, 74:307-316, 2007). Furthermore, FTY720 has been found to have therapeutic efficacy for multiple sclerosis in clinical trials. In Phase II clinical trials for relapsing-remitting multiple sclerosis, FTY720 was found to reduce the number of lesions detected by magnetic resonance imaging (MRI) and clinical disease activity in patients with multiple sclerosis (Kappos et al, N. Engl. J. Med., 355:1124-1140, 2006; Martini et al, Expert Opin. Investig.
Drugs, 16:505-518, 2007; Zhang et al, Mini-Reviews in Medicinal Chemistry, 7:845-850, 2007; Brinkmann, Pharmacology & Therapeutics, 115:84-105, 2007). FTY720 is currently in Phase HI studies of remitting-relapsing multiple sclerosis (Brinkmann, Pharmacology & Therapeutics, 115:84-105, 2007; Baumruker et al, Expert. Opin. Investig. Drugs, 16:283-289, 2007; Dev et al, Pharmacology and Therapeutics, 117:77-93, 2008).
Recently, FTY720 has been reported to have anti-viral activity. Specific data has been presented in the lymphocytic choriomeningitis virus (LCMV) mouse model, wherein the mice were infected with either the Armstrong or the clone 13 strain of LCMV (Premenko-Lanier et al., Nature, 454, 894, 2008).
FTY720 has been reported to impair migration of dendritic cells infected with
Francisella tularensis to the mediastinal lymph node, thereby reducing the bacterial colonization of it. Francisella tularensis is associated with tularemia, ulceroglandular infection, respiratory infection and a typhoidal disease (E. Bar-Haim et al, PLoS Pathogens, 4(11):
el000211. doi:10.1371/journal.ppat.l000211, 2008).
It has also been recently reported that a short-term high dose of FTY720 rapidly reduced ocular infiltrates in experimental autoimmune uveoretinitis. When given in the early stages of ocular inflammation, FTY720 rapidly prevented retinal damage. It was reported to not only prevent infiltration of target organs, but also reduce existing infiltration (Raveney et al, Arch. Ophthalmol. 126(10), 1390, 2008).
It has been reported that treatment with FTY720 relieved ovariectomy-induced osteoporosis in mice by reducing the number of mature osteoclasts attached to the bone surface. The data provided evidence that SlP controlled the migratory behavior of osteoclast precursors, dynamically regulating bone mineral homeostasis (Ishii et al., Nature, advance online publication, 8 February 2009, doi:10.1038/nature07713).
Agonism of the SlPl receptor has been implicated in enhancement of survival of oligodendrocyte progenitor cells. Survival of oligodendrocyte progenitor cells is a required component of the remyelination process. Remyelination of multiple sclerosis lesions is considered to promote recovery from clinical relapses. (Miron et al, Ann. Neurol., 63:61-71, 2008; Coelho et al, J. Pharmacol. Exp. Ther., 323:626-635, 2007; Dev et al, Pharmacology and Therapeutics, 117:77-93, 2008). It also has been shown that the SlPl receptor plays a role in platelet-derived growth factor (PDGF)-induced oligodendrocyte progenitor cell mitogenesis (Jung et al, GHa, 55:1656-1667, 2007).
Agonism of the SlPl receptor has also been reported to mediate migration of neural stem cells toward injured areas of the central nervous system (CNS), including in a rat model of spinal cord injury (Kimura et al, Stem Cells, 25:115-124, 2007).
Agonism of the SlPl receptor has been implicated in the inhibition of keratinocyte proliferation (Sauer et al, J. Biol Chem., 279:38471-38479, 2004), consistent with reports that SlP inhibits keratinocyte proliferation (Kim et al, Cell Signal, 16:89-95, 2004). The hyperproliferation of keratinocytes at the entrance to the hair follicle, which can then become blocked, and an associated inflammation are significant pathogenetic factors of acne (Koreck et al, Dermatology, 206:96-105, 2003; Webster, Cutis, 16:4-1, 2005).
FTY720 has been reported to have therapeutic efficacy in inhibiting pathologic angiogenesis, such as that as may occur in tumor development. Inhibition of angiogenesis by FTY720 is thought to involve agonism of the SlPl receptor (Oo et al, J. Biol Chem., 282;9082-9089, 2007; Schmid et al, J. Cell Biochem., 101 :259-270, 2007). FTY720 has been reported to have therapeutic efficacy for inhibiting primary and metastatic tumor growth in a mouse model of melanoma (LaMontagne et al, Cancer Res., 66:221-231, 2006). FTY720 has been reported to have therapeutic efficacy in a mouse model for metastatic hepatocellular carcinoma (Lee et al, CHn. Cancer Res., 11:84588466, 2005).
It has been reported that oral administration of FTY720 to mice potently blocked VEGF-induced vascular permeability, an important process associated with angiogenesis, inflammation, and pathological conditions such as sepsis, hypoxia, and solid tumor growth (T Sanchez et al, J. Biol. Chem., 278(47), 47281-47290, 2003).
Cyclosporin A and FK506 (calcineurin inhibitors) are drugs used to prevent rejection of transplanted organs. Although they are effective in delaying or suppressing transplant rejection, classical immunosuppressants such as cyclosporin A and FK506 are known to cause several undesirable side effects including nephrotoxicity, neurotoxicity, /3-cell toxicity and
gastrointestinal discomfort. There is an unmet need in organ transplantation for an
immunosuppressant without these side effects which is effective as a monotherapy or in combination with a classical immunosuppressant for inhibiting migration of, e.g., alloantigen- reactive T-cells to the grafted tissue, thereby prolonging graft survival.
FTY720 has been shown to have therapeutic efficacy in transplant rejection both as a monotherapy and in synergistic combination with a classical immunosuppressant, including cyclosporin A, FK506 and RAD (an mTOR inhibitor). It has been shown that, unlike the classical immunosuppressants cyclosporin A, FK506 and RAD, FTY720 has efficacy for prolonging graft survival without inducing general immunosuppression, and this difference in drug action is believed to be relevant to the synergism observed for the combination (Brinkmann et al, Transplant Proc, 33:530-531, 2001; Brinkmann et al, Transplantation, 72:764-769, 2001).
Agonism of the SlPl receptor has been reported to have therapeutic efficacy for prolonging allograft survival in mouse and rat skin allograft models (Lima et al., Transplant Proc, 36:1015-1017, 2004; Yan et al, Bioorg. & Med. Chem. Lett., 16:3679-3683, 2006). FTY720 has been reported to have therapeutic efficacy for prolonging allograft survival in a rat cardiac allograft model (Suzuki et al., Transpl. Immunol., 4:252-255, 1996). FTY720 has been reported to act synergistically with cyclosporin A to prolong rat skin allograft survival (Yanagawa et al., J. Immunol, 160:5493-5499, 1998), to act synergistically with cyclosporin A and with FK506 to prolong rat cardiac allograft survival, and to act synergistically with cyclosporin A to prolong canine renal allograft survival and monkey renal allograft survival (Chiba et al., Cell MoI. Biol, 3:11-19, 2006). KRP-203, an SlP receptor agonist has been reported to have therapeutic efficacy for prolonging allograft survival in a rat skin allograft model and both as monotherapy and in synergistic combination with cyclosporin A in a rat cardiac allograft model (Shimizu et al, Circulation, 111:222-229, 2005). KRP-203 also has been reported to have therapeutic efficacy in combination with mycophenolate mofetil (MMF; a prodrug for which the active metabolite is mycophenolic acid, an inhibitor of purine biosynthesis) for prolonging allograft survival both in a rat renal allograft model and in a rat cardiac allograft model (Suzuki et al, J. Heart Lung Transplant, 25:302-209, 2006; Fujishiro et al, J. Heart Lung Transplant, 25:825-833, 2006). It has been reported that an agonist of the SlPl receptor, AUY954, in combination with a subtherapeutic dose of RADOOl
(Certican/Everolimus, an mTOR inhibitor) can prolong rat cardiac allograft survival (Pan et al, Chemistry & Biology, 13:1227-1234, 2006). In a rat small bowel allograft model, FTY720 has been reported to act synergistically with cyclosporin A to prolong small bowel allograft survival (Sakagawa et al, Transpl Immunol, 13:161-168, 2004). FTY720 has been reported to have therapeutic efficacy in a mouse islet graft model (Fu et al, Transplantation, 73:1425-1430, 2002; Liu et al, Microsurgery, 27:300-304; 2007) and in a study using human islet cells to evidence no detrimental effects on human islet function (Truong et al, American Journal of Transplantation, 7:2031-2038, 2007).
FTY720 has been reported to reduce the nociceptive behavior in the spared nerve injury model for neuropathic pain which does not depend on prostaglandin synthesis (O. Costu et al, Journal of Cellular and Molecular Medicine 12(3), 995-1004, 2008).
FTY720 has been reported to impair initiation of murine contact hypersensitivity (CHS). Adoptive transfer of immunized lymph node cells from mice treated with FTY720 during the sensitization phase was virtually incapable of inducing CHS response in recipients (D. Nakashima et al, J. Investigative Dermatology (128(12), 2833-2841, 2008).
It has been reported that prophylactic oral administration of FTY720 (1 mg/kg, three times a week), completely prevented the development of experimental autoimmune myasthenia gravis (EAMG) in C57BL/6 mice (T. Kohono et al., Biological & Pharmaceutical Bulletin, 28(4), 736-739, 2005).
In one embodiment, the present invention encompasses compounds which are agonists of the SlPl receptor having selectivity over the S1P3 receptor. The S1P3 receptor, and not the SlPl receptor, has been directly implicated in bradycardia (Sanna et al., J. Biol. Chem., 279:13839-13848, 2004). An SlPl receptor agonist selective over at least the S1P3 receptor has advantages over current therapies by virtue of an enhanced therapeutic window, allowing better tolerability with higher dosing and thus improving efficacy as therapy. The present invention encompasses compounds which are agonists of the SlPl receptor and which exhibit no or substantially no activity for bradycardia.
SlPl receptor agonists are useful for treating or preventing conditions where suppression of the immune system or agonism of the SlPl receptor is in order, such as diseases and disorders mediated by lymphocytes, transplant rejection, autoimmune diseases and disorders, inflammatory diseases and disorders, and conditions that have an underlying defect in vascular integrity or that relate to angiogenesis such as may be pathologic.
Citation of any reference throughout this application is not to be construed as an admission that such reference is prior art to the present application.
SUMMARY OF THE INVENTION
The present invention encompasses compounds of Formula (Ia) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000009_0001
(Ia)
wherein:
E is N or CR1;
G is N or CR3;
R1 is selected from the group consisting Of Ci-C6 alkyl, C1-C6 alkylsulfonyl, cyano, Ci-C6 haloalkyl, Cj-C6 haloacyl, and halogen; R2, R3, and R4 are each independently selected from the group consisting of H, Ci-C6 alkoxy, CrC6 alkyl, cyano, C3-C7 cycloalkyl, C3-C7 cycloalkyloxy, Ci-C6 haloalkoxy, C1-C6 haloalkyl, and halogen; and
R5 is H or halogen.
The present invention encompasses compounds which are SlPl receptor agonists having at least immunosuppressive, anti-inflammatory and/or hemostatic activities, e.g. by virtue of modulating leukocyte trafficking, sequestering lymphocytes in secondary lymphoid tissues, and/or enhancing vascular integrity.
SlPl receptor agonists are useful for treating or preventing conditions where suppression of the immune system or agonism of the SlPl receptor is in order, such as diseases and disorders mediated by lymphocytes, transplant rejection, autoimmune diseases and disorders, inflammatory diseases and disorders (e.g., acute and chronic inflammatory conditions), cancer, and conditions that have an underlying defect in vascular integrity or that are associated with angiogenesis such as may be pathologic {e.g. , as may occur in inflammation, tumor development and atherosclerosis). Such conditions where suppression of the immune system or agonism of the SlPl receptor is in order include diseases and disorders mediated by lymphocytes; conditions that have an underlying defect in vascular integrity; autoimmune diseases and disorders; inflammatory diseases and disorders (e.g., acute and chronic inflammatory conditions); acute or chronic rejection of cells; tissue or solid organ grafts;
arthritis, including psoriatic arthritis, and rheumatoid arthritis; diabetes, including type I diabetes; demyelinating disease, including multiple sclerosis; ischemia-reperfusion injury, including renal and cardiac ischemia-reperfusion injury; inflammatory skin disease, including psoriasis, atopic dermatitis, and acne; hyperproliferative skin disease, including acne;
inflammatory bowel disease, including Crohn's disease, and ulcerative colitis; systemic lupus erythematosus; asthma; uveitis; myocarditis; allergy; atherosclerosis; brain inflammation, including Alzheimer's disease, and brain inflammatory reaction following traumatic brain injury; central nervous system disease, including spinal cord injury, or cerebral infarction; pathologic angiogenesis, including as may occur in primary and metastatic tumor growth;
rheumatoid arthritis; diabetic retinopathy, atherosclerosis; cancer; chronic pulmonary disease; acute lung injury; acute respiratory disease syndrome; sepsis; and the like.
One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to methods for treating an SlPl receptor- associated disorder in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof. One aspect of the present invention pertains to methods for treating a disease or disorder mediated by lymphocytes in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating an autoimmune disease or disorder in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating' an inflammatory disease or disorder in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating a microbial infection or disease in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating a viral infection or disease in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating cancer in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
Examples of cancer include, but are not limited to, lung cancer, colon cancer, breast cancer, prostate cancer, pancreatic cancer, and hepatocellular carcinoma.
One aspect of the present invention pertains to methods for treating a disorder in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof, wherein said disorder is selected from the group consisting of psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy, glomerulosclerosis, gastritis, polymyositis, thyroiditis, vitiligo, hepatitis, and biliary cirrhosis.
One aspect of the present invention pertains to methods for treating psoriasis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof. One aspect of the present invention pertains to methods for treating rheumatoid arthritis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating Crohn's disease in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating transplant rejection in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating multiple sclerosis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating systemic lupus erythematosus in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating ulcerative colitis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating type I diabetes in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating acne in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating myocardial ischemia-reperfusion injury in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating hypertensive nephropathy in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating glomerulosclerosis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating gastritis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating polymyositis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating thyroiditis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating vitiligo in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating hepatitis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to methods for treating biliary cirrhosis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of an SlPl receptor-associated disorder.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of a disease or disorder mediated by lymphocytes.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of an autoimmune disease or disorder.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of an inflammatory disease or disorder. One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of a microbial infection or disease.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of a viral infection or disease.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of cancer. Examples of cancer include, but are not limited to, lung cancer, colon cancer, breast cancer, prostate cancer, pancreatic cancer, and hepatocellular carcinoma.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of an SlPl receptor-associated disorder selected from the group consisting of psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy, glomerulosclerosis, gastritis, polymyositis, thyroiditis, vitiligo, hepatitis, and biliary cirrhosis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of psoriasis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of rheumatoid arthritis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of Crohn's disease.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of transplant rejection.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of multiple sclerosis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of systemic lupus erythematosus.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of ulcerative colitis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of type I diabetes.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of acne.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of myocardial ischemia- reperfusion injury. One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of hypertensive nephropathy.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of glomerulosclerosis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of gastritis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of polymyositis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of thyroiditis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of vitiligo.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of hepatitis.
One aspect of the present invention pertains to the use of compounds of the present invention in the manufacture of a medicament for the treatment of biliary cirrhosis.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of the human or animal body by therapy.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of an SlPl receptor-associated disorder.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of a disease or disorder mediated by lymphocytes.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of an autoimmune disease or disorder.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of an inflammatory disease or disorder.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of a microbial infection or disease.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of a viral infection or disease.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of cancer. Examples of cancer include, but are not limited to, lung cancer, colon cancer, breast cancer, prostate cancer, pancreatic cancer, and hepatocellular carcinoma.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of an SlPl receptor-associated disorder selected from the group consisting of psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy, glomerulosclerosis, gastritis, polymyositis, thyroiditis, vitiligo, hepatitis, and biliary cirrhosis.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of psoriasis.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of rheumatoid arthritis
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of Crohn's disease.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of transplant rejection.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of multiple sclerosis.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of systemic lupus erythematosus.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of ulcerative colitis.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of type I diabetes.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of acne.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of myocardial ischemia-reperfusion injury.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of hypertensive nephropathy.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of glomerulosclerosis.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of gastritis.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of polymyositis.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of thyroiditis.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of vitiligo.
One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of hepatitis. One aspect of the present invention pertains to compounds of the present invention for use in a method for the treatment of biliary cirrhosis.
One aspect of the present invention pertains to processes for preparing a composition comprising admixing a compound of the present invention and a pharmaceutically acceptable carrier.
These and other aspects of the invention disclosed herein will be set forth in greater detail as the patent disclosure proceeds.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the results of an experiment which measured the ability of the exemplified enantiomer of Compound 4 (Example 1.3) to lower the absolute count of peripheral lymphocytes in rats compared to vehicle.
Figure 2 shows a general synthetic scheme for the preparation of compounds of Formula (Ia) wherein E is CR1. The synthetic scheme shows the coupling of carboxylic acids or acid chlorides with N-hydroxy-carbamimidoyl intermediates and subsequent formation of the oxadiazole ring to provide ester intermediates. The esters can be converted to carboxylic acid using methods known in the art, for example, /-butyl esters are converted to carboxylic acids by treatment with an acid, such as TFA in the presence or absence of thioanisole or cysteine, methyl esters are converted to carboxylic acids by treatment with a base, such as LiOH, NaOH, KOH, and the like. The acid can then be converted to compounds of Formula (Ia) by using appropriate electrophiles known in the art, such as N-halosuccinimide.
Figure 3 shows alternative routes to give compounds of Formula (Ia) by introducing the R1 group before the ester is hydrolyzed to the carboxylic acid and by subsequently introducing the alkyl group through a halogen intermediate.
Figure 4 shows synthetic methods to give compounds of Formula (Ia) wherein R1 is alkylsulfonyl, cyano, or haloalkyl.
Figure 5 shows a general synthetic scheme for the preparation of compounds of Formula (Ia) wherein E is Ν.
DETAILED DESCRIPTION OF THE INVENTION DEFINITIONS
For clarity and consistency, the following definitions will be used throughout this patent document.
The term "agonist" is intended to mean a moiety that interacts with and activates a G- protein-coupled receptor, such as the SlPl receptor, such as can thereby initiate a physiological or pharmacological response characteristic of that receptor. For example, an agonist activates an intracellular response upon binding to the receptor, or enhances GTP binding to a membrane. In certain embodiments, an agonist of the invention is an SlPl receptor agonist that is capable of facilitating sustained SlPl receptor internalization (see e.g. , Matloubian et ai, Nature, 427, 355, 2004).
The term "antagonist" is intended to mean a moiety that competitively binds to the receptor at the same site as an agonist (for example, the endogenous ligand), but which does not activate the intracellular response initiated by the active form of the receptor and can thereby inhibit the intracellular responses by an agonist or partial agonist. An antagonist does not diminish the baseline intracellular response in the absence of an agonist or partial agonist.
The term "hydrate" as used herein means a compound of the invention or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of water bound by non- covalent intermolecular forces.
The term "solvate" as used herein means a compound of the invention or a salt, thereof, that further includes a stoichiometric or non-stoichiometric amount of a solvent bound by non- covalent intermolecular forces. Preferred solvents are volatile, non-toxic, and/or acceptable for administration to humans in trace amounts.
The term "in need of treatment" and the term "in need thereof when referring to treatment are used interchangeably to mean a judgment made by a caregiver (e.g. physician, nurse, nurse practitioner, etc. in the case of humans; veterinarian in the case of animals, including non-human mammals) that an individual or animal requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a caregiver's expertise, but that includes the knowledge that the individual or animal is ill, or will become ill, as the result of a disease, condition or disorder that is treatable by the compounds of the invention. Accordingly, the compounds of the invention can be used in a protective or preventive manner; or compounds of the invention can be used to alleviate, inhibit or ameliorate the disease, condition or disorder.
The term "individual" is intended to mean any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates and most preferably humans.
The term "inverse agonist" is intended to mean a moiety that binds to the endogenous form of the receptor or to the constitutively activated form of the receptor and which inhibits the " baseline intracellular response initiated by the active form of the receptor below the normal base level of activity which is observed in the absence of an agonist or partial agonist, or decreases GTP binding to a membrane. In some embodiments, the baseline intracellular response is inhibited in the presence of the inverse agonist by at least 30%. In some embodiments, the baseline intracellular response is inhibited in the presence of the inverse agonist by at least 50%. In some embodiments, the baseline intracellular response is inhibited in the presence of the inverse agonist by at least 75%, as compared with the baseline response in the absence of the inverse agonist. The term "modulate or modulating" is intended to mean an increase or decrease in the amount, quality, response or effect of a particular activity, function or molecule.
The term "pharmaceutical composition" is intended to mean a composition comprising at least one active ingredient; including but not limited to, salts, solvates, and hydrates of compounds of the present invention, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human). Those of ordinary skill in the art will understand and appreciate the techniques appropriate for determining whether an active ingredient has a desired efficacious outcome based upon the needs of the artisan.
The term "therapeutically effective amount" is intended to mean the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, caregiver or by an individual, which includes one or more of the following:
(1) Preventing the disease, for example, preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease;
(2) Inhibiting the disease, for example, inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or
symptomatology); and
(3) Ameliorating the disease, for example, ameliorating a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology). CHEMICAL GROUP, MOIETY OR RADICAL
The term "Ci-C6 acyl" is intended to mean a Q-Ce alkyl radical attached to the carbon of a carbonyl group wherein the definition of alkyl has the same definition as described herein; some examples include, but are not limited to, acetyl, propionyl, «-butanoyl, sec-butanoyl, pivaloyl, pentanoyl, and the like.
The term "Ci-C6 alkoxy" is intended to mean a Ci-C6 alkyl radical, as defined herein, attached directly to an oxygen atom. Some embodiments are 1 to 5 carbons, some embodiments are 1 to 4 carbons, some embodiments are 1 to 3 carbons and some embodiments are 1 or 2 carbons. Examples include methoxy, ethoxy, w-propoxy, isopropoxy, n-butoxy, te/7-butoxy, isobutoxy, sec-butoxy, and the like.
The term "Ci-C6 alkyl" is intended to mean a straight or branched carbon radical containing 1 to 6 carbons. Some embodiments are 1 to 5 carbons, some embodiments are 1 to 4 carbons, some embodiments are 1 to 3 carbons and some embodiments are 1 or 2 carbons. Examples of an alkyl include, but are not limited to, methyl, ethyl, /i-propyl, isopropyl, π-butyl, sec-butyl, isobutyl, ter/-butyl, pentyl, isopentyl, /ert-pentyl, πeø-pentyl, 1 -methylbutyl [i.e., -CH(CH3)CH2CH2CH3], 2-methylbutyl [i.e., -CH2CH(CH3)CH2CH3], n-hexyl, and the like.
The term "Ci-C6 alkylsulfonyl" is intended to mean a C]-C6 alkyl radical attached to the sulfur of a sulfone radical having the formula: -S(O)2- wherein the alkyl radical has the same definition as described herein. Examples include, but are not limited to, methylsulfonyl, ethylsulfonyl, n-propylsulfonyl, isopropylsulfonyl, n-butylsulfonyl, sec-butylsulfonyl, isobutylsulfonyl, tert-butylsulfonyl, and the like.
The term "cyano" is intended to mean the group -CN.
The term "C3-C7 cycloalkoxy" is intended to mean a saturated ring radical containing 3 to 7 carbons directly bonded to an oxygen atom. Some examples include cyclopropyl-O-, cyclobutyl-O-, cyclopentyl-O-, cyclohexyl-O-, and the like.
The term "C3-C7 cycloalkyl" is intended to mean a saturated ring radical containing 3 to 7 carbons. Some embodiments contain 3 to 6 carbons. Some embodiments contain 3 to 5 carbons. Some embodiments contain 5 to 7 carbons. Some embodiments contain 3 to 4 carbons. Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like.
The term "Ci-C6 haloalkoxy" is intended to mean a C)-C6 haloalkyl, as defined herein, which is directly attached to an oxygen atom. Examples include, but are not limited to, difluoromethoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy, pentafluoroethoxy, and the like.
The term "Ci-C6 haloalkyl" is intended to mean an Ci-C6 alkyl group, defined herein, wherein the alkyl is substituted with between one halogen up to folly substituted wherein a folly substituted Ci-C6 haloalkyl can be represented by the formula CzL2z+i wherein L is a halogen and "z" is 1, 2, 3, 4, 5, or 6. When more than one halogen is present, the halogens may be the same or different and selected from the group consisting of fluoro, chloro, bromo or iodo, preferably fluoro. Some embodiments are 1 to 5 carbons, some embodiments are 1 to 4 carbons, some embodiments are 1 to 3 carbons and some embodiments are 1 or 2 carbons. Examples of haloalkyl groups include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, chlorodifluoromethyl, 2,2,2-trifluoroethyl, pentafiuoroethyl, and the like.
The term "Ci-C6 haloacyl" is intended to mean a Ci-C6 haloalkyl radical attached to the carbon of a carbonyl group wherein the definition of haloalkyl has the same definition as described herein; some examples include, but are not limited to, 2,2,2-trifluoroacetyl, 3,3,3- trifluoropropanoyl, 2,2-difluoropropanoyl, 2,2,3,3,3-pentafluoropropanoyl, 3,3,3-trifluoro-2- (trifluoromethyl)propanoyl, and the like.
The term "halogen" or "halo" is intended to mean a fluoro, chloro, bromo or iodo group.
COMPOUNDS OF THE INVENTION: One aspect of the present invention pertains to certain compounds of Formula (Ia) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000021_0001
(Ia)
wherein:
E, G, R2, R4, and R5 have the same definitions as described herein , supra and infra.
It is understood that the present invention embraces compounds, solvates and/or hydrates of compounds, pharmaceutically acceptable salts of compounds, and solvates and/or hydrates of pharmaceutically acceptable salts of compounds, wherein the compounds are as described herein.
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination. All combinations of the embodiments pertaining to the chemical groups represented by the variables (e.g., E, G, R1, R2, R3, R4, and R5) contained within the generic chemical formulae described herein, for example, (Ia), (Ib), (Ic), (Ie), (Ig), (Ii), (Ik), (Qa), (IHa), (iπb), (HIc), (Hid), etc. are specifically embraced by the present invention just as if each and every combination was individually explicitly recited, to the extent that such combinations embrace stable compounds (i.e., compounds that can be isolated, characterized and tested for biological activity). In addition, all subcombinations of the chemical groups listed in the embodiments describing such variables, as well as all subcombinations of uses and medical indications described herein, are also specifically embraced by the present invention just as if each and every subcombination of chemical groups and subcombination of uses and medical indications was individually and explicitly recited herein.
As used herein, "substituted" indicates that at least one hydrogen atom of the chemical group is replaced by a non-hydrogen substituent or group. The non-hydrogen substituent or group can be monovalent or divalent. When the substituent or group is divalent, then it is understood that this group is further substituted with another substituent or group. When a chemical group herein is "substituted" it may have up to the full valence of substitution, for example, a methyl group can be substituted by 1, 2, or 3 substituents, a methylene group can be substituted by 1 or 2 substituents, a phenyl group can be substituted by 1, 2, 3, 4, or 5 substituents, a naphthyl group can be substituted by 1, 2, 3, 4, 5, 6, or 7 substituents and the like. Likewise, "substituted with one or more substituents" refers to the substitution of a group with one substituent up to the total number of substituents physically allowed by the group. Further, when a group is substituted with more than one substituent, the substituents can be identical or they can be different.
Compounds of the invention also include tautomeric forms, such as keto-enol tautomers and the like. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. It is understood that the various tautomeric forms are within the scope of the compounds of the present invention.
The present disclosure includes all isotopes of atoms occurring in the present compounds, intermediates, salts and crystalline forms thereof. Isotopes include those atoms having the same atomic number but different mass numbers. One aspect of the present invention includes every combination of one or more atoms in the present compounds, intermediates, salts, and crystalline forms thereof that is replaced with an atom having the same atomic number but a different mass number. One such example is the replacement of an atom that is the most naturally abundant isotope, such as 1H or 12C, found in one the present compounds,
intermediates, salts, and crystalline forms thereof, with a different atom that is not the most naturally abundant isotope, such as 2H or 3H (replacing 1H), or 11C, 13C, or 14C (replacing 12C). A compound wherein such a replacement has taken place is commonly referred to as being an isotopically-labeled compound. Isotopic-labeling of the present compounds, intermediates, salts, and crystalline forms thereof can be accomplished using any one of a variety of different synthetic methods know to those of ordinary skill in the art and they are readily credited with understanding the synthetic methods and available reagents needed to conduct such isotopic- labeling. By way of general example, and without limitation, isotopes of hydrogen include 2H (deuterium) and 3H (tritium). Isotopes of carbon include 11C, 13C, and 14C. Isotopes of nitrogen include 13N and 15N. Isotopes of oxygen include 150, 17O, and 18C. An isotope of fluorine includes 18F. An isotope of sulfur includes 35S. An isotope of chlorine includes 36Cl. Isotopes of bromine include 75Br, 76Br, 77Br, and 82Br. Isotopes of iodine include 123I, 1241, 125I, and 131I. Another aspect of the present invention includes compositions, such as, those prepared during synthesis, preformulation, and the like, and pharmaceutical compositions, such as, those prepared with the intent of using in a mammal for the treatment of one or more of the disorders described herein, comprising one or more of the present compounds, intermediates, salts, and crystalline forms thereof, wherein the naturally occurring distribution of the isotopes in the composition is perturbed. Another aspect of the present invention includes compositions and pharmaceutical compositions comprising compounds as described herein wherein the compound is enriched at one or more positions with an isotope other than the most naturally abundant isotope. Methods are readily available to measure such isotope perturbations or enrichments, such as, mass spectrometry, and for isotopes that are radio-isotopes additional methods are available, such as, radio-detectors used in connection with HPLC or GC. It is understood and appreciated that compounds of Formula (Ia) and formulae related thereto may have one or more chiral centers and therefore can exist as enantiomers and/or diastereomers. The invention is understood to extend to and embrace all such enantiomers, diastereomers and mixtures thereof: including but not limited to racemates. It is understood that Formula (Ia) and formulae used throughout this disclosure are intended to represent all individual enantiomers and mixtures thereof unless stated or shown otherwise.
The Variables E and G
In some embodiments, E is N.
In some embodiments, E is CR1.
In some embodiments, E is CR1; and G is CR3.
Some embodiments of the present invention pertain to compounds of Formula (Ig) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000023_0001
wherein each variable in Formula (Ig) has the same meaning as described herein, supra and infra.
In some embodiments, G is N.
In some embodiments, G is CR3.
In some embodiments, E is CR1; and G is N.
Some embodiments of the present invention pertain to compounds of Formula (Ii) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000023_0002
wherein each variable in Formula (Ii) has the same meaning as described herein, supra and infra.
In some embodiments, E is N; and G is CR3.
Some embodiments of the present invention pertain to compounds of Formula (Ik) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000024_0001
(Ik)
wherein each variable in Formula (Ik) has the same meaning as described herein, supra and infra.
The Group R1
In some embodiments, R1 is selected from the group consisting OfC1-C6 alkyl, C]-C6 alkylsulfonyl, Ci-C6 haloalkyl, Ci-C6 haloacyl, cyano, and halogen.
In some embodiments, R1 is selected from the group consisting of chloro, cyano, fluoro, iodo, methyl, methylsulfonyl, trifluoroacetyl, and trifluoromethyl.
In some embodiments, R1 is chloro or fluoro.
In some embodiments, R1 is chloro
In some embodiments, R1 is cyano.
In some embodiments, R1 is fluoro.
In some embodiments, R1 is iodo
In some embodiments, R1 is methyl.
In some embodiments, R1 is methylsulfonyl.
In some embodiments, R1 is trifluoroacetyl.
In some embodiments, R1 is trifluoromethyl. The Group R2
In some embodiments, R2 is selected from the group consisting of H, Ci-C6 alkoxy, Ci- C6 alkyl, cyano, C3-C7 cycloalkyl, C3-C7 cycloalkyloxy, Ci-C6 haloalkoxy, Ci-C6 haloalkyl, and halogen.
In some embodiments, R2 is selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, isopropoxy, methoxy, methyl, trifiuoromethoxy, and trifluoromethyl.
In some embodiments, R2 is selected from the group consisting of H, chloro, cyano, methoxy, methyl, trifiuoromethoxy, and trifluoromethyl.
In some embodiments, R2 is selected from the group consisting of cyano, methoxy, methyl, trifiuoromethoxy, and trifluoromethyl.
In some embodiments, R2 is selected from the group consisting of cyano, cyclohexyl, cyclopentyl, and isopropoxy.
In some embodiments, R2 is selected from the group consisting of CN and
trifiuoromethoxy. In some embodiments, R2 is selected from the group consisting of chloro, cyano, fluoro, methyl, and trifluoromethyl.
In some embodiments, R2 is H.
In some embodiments, R2 is cyano.
In some embodiments, R2 is fluoro.
In some embodiments, R2 is methoxy.
In some embodiments, R2 is methyl.
In some embodiments, R2 is chloro.
In some embodiments, R2 is trifluoromethyl.
In some embodiments, R2 is trifluoromethoxy.
The Group R3
In some embodiments, R3 is selected from the group consisting of H, C]-C6 alkoxy, C1- C6 alkyl, cyano, C3-C7 cycloalkyl, C3-C7 cycloalkyloxy, Ci-C6 haloalkoxy, Ci -C6 haloalkyl, and halogen.
In some embodiments, R3 is selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, isopropoxy, methoxy, methyl, trifluoromethoxy, and trifluoromethyl.
In some embodiments, R3 is selected from the group consisting of H, cyclohexyl, cyclopentyl, and isopropoxy.
In some embodiments, R3 is selected from the group consisting of cyano, isopropoxy, cyclopentyl, and cyclohexyl.
In some embodiments, R3 is H.
In some embodiments, R3 is cyano.
In some embodiments, R3 is cyclopentyl.
In some embodiments, R3 is cyclohexyl.
In some embodiments, R3 is isopropoxy.
In some embodiments, R3 is trifluoromethyl.
In some embodiments, R3 is trifluoromethoxy. The Group R4
In some embodiments, R4 is selected from the group consisting of H, Ci-C6 alkoxy, C1- C6 alkyl, cyano, C3-C7 cycloalkyl, C3-C7 cycloalkyloxy, Ci-C6 haloalkoxy, Q-C6 haloalkyl, and halogen.
In some embodiments, R4 is selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, isopropoxy, methoxy, methyl, trifluoromethoxy, and trifluoromethyl.
In some embodiments, R4 is selected from the group consisting of H, chloro, cyano, methoxy, methyl, trifluoromethoxy, and trifluoromethyl. In some embodiments, R4 is selected from the group consisting of cyano, methoxy, methyl, trifluoromethoxy, and trifluoromethyl.
In some embodiments, R4 is selected from the group consisting of cyano and trifluoromethoxy.
In some embodiments, R4 is selected from the group consisting of H, chloro, methoxy, trifluoromethoxy, and trifluoromethyl.
In some embodiments, R4 is H.
In some embodiments, R4 is cyano.
In some embodiments, R4 is methoxy.
In some embodiments, R4 is methyl.
In some embodiments, R4 is chloro.
In some embodiments, R4 is trifluoromethyl.
In some embodiments, R4 is trifluoromethoxy. The Group R5
In some embodiments, R5 is H or halogen.
In some embodiments, R5 is H or fluoro.
In some embodiments, R5 is H.
In some embodiments, R5 is fluoro.
Certain Combinations
In some embodiments, R2, R3, and R4 are each independently selected from the group consisting of H, C1-C6 alkoxy, C]-C6 alkyl, cyano, C3-C7 cycloalkyl, C3-C7 cycloalkyloxy, Ci-C6 haloalkoxy, C]-C6 haloalkyl, and halogen.
In some embodiments, R2, R3, and R4 are each independently selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, fluoro, iodo, isopropoxy, methoxy, methyl, trifluoromethoxy, and trifluoromethyl.
In some embodiments, R2, R3, and R4 are each independently selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, isopropoxy, methoxy, methyl, trifluoromethoxy, and trifluoromethyl.
In some embodiments, R2 and R4 are each independently selected from the group consisting of H, chloro, cyano, methoxy, methyl, trifluoromethoxy, and trifluoromethyl.
In some embodiments, R2 and R4 are each independently selected from the group consisting of cyano, methoxy, methyl, trifluoromethoxy, and trifluoromethyl; and R3 is H.
In some embodiments, R2 and R4 are each independently Ci-C6 alkyl or halogen.
In some embodiments, R2 and R4 are each independently chloro or methyl. In some embodiments, R2 and R3 are each independently selected from the group consisting of cyano, isopropoxy, cyclohexyl, and cyclopentyl; and R4 is H.
In some embodiments, R2 is cyano; R3 is isopropoxy, cyclohexyl, or cyclopentyl; and R4 is H.
In some embodiments, R1 is selected from the group consisting of chloro, cyano, fluoro, iodo, methyl, methylsulfonyl, trifluoroacetyl, and trifluoromethyl;
R2, R3, and R4 are each independently selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, fluoro, iodo, isopropoxy, methoxy, methyl, trifluoromethoxy, and trifluoromethyl; and
R5 is H or F.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ia) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000027_0001
wherein:
E is N or CR1;
G is N or CR3;
R1 is selected from the group consisting of methyl, trifluoromethyl, cyano,
trifluoroacetyl, methylsulfonyl, chloro, iodo, and fluoro;
R2 is selected from the group consisting of H, chloro, cyano, methoxy, methyl, trifluoromethoxy, and trifluoromethyl;
R3 is selected from the group consisting of H, cyclohexyl, cyclopentyl, and isopropoxy; R4 is selected from the group consisting of H, chloro, cyano, methoxy, methyl, trifluoromethoxy, and trifluoromethyl; and
R4 is H or fluoro.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ib) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000028_0001
wherein:
R1 is selected from the group consisting Of C]-C6 alkyl, Ci-Ce alkylsulfonyl, Ci- C6 haloalkyl, Ci-C6 haloacyl, cyano, and halogen; and
R2, R3, and R4 are each independently selected from the group consisting of H,
Ci-C6 alkoxy, cyano, C3-C7 cycloalkyl, Ci-C6 haloalkoxy, Ci-C6 haloalkyl, and halogen.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ib) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000028_0002
wherein:
R1 is selected from the group consisting of chloro, cyano, fluoro, iodo, methyl, methylsulfonyl, trifluoroacetyl, and trifluoromethyl; and
R2, R3, and R4 are each independently selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, fluoro, iodo, isopropoxy, methoxy,
methylsulfonyl, trifluoromethoxy, and trifluoromethyl.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ib) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000028_0003
wherein:
R1 is selected from the group consisting of chloro, cyano, fluoro, iodo, methyl, methylsulfonyl, trifluoroacetyl, and trifluoromethyl; R2 is selected from the group consisting of H, chloro, cyano, fluoro, methoxy, trifluoromethoxy, and trifluoromethyl;
R3 is selected from the group consisting of H, cyclohexyl, cyclopentyl, and isopropoxy; and
R4 is selected from the group consisting of H, chloro, cyano, fluoro, methoxy, trifluoromethoxy, and trifluoromethyl.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ic) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000029_0001
wherein:
R1 is selected from the group consisting Of C1-C6 alkyl, C1-C6 alkylsulfonyl, Q C6 haloalkyl, cyano, and halogen; and
R2 and R4 are each independently selected from the group consisting of cyano,
CJ-C6 alkoxy, C1-C6 haloalkoxy, Ci-C6 haloalkyl, and halogen.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ic) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000029_0002
wherein:
R1 is selected from the group consisting of methyl, trifluoromethyl, cyano, methylsulfonyl, chloro, iodo, and fluoro;
R2 and R4 are each independently selected from the group consisting of chloro, cyano, methoxy, trifluoromethoxy, and trifluoromethyl. Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ic) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000030_0001
wherein:
R1 is selected from the group consisting of methyl, trifluoromethyl, cyano, methylsulfonyl, chloro, iodo, and fluoro;
R2 is cyano; and
R4 is trifluoromethoxy.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ie) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000030_0002
wherein:
R1 is selected from the group consisting of Q-C6 alkyl, Ci-C6 alkylsulfonyl, Cr C6 haloalkyl, Q-C6 haloacyl, cyano, and halogen; and
R2 and R3 are each independently selected from the group consisting of CpC6 alkoxy, cyano, C3-C7 cycloalkyl, and halogen.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ie) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000030_0003
wherein: R1 is Ci-C6 haloacyl or halogen; and
R2 and R3 are each independently selected from the group consisting OfCj-C6 alkoxy, cyano, and C3-C7 cycloalkyl.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ie) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000031_0001
wherein:
R1 is Ci-C6 haloacyl or halogen;
R2 is cyano, CrC6 haloalkyl, or halogen; and
R3 is C1-C6 alkoxy or C3-C7 cycloalkyl.
Some embodiments of the present invention pertain to compounds selected from compounds of Formula (Ie) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000031_0002
wherein:
R1 is chloro or trifluoroacetyl;
R2 is cyano, fluoro, or trifluoromethyl; and
R3 is cyclohexyl, cyclopentyl, isopropoxy, or methoxy.
Some embodiments of the present invention pertain to compounds of Formula (DIa) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000031_0003
wherein:
E is N or CR1;
G is N or CR3;
R1 is selected from the group consisting of Q-C6 alkyl, C1-C6 alkylsulfonyl, C1-C6 haloalkyl, C1-C6 haloacyl, cyano, and halogen;
R2, R3, and R4 are each independently selected from the group consisting of H, C1-C6 alkoxy, C1-C6 alkyl, cyano, C3-C7 cycloalkyl, C3-C7 cycloalkyloxy, C1-C6 haloalkoxy, C1-C6 haloalkyl, and halogen;
R5 is H or halogen; and
R6 is H or C1-C6 alkyl.
Some embodiments of the present invention pertain to compounds of Formula (HIb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000032_0001
wherein each variable in Formula (TOb) has the same meaning as described herein, supra and infra.
Some embodiments of the present invention pertain to compounds of Formula (Hie) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000032_0002
wherein:
E is N or CR1;
G is N or CR3;
R1 is selected from the group consisting Of C1-C6 alkyl, CrC6 alkylsulfonyl, C1-C6 haloalkyl, C1-C6 haloacyl, cyano, and halogen;
R2, R3, and R4 are each independently selected from the group consisting of H, CrC6 alkoxy, C1-C6 alkyl, cyano, C3-C7 cycloalkyl, C3-C7 cycloalkyloxy, Q-C6 haloalkoxy, C1-C6 haloalkyl, and halogen;
R5 is H or halogen; and
R6 is H or C1-C6 alkyl. Some embodiments of the present invention pertain to compounds of Formula (HId) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000033_0001
(HId)
wherein each variable in Formula (Illd) has the same meaning as described herein, supra and infra.
Esters and Prodrugs
One aspect of the present invention pertains to compounds selected from compounds of Formula (Ha) and pharmaceutically acceptable salts, solvates, and hydrates thereof, useful as synthetic intermediates in the preparation of compounds of Formula (Ia) and/or useful as prodrugs for the oral delivery of compounds of Formula (Ia):
Figure imgf000033_0002
wherein:
E, G, R2, R4, and R5 have the same definitions as described herein, supra and infra, and R6 is CrC6 alkyl.
One aspect of the present invention pertains to compounds selected from compounds of Formula (Ha) and pharmaceutically acceptable salts, solvates, and hydrates thereof.
In some embodiments, R6 is methyl.
In some embodiments, R6 is ter/-butyl.
It is appreciated that all of the embodiments described herein, supra and infra, that relate to the common variables shared between compounds of Formula (Ia) and (Ha) namely, E, G, R2, R4, and R5, apply to compounds of Formula (TIa) just as if they were each individually disclosed herewith with specific reference to Formula (Ila).
One aspect of the present invention pertains to compounds of Formula (Ila) as synthetic intermediates useful in the preparation of compounds of Formula (Ia).
One aspect of the present invention pertains to compounds of Formula (Ila) as esters of compounds, described and shown herein, such as compounds in Table A, where R6 is methyl.
One aspect of the present invention pertains to compounds of Formula (Ila) as esters of compounds, described and shown herein, such as compounds in Table A, where R is ter/-butyl. One aspect of the present invention pertains to compounds of Formula (Ila) as prodrugs useful for the oral delivery of compounds of Formula (Ia).
One aspect of the present invention pertains to compounds of Formula (Ila) useful as prodrugs of compounds of Formula (Ia).
Some embodiments of the present invention include every combination of one or more compounds selected from the following group shown in Table A.
Table A
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
C(I) Ring Carbon Stereochemistry
Certain compounds of the present invention contain the fused tricyclic system referred to as 2,3-dihydro-lH-pyrrolo[l,2-a]indole. Certain other compounds of the present invention contain the fused tricyclic system referred to as 2,3-dihydro-lH-benzo[d]pyrrolo[l,2- a]imidazole. Present on one of the πngs of both of the fused tricyclic systems is a -CH2CO2H group. The nng carbon to which the -CH2CO2H acid group is bonded is referred to herein as the C(I) nng carbon. It is understood that the stereochemistry for the C(I) nng carbon contained in the 2,3-dihydro-lH-pyrrolo[l,2-a]mdole or 2,3-dihydro-lH-benzo[d]pyrrolo[l,2-a]imidazole ring system can be either R or S, see Formulae (Ilia) through (Illd) for specific designation of C(I).
A. C(I) Ring Carbon R Stereochemistry
In some embodiments, the stereochemistry for the C(I) ring carbon is R.
Some embodiments of the present invention include every combination of one or more compounds selected from the following group and pharmaceutically acceptable salts, solvates, and hydrates thereof: (R)-2-(7-(5-(3-cyano-4-isopropoxyphenyl)-l,2,4-oxadiazol-3-yl)-9-(2,2,2- trifluoroacetyl)-2,3-dihydro-lH-pyrrolo[l ,2-a]indol-l -yl)acetic acid; (/?)-2-(7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-fluoro-2,3-dihydro-lH-pyrrolo[l,2-a]indol- l-yl)acetic acid; (R)-2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-l,9- difluoro-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (R)-2-(9-chloro-7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (R)-2-(9-chloro-7-(5-(3-cyano-4-cyclohexylphenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH- pyrrolo[l ,2-a]indol-l -yl)acetic acid; (i?)-2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l ,2,4- oxadiazol-3-yl)-9-iodo-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (Λ)-2-(7-(5-(3- cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-methyl-2,3-dihydro-lH-pyrrolo[l,2- a]indol-l-yl)acetic acid; (i?)-2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3- yl)-9-(trifluoromethyl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (R)-2-(9-cyano-7- (5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2- a]indol-l-yl)acetic acid; (R)-2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3- yO^^methylsulfonyO^^-dihydro-lH-pyrrolofl^-alindol-l-yOacetic acidj ^^^V^S^.S- bis(trifluoromethyl)phenyl)-l,2,4-oxadiazol-3-yl)-9-chloro-2,3-dihydro-lH-pyrrolo[l,2-a]indol- l-yl)acetic acid; (R)-2-(9-chloro-7-(5-(4-cyclopentyl-3-(trifluoromethyl)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (R)-2-(9-chloro-7-(5-(2- chloro-ό-methylpyridin^-y^-l^^-oxadiazol-S-y^^^-dihydro-lH-pyrrolotl^-alindol-l- yl)acetic acid; (R)-2-(9-chloro-7-(5-(3-cyano-5-methoxyphenyl)-l ,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (R)-2-(6-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-benzo[d]pyrrolo[l,2- a]imidazol-3-yl)acetic acid; (/?)-2-(9-chloro-7-(5-(3-chloro-5-(trifluoromethoxy) phenyl)- 1,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (R)-2-(9-chloro-7-(5-(3- fluoro-4-methoxyphenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; and (R)-2-(9-chloro-7-(5-(4-isopropoxy-3-methoxyphenyl)-l ,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid. B. C(I) Ring Carbon "5" Stereochemistry
In some embodiments, the stereochemistry for the C(I) ring carbon is S. Some embodiments of the present invention include every combination of one or more compounds selected from the following group and pharmaceutically acceptable salts, solvates, and hydrates thereof: (S)-2-(7-(5-(3-cyano-4-isopropoxyphenyl)-l,2,4-oxadiazol-3-yl)-9-(2,2,2- trifluoroacetyO^jS-dihydro-lH-pyrrolofl^-ajindol-l-yOacetic acid; (5)-2-(7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-fluoro-2,3-dihydro-lH-pyiτolo[l,2-a]indol- 1 -yl)acetic acid; (S)-2-(7-(5 -(3 -cyano-5-(trifluoromethoxy)phenyl)- 1 ,2,4-oxadiazol-3 -yl)- 1 ,9- difluoro-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (S)-2-(9-chloro-7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (5)-2-(9-chloro-7-(5-(3-cyano-4-cyclohexylphenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetic acid; (S)-2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4- oxadiazol^-yO^-iodo^^-dihydro-lH-pyrrolofl^^indol-l-yOacetic acid; (S)-2-(7-(5-(3- cyano-5-(trifluoromethoxy)phenyl)-l,2>4-oxadiazol-3-yl)-9-methyl-2,3-dihydro-lH-pyrrolo[l,2- a]indol-l -yl)acetic acid; (5)-2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l ,2,4-oxadiazol-3- yl)-9-(trifluoromethyl)-2,3-dihydro-lH-pyrrolo[l ,2-a]indol-l -yl)acetic acid; (S)-2-(9-cyano-7- (5 -(3 -cyano-5 -(trifluoromethoxy)phenyl)- 1 ,2 ,4-oxadiazol-3 -yl)-2 ,3 -dihydro- lH-pyrrolo [1,2- a]indol-l -yl)acetic acid; (5)-2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l ,2,4-oxadiazol-3- yl)-9-(methylsulfonyl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (5)-2-(7-(5-(3,5- bis(trifluoromethyl)phenyl)-l,2,4-oxadiazol-3-yl)-9-chloro-2,3-dihydro-lH-pyrrolo[l,2-a]indol- l-yl)acetic acid; (5)-2-(9-chloro-7-(5-(4-cyclopentyl-3-(trifluoromethyl)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (5)-2-(9-chloro-7-(5-(2- chloro-6-methylpyridin-4-yl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetic acid; (5)-2-(9-chloro-7-(5-(3-cyano-5-methoxyphenyl)-l ,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (5)-2-(6-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-benzo[d]pyrrolo[l,2- a]imidazol-3-yl)acetic acid; (5)-2-(9-chloro-7-(5-(3-chloro-5-(trifluoromethoxy) phenyl)- 1,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; (5)-2-(9-chloro-7-(5-(3- fluoro-4-methoxyphenyl)- 1 ,2,4-oxadiazol-3 -yl)-2,3 -dihydro- lH-pyrrolo[ 1 ,2-a]indol- 1 -yl)acetic acid; and (.S)-2-(9-chloro-7-(5-(4-isopropoxy-3-methoxyphenyl)-l ,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid.
Additionally, individual compounds and chemical genera of the present invention, for example, those compounds found in Table A including diastereomers and enantiomers thereof, encompass all pharmaceutically acceptable salts, solvates, and hydrates, thereof.
It is understood that the present invention embraces each diastereomer, each enantiomer and mixtures thereof of each compound and generic formulae disclosed herein just as if they were each individually disclosed with the specific stereochemical designation for each chiral carbon. Separation of the individual isomers (such as, by chiral ΗPLC, recrystallization of diastereomeπc mixtures, and the like) or selective synthesis (such as, by enantiomeπc selective syntheses, and the like) of the individual isomers is accomplished by application of vaπous methods which are well known to practitioners in the art.
The compounds of the Formula (Ia) of the present invention may be prepared according to relevant published literature procedures that are used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter in the working examples.
Protection and deprotection may be earned out by procedures generally known in the art (see, for example, Greene, T. W. and Wuts, P. G. M., Protecting Groups in Organic Synthesis, 3rd Edition, 1999 [Wiley]).
PHARMACEUTICAL COMPOSITIONS
A further aspect of the present invention pertains to pharmaceutical compositions comprising one or more compounds as descπbed herein and one or more pharmaceutically acceptable earners. Some embodiments pertain to pharmaceutical compositions compnsing a compound of the present invention and a pharmaceutically acceptable earner.
Some embodiments of the present invention include a method of producing a pharmaceutical composition compnsmg admixing at least one compound according to any of the compound embodiments disclosed herein and a pharmaceutically acceptable earner.
Formulations may be prepared by any suitable method, typically by uniformly mixing the active compound(s) with liquids or finely divided solid earners, or both, in the required proportions and then, if necessary, forming the resulting mixture into a desired shape.
Conventional excipients, such as binding agents, fillers, acceptable wetting agents, tabletting lubncants and disintegrants may be used m tablets and capsules for oral
administration. Liquid preparations for oral administration may be in the form of solutions, emulsions, aqueous or oily suspensions and syrups. Alternatively, the oral preparations may be in the form of dry powder that can be reconstituted with water or another suitable liquid vehicle before use. Additional additives such as suspending or emulsifying agents, non-aqueous vehicles (including edible oils), preservatives and flavonngs and colorants may be added to the liquid preparations. Parenteral dosage forms may be prepared by dissolving the compound of the invention m a suitable liquid vehicle and filter stenlizing the solution before filling and sealing an appropnate vial or ampule. These are just a few examples of the many appropnate methods well known m the art for prepanng dosage forms.
A compound of the present invention can be formulated into pharmaceutical compositions using techniques well known to those in the art. Suitable pharmaceutically acceptable earners, outside those mentioned herein, are known in the art; for example, see
Remington, The Science and Practice of Pharmacy, 20th Edition, 2000, Lippincott Williams & Wilkms, (Editors: Gennaro et al.) While it is possible that, for use in the prophylaxis or treatment, a compound of the invention may, in an alternative use, be administered as a raw or pure chemical, it is preferable however to present the compound or active ingredient as a pharmaceutical formulation or composition further comprising a pharmaceutically acceptable carrier.
The invention thus further provides pharmaceutical formulations comprising a compound of the invention or a pharmaceutically acceptable salt, solvate, hydrate or derivative thereof together with one or more pharmaceutically acceptable carriers thereof and/or prophylactic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not overly deleterious to the recipient thereof.
Pharmaceutical formulations include those suitable for oral, rectal, nasal, topical
(including buccal and sub-lingual), vaginal or parenteral (including intramuscular, subcutaneous and intravenous) administration or in a form suitable for administration by inhalation, insufflation or by a transdermal patch. Transdermal patches dispense a drug at a controlled rate by presenting the drug for absorption in an efficient manner with a minimum of degradation of the drug. Typically, transdermal patches comprise an impermeable backing layer, a single pressure sensitive adhesive and a removable protective layer with a release liner. One of ordinary skill in the art will understand and appreciate the techniques appropriate for manufacturing a desired efficacious transdermal patch based upon the needs of the artisan.
The compounds of the invention, together with a conventional adjuvant, carrier, or diluent, may thus be placed into the form of pharmaceutical formulations and unit dosages thereof and in such form may be employed as solids, such as tablets or filled capsules, or liquids such as solutions, suspensions, emulsions, elixirs, gels or capsules filled with the same, all for oral use; in the form of suppositories for rectal administration; or in the form of sterile injectable solutions for parenteral (including subcutaneous) use. Such pharmaceutical compositions and unit dosage forms thereof may comprise conventional ingredients in conventional proportions, with or without additional active compounds or principles and such unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
For oral administration, the pharmaceutical composition may be in the form of, for example, a tablet, capsule, suspension or liquid. The pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient.
Examples of such dosage units are capsules, tablets, powders, granules or suspensions, with conventional additives such as lactose, mannitol, corn starch or potato starch; with binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators such as corn starch, potato starch or sodium carboxymethyl-cellulose; and with lubricants such as talc or magnesium stearate. The active ingredient may also be administered by injection as a composition wherein, for example, saline, dextrose or water may be used as a suitable pharmaceutically acceptable carrier.
Compounds of the present invention or a salt, solvate, hydrate or physiologically functional derivative thereof can be used as active ingredients in pharmaceutical compositions, specifically as SlPl receptor modulators. The term "active ingredient" is defined in the context of a "pharmaceutical composition" and is intended to mean a component of a pharmaceutical composition that provides the primary pharmacological effect, as opposed to an "inactive ingredient" which would generally be recognized as providing no pharmaceutical benefit.
The dose when using the compounds of the present invention can vary within wide limits and as is customary and known to the physician, it is to be tailored to the individual conditions in each individual case. It depends, for example, on the nature and severity of the illness to be treated, on the condition of the patient, on the compound employed or on whether an acute or chronic disease state is treated or prophylaxis is conducted or on whether further active compounds are administered in addition to the compounds of the present invention. Representative doses of the present invention include, but are not limited to, about 0.001 mg to about 5000 mg, about 0.001 mg to about 2500 mg, about 0.001 mg to about 1000 mg, 0.001 mg to about 500 mg, 0.001 mg to about 250 mg, about 0.001 mg to 100 mg, about 0.001 mg to about 50 mg and about 0.001 mg to about 25 mg. Multiple doses may be administered during the day, especially when relatively large amounts are deemed to be needed, for example 2, 3 or 4 doses. Depending on the individual and as deemed appropriate by the patient's physician or caregiver it may be necessary to deviate upward or downward from the doses described herein.
The amount of active ingredient or an active salt, solvate or hydrate derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will ultimately be at the discretion of the attendant physician or clinician. In general, one skilled in the art understands how to extrapolate in vivo data obtained in one model system, typically an animal model, to another, such as a human. In some circumstances, these extrapolations may merely be based on the weight of the animal model in comparison to another, such as a mammal, preferably a human, however, more often, these extrapolations are not simply based on weights, but rather incorporate a variety of factors. Representative factors include the type, age, weight, sex, diet and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetic and toxicology profiles of the particular compound employed, whether a drug delivery system is utilized, whether an acute or chronic disease state is being treated or prophylaxis is conducted or whether further active compounds are administered in addition to the compounds of the present invention and as part of a drug combination. The dosage regimen for treating a disease condition with the compounds and/or compositions of this invention is selected in accordance with a variety factors including those cited above. Thus, the actual dosage regimen employed may vary widely and therefore may deviate from a preferred dosage regimen and one skilled in the art will recognize that dosage and dosage regimens outside these typical ranges can be tested and, where appropriate, may be used in the methods of this invention.
The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as 2, 3, 4 or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations. The daily dose can be divided, especially when relatively large amounts are administered as deemed appropriate, into several, for example 2, 3 or 4 part administrations. If appropriate, depending on individual behavior, it may be necessary to deviate upward or downward from the daily dose indicated.
For preparing pharmaceutical compositions from the compounds of the present invention, the suitable pharmaceutically acceptable carrier can be either solid, liquid or a mixture of both. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories and dispersible granules. A solid carrier can be one or more substances which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or encapsulating materials.
In powders, the carrier is a finely divided solid which is in a mixture with the finely divided active component.
In tablets, the active component is mixed with the carrier having the necessary binding capacity in suitable proportions and compacted to the desired shape and size.
The powders and tablets may contain varying percentages of the active compound. A representative amount in a powder or tablet may be from 0.5 to about 90 percent of the active compound. However, an artisan would know when amounts outside of this range are necessary. Suitable carriers for powders and tablets include magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter and the like. The term "preparation" is intended to include the formulation of the active compound with encapsulating material as carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it. Similarly, cachets and lozenges are included. Tablets, powders, capsules, pills, cachets and lozenges can be used as solid forms suitable for oral administration.
For preparing suppositories, a low melting wax, such as an admixture of fatty acid glycerides or cocoa butter, is first melted and the active component is dispersed homogeneously therein {e.g., by stirring). The molten homogenous mixture is then poured into convenient sized molds, allowed to cool and thereby to solidify. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
Liquid form preparations include solutions, suspensions and emulsions, for example, water or water-propylene glycol solutions. For example, parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution. Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of iηjectables.
The compounds according to the present invention may thus be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-fϊlled syringes, small volume infusion or in multi-dose containers with an added preservative. The pharmaceutical compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
Alternatively, the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
Aqueous formulations suitable for oral use can be prepared by dissolving or suspending the active component in water and adding suitable colorants, flavors, stabilizing and thickening agents, as desired.
Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well-known suspending agents.
Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration. Such liquid forms include solutions, suspensions and emulsions. These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents and the like.
For topical administration to the epidermis the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch. Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
Formulations suitable for topical administration in the mouth include lozenges comprising the active agent in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray. The formulations may be provided in single or multi-dose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension. In the case of a spray, this may be achieved for example by means of a metering atomizing spray pump.
Administration to the respiratory tract may also be achieved by means of an aerosol formulation in which the active ingredient is provided in a pressurized pack with a suitable propellant. If the compounds of the present invention or pharmaceutical compositions comprising them are administered as aerosols (e.g., nasal aerosols, by inhalation), this can be carried out, for example, using a spray, a nebulizer, a pump nebulizer, an inhalation apparatus, a metered inhaler or a dry powder inhaler. Pharmaceutical forms for administration of the compounds of the present invention as an aerosol can be prepared by processes well known to the person skilled in the art. Solutions or dispersions of the compounds of the present invention or a pharmaceutically acceptable salt, solvate, hydrate or derivative thereof in water, water/alcohol mixtures or suitable saline solutions, for example, can be employed using customary additives (e.g., benzyl alcohol or other suitable preservatives), absorption enhancers for increasing the bioavailability, solubilizers, dispersants and others and, if appropriate, customary propellants (e.g., carbon dioxide, CFCs, such as, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane and the like). The aerosol may conveniently also contain a surfactant such as lecithin. The dose of drug may be controlled by provision of a metered valve.
In formulations intended for administration to the respiratory tract, including intranasal formulations, the compound will generally have a small particle size for example of the order of 10 microns or less. Such a particle size may be obtained by means known in the art, for example by micronization. When desired, formulations adapted to give sustained release of the active ingredient may be employed.
Alternatively the active ingredients may be provided in the form of a dry powder (e.g., a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP)). Conveniently the powder carrier will form a gel in the nasal cavity. The powder composition may be presented in unit dose form (e.g., capsules, cartridges) as for gelatin or blister packs from which the powder may be administered by means of an inhaler.
The pharmaceutical preparations are preferably in unit dosage forms. In such form, the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
In some embodiments, the compositions are tablets or capsules for oral administration.
In some embodiments, the compositions are liquids for intravenous administration. The compounds according to the invention may optionally exist as pharmaceutically acceptable salts including pharmaceutically acceptable acid addition salts prepared from pharmaceutically acceptable non-toxic acids including inorganic and organic acids.
Representative acids include, but are not limited to, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, dichloroacetic, formic, fumaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic,
methanesulfonic, mucic, nitric, oxalic, pamoic, pantothenic, phosphoric, succinic, sulfiric, tartaric, oxalic, />-toluenesulfonic and the like, such as those pharmaceutically acceptable salts listed by Berge et al, Journal of Pharmaceutical Sciences, 66: 1-19 (1977).
The acid addition salts may be obtained as the direct products of compound synthesis. In the alternative, the free base may be dissolved in a suitable solvent containing the appropriate acid and the salt isolated by evaporating the solvent or otherwise separating the salt and solvent. The compounds of this invention may form solvates with standard low molecular weight solvents using methods known to the skilled artisan.
Compounds of the present invention can be converted to "pro-drugs." The term "prodrugs" refers to compounds that have been modified with specific chemical groups known in the art and that when administered into an individual undergo biotransformation to give the parent compound. Pro-drugs can thus be viewed as compounds of the invention containing one or more specialized non-toxic protective groups used in a transient manner to alter or to eliminate a property of the compound. In one general aspect, the "pro-drug" approach is utilized to facilitate oral absorption. A thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems Vol. 14 of the A.C.S. Symposium Series; and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
Some embodiments of the present invention include a method of producing a pharmaceutical composition for "combination-therapy" comprising admixing at least one compound according to any of the compound embodiments disclosed herein, together with at least one known pharmaceutical agent as described herein and a pharmaceutically acceptable carrier.
It is noted that when SlPl receptor agonists are utilized as active ingredients in a pharmaceutical composition, these are not intended for use only in humans, but in other non- human mammals as well. Indeed, recent advances in the area of animal health-care mandate that consideration be given for the use of active agents, such as SlPl receptor agonists, for the treatment of an SlPl receptor-associated disease or disorder in companionship animals (e.g., cats, dogs, etc.) and in livestock animals (e.g., cows, chickens, fish, etc.). Those of ordinary skill in the art are readily credited with understanding the utility of such compounds in such settings.
Hydrates and Solvates
It is understood that when the phrase "pharmaceutically acceptable salts, solvates, and hydrates" is used when referring to a particular formula herein, it is intended to embrace solvates and/or hydrates of compounds of the particular formula, pharmaceutically acceptable salts of compounds of the particular formula and solvates and hydrates of pharmaceutically acceptable salts of compounds of the particular formula. It is understood by one of ordinary skill in the art that hydrates is a subset of solvates.
The compounds of the present invention can be administrated in a wide variety of oral and parenteral dosage forms. It will be apparent to those skilled in the art that the following dosage forms may comprise, as the active component, either a compound of the invention or a pharmaceutically acceptable salt or as a solvate or hydrate thereof. Moreover, various hydrates and solvates of the compounds of the invention and their salts will find use as intermediates in the manufacture of pharmaceutical compositions. Typical procedures for making and identifying suitable hydrates and solvates, outside those mentioned herein, are well known to those in the art; see for example, pages 202-209 of KJ. Guillory, "Generation of Polymorphs, Hydrates, Solvates, and Amorphous Solids," in: Polymorphism in Pharmaceutical Solids, ed. Harry G. Brittan, Vol. 95, Marcel Dekker, Inc., New York, 1999. Accordingly, one aspect of the present invention pertains to hydrates and solvates of compounds of Formula (Ia) or Formula (Ila) and/or their pharmaceutical acceptable salts, as described herein, that can be isolated and characterized by methods known in the art, such as, thermogravimetric analysis (TGA), TGA- mass spectroscopy, TGA-Infrared spectroscopy, powder X-ray diffraction (XRPD), Karl Fisher titration, high resolution X-ray diffraction, and the like. There are several commercial entities that provide quick and efficient services for identifying solvates and hydrates on a routine basis. Example companies offering these services include Wilmington PharmaTech (Wilmington, DE), Avantium Technologies (Amsterdam) and Aptuit (Greenwich, CT). POLYMOPHS AND PSEUDOPOL YMORPHS
Polymorphism is the ability of a substance to exist as two or more crystalline phases that have different arrangements and/or conformations of the molecules in the crystal lattice.
Polymorphs show the same properties in the liquid or gaseous state but they behave differently in the solid state.
Besides single-component polymorphs, drugs can also exist as salts and other multicomponent crystalline phases. For example, solvates and hydrates may contain an API host and either solvent or water molecules, respectively, as guests. Analogously, when the guest compound is a solid at room temperature, the resulting form is often called a cocrystal. Salts, solvates, hydrates, and cocrystals may show polymorphism as well. Crystalline phases that share the same API host, but differ with respect to their guests, may be referred to as
pseudopolymorphs of one another.
Solvates contain molecules of the solvent of crystallization in a definite crystal lattice. Solvates, in which the solvent of crystallization is water, are termed hydrates. Because water is a constituent of the atmosphere, hydrates of drugs may be formed rather easily.
By way of example, Stahly recently published a polymorph screen of 245 compounds consisting of a "wide variety of structural types" that revealed about 90% of the compounds exhibiting multiple solid forms. Overall, approximately half the compounds were polymorphic, often having one to three forms. About one-third of the compounds formed hydrates, and about one-third formed solvates. Data from cocrystal screens of 64 compounds showed that 60% formed cocrystals other than hydrates or solvates. (G. P. Stahly, Crystal Growth & Design (2007), 7(6), 1007-1026.)
OTHER UTILITIES
Another object of the present invention relates to radiolabeled compounds of the present invention that are useful not only in radio-imaging but also in assays, both in vitro and in vivo, for localizing and quantitating the SlPl receptor in tissue samples, including human and for identifying SlPl receptor ligands by inhibition binding of a radiolabeled compound. It is a further object of this invention to develop novel SlPl receptor assays which comprise such radiolabeled compounds.
The present invention embraces isotopically-labeled compounds of the present invention. Isotopically or radiolabeled compounds are those which are identical to compounds disclosed herein, but for the fact that one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number most commonly found in nature. Suitable radionuclides that may be incorporated in compounds of the present invention include, but are not limited, to 2H (also written as D for deuterium), 3H (also written as T for tritium), 11C, 13C, 14C, 13N, 15N, 150, 170, 18O, 18F, 35S, 36Cl, 75Br, 76Br, 77Br, 82Br, 1231, 1241, 125I, and 131I. The radionuclide that is incorporated in the instant radiolabeled compounds will depend on the specific application of that radiolabeled compound. For example, for in vitro SlPl receptor labeling and competition assays, compounds that incorporate 3H, 14C, 82Br, 1251, 131I, or 35S will generally be most useful. For radio-imaging applications 11C, 18F, 125I, 1231, 1241, 1311, 75Br, 76Br, or 77Br will generally be most useful.
It is understood that a "radiolabeled " or "labeled compound" is a compound of Formula (Ia), (Ib), (Ic), (Ie), (Ig), (Ii), (Ik), (TIa), (HIa), (HIb), (HIc), (HId), etc. containing at least one radionuclide. In some embodiments the radionuclide is selected from the group consisting of 3H, 14C, 1251 , 35S, and 82Br.
Certain isotopically-labeled compounds of the present invention are useful in compound and/or substrate tissue distribution assays. In some embodiments the radionuclide 3H and/or 14C isotopes are useful in these studies. Further, substitution with heavier isotopes such as deuterium {i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability {e.g. , increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in Figures 2 to 5 and Examples infra, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent. Other synthetic methods that are useful are discussed infra. Moreover, it should be understood that all of the atoms represented in the compounds of the invention can be either the most commonly occurring isotope of such atoms or a scarcer radio-isotope or nonradioactive isotope.
Synthetic methods for incorporating radio-isotopes into organic compounds are applicable to compounds of the invention and are well known in the art. Certain synthetic methods, for example, for incorporating activity levels of tritium into target molecules, are as follows:
A. Catalytic Reduction with Tritium Gas: This procedure normally yields high specific activity products and requires halogenated or unsaturated precursors.
B. Reduction with Sodium Borohydride [3H]: This procedure is rather inexpensive and requires precursors containing reducible functional groups such as aldehydes, ketones, lactones, esters and the like.
C. Reduction with Lithium Aluminum Hydride [3H]: This procedure offers products at almost theoretical specific activities. It also requires precursors containing reducible functional groups such as aldehydes, ketones, lactones, esters, and the like.
D. Tritium Gas Exposure Labeling: This procedure involves exposing precursors containing exchangeable protons to tritium gas in the presence of a suitable catalyst.
E. N-Methylation using Methyl Iodide [3H]: This procedure is usually employed to prepare 0-methyl or N-methyl [3H] products by treating appropriate precursors with high specific activity methyl iodide [3H]. This method in general allows for higher specific activity, such as for example, about 70-90 Ci/mmol.
Synthetic methods for incorporating activity levels of 125I into target molecules include:
A. Sandmeyer and like reactions: This procedure transforms an aryl amine or a heteroaryl amine into a diazonium salt, such as a diazonium tetrafluoroborate salt and subsequently to 125I labeled compound using Na125I. A represented procedure was reported by Zhu, G-D. and co-workers in J. Org. Chem., 2002, 67, 943-948.
B. Ortho 125Iodination of phenols: This procedure allows for the incorporation of 125I at the ortho position of a phenol as reported by Collier, T. L. and co-workers in J. Labelled Compd. Radiopharm., 1999, 42, S264-S266.
C. Aryl and heteroaryl bromide exchange with 125I: This method is generally a two step process. The first step is the conversion of the aryl or heteroaryl bromide to the corresponding tri-alkyltin intermediate using for example, a Pd catalyzed reaction [i.e. Pd(Ph3P)4] or through an aryl or heteroaryl lithium, in the presence of a tri-alkyltinhalide or hexaalkylditin [e.g., (CH3)3SnSn(CH3)3]. A representative procedure was reported by Le Bas, M.-D. and co-workers in J. Labelled Compd. Radiopharm. 2001, 44, S280-S282.
A radiolabeled SlPl receptor compound of Formula (Ia) or Formula (Ila) can be used in a screening assay to identify/evaluate compounds. In general terms, a newly synthesized or identified compound (i.e., test compound) can be evaluated for its ability to reduce binding of the "radiolabeled compound of Formula (Ia) or Formula (Ila)" to the SlPl receptor.
Accordingly, the ability of a test compound to compete with the "radiolabeled compound of Formula (Ia) or Formula (Ha)" for the binding to the SlPl receptor directly correlates to its binding affinity.
The labeled compounds of the present invention bind to the SlPl receptor. In one embodiment the labeled compound has an IC50 less than about 500 μM, in another embodiment the labeled compound has an IC50 less than about 100 μM, in yet another embodiment the labeled compound has an IC50 less than about 10 μM, in yet another embodiment the labeled compound has an IC50 less than about 1 μM and in still yet another embodiment the labeled inhibitor has an IC50 less than about 0.1 μM.
Other uses of the disclosed receptors and methods will become apparent to those of skill in the art based upon, inter alia, a review of this disclosure.
As will be recognized, the steps of the methods of the present invention need not be performed any particular number of times or in any particular sequence. Additional objects, advantages and novel features of this invention will become apparent to those skilled in the art upon examination of the following examples thereof, which are intended to be illustrative and not intended to be limiting. EXAMPLES
Example 1: Syntheses of Compounds of the Present Invention.
Illustrated syntheses for compounds of the present invention are shown in Figures 2 through 5 where the variables have the same definitions as used throughout this disclosure.
The compounds of the invention and their syntheses are further illustrated by the following examples. The following examples are provided to further define the invention without, however, limiting the invention to the particulars of these examples. The compounds described herein, supra and infra, are named according to the AutoNom version 2.2, or CS ChemDraw Ultra Version 9.0.7. In certain instances common names are used and it is understood that these common names would be recognized by those skilled in the art.
Chemistry: Proton nuclear magnetic resonance (1H NMR) spectra were recorded on a Bruker Avance-400 equipped with a QNP (Quad Nucleus Probe) or a BBI (Broad Band Inverse) and z-gradient. Proton nuclear magnetic resonance (1H NMR) spectra were also recorded on a Bruker Avance-500 equipped a BBI (Broad Band Inverse) and z-gradient. Chemical shifts are given in parts per million (ppm) with the residual solvent signal used as reference. NMR abbreviations are used as follows: s = singlet, d = doublet, dd = doublet of doublets, dt = doublet of triplets, ddd = doublet of doublet of doublets, t = triplet, q = quartet, m = multiplet, bs = broad singlet, br = broad. Microwave irradiations were carried out using a Smith Synthesizer™ or an Emrys Optimizer™ (Biotage). Thin-layer chromatography (TLC) was performed on silica gel 60 F254 (Merck), preparatory thin-layer chromatography (prep TLC) was preformed on PK6F silica gel 60 A 1 mm plates (Whatman) and column chromatography was carried out on a silica gel column using Kieselgel 60, 0.063-0.200 mm (Merck). Evaporation was done under reduced pressure on a Bϋchi rotary evaporator. Celite® 545 was used for filtration of palladium.
LCMS spec: HPLC-pumps: LC-IOAD VP, Shimadzu Inc.; HPLC system controller:
SCL-IOA VP, Shimadzu Inc; UV-Detector: SPD-IOA VP, Shimadzu Inc; Autosampler: CTC HTS, PAL, Leap Scientific; Mass spectrometer: API 150EX with Turbo Ion Spray source, AB/MDS Sciex; Software: Analyst 1.2. Example 1.1: Preparation of 2-(7-(5-(3-Cyano-5-(trifluoromethoxy)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indoI-l-yl)acetic Acid (Intermediate 1).
Step A: Preparation of 7-Bromo-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-one.
To a solution of ethyl 5-bromo-lH-indole-2-carboxylate (30 g, 112 mmol) in toluene (500 mL) was added portionwise sodium hydride (60% dispersion in mineral oil, 9.40 g, 235 mmol). Vigorous gas evolution was observed. The resulting white suspension was heated to 110 °C. Butyl acrylate (35.1 mL, 246 mmol) was added dropwise (using a syringe pump) over 24 h while stirring vigorously at an internal temperature of 110 0C. Additional butyl acrylate (10 mL) was added in one portion and stirring was continued at 110 0C for 4 h followed by additional sodium hydride (60% dispersion in mineral oil, 5 g) and butyl acrylate (10 mL). 4 h later another 6 mL of butyl acrylate was added. Stirring was continued at 110 0C for a total of 48 h. The reaction was cooled in an ice-bath, and 2 M HCl (400 mL) was added carefully. The layers were separated and the aqueous layer was extracted with dichloromethane (2 x 200 mL). The combined organic extracts were washed with brine, dried over MgSO4, filtered, and
concentrated. The resulting orange residue was dissolved in acetic acid (900 mL) and water (100 mL). The orange solution was refluxed for 16 h. The solvents were removed under reduced pressure. To the residue was added dichloromethane (300 mL). The resulting precipitate was collected by filtration and rinsed twice with dichloromethane to provide the title compound. LCMS m/z = 250.2 [M+H]+; 1H NMR (400 MHz, DMSO-rfβ) δ ppm 3.20 (t, J= 6.1 Hz, 2H), 4.46 (t, 7= 6.1 Hz, 2H), 6.92 (s, IH), 7.46 (dd, J= 8.8, 1.8 Hz, IH), 7.63 (d, 7= 8.8 Hz, IH), 7.98 (d, J= 2.0 Hz, IH).
Step B: Preparation of l-Oxo-2,3-dihydro-lH-pyrrolo[l,2-a]indole-7-carbonitrile. To a suspension of 7-bromo-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-one (6.7 g, 26.8 mmol) in N-methylpyrrolidinone (20 mL) was added CuCN (3.12 g, 34.8 mmol). The mixture was heated under microwave irradiation in a sealed, thick-walled glass tube at 200 0C for 2 h. The reaction was diluted with dichloromethane (400 mL). CeIi te® was added and the mixture was stirred vigorously for 5 min. The solids were removed by filtration. The dichloromethane was removed under reduced pressure. The remaining brown liquid was poured into water (800 mL). The precipitate was collected by filtration, rinsed with water, and dried under reduced pressure to give the title compound as a beige solid (5.16 g). LCMS m/z = 197.0 [M+Η]+.
Step C: Preparation of terf-Butyl 2-(7-Cyano-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- ylidene)acetate.
To a solution of l-oxo-2,3-dihydro-lH-pyrrolo[l,2-a]indole-7-carbonitrile (3.30 g,
16.82 mmol) in TΗF (50 mL) was added (terΝbutoxycarbonylmethylene)triphenylphosphorane (15.83 g, 42.0 mmol). The mixture was stirred at 65 0C for 22 h and concentrated. The residue was purified by silica gel flash chromatography to provide the title compound as a pale yellow solid. LCMS m/z = 295.1 [M+Η]+.
Step D: Preparation of tert-Butyl 2-(7-Cyano-2,3-dihydro-lH-pyrrolo[l,2-a]indoI-l- yl)acetate.
tert-Butyl 2-(7-cyano-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-ylidene)acetate (4.6 g, 15.63 mmol) was dissolved in EtOAc (100 mL). The flask was flushed with nitrogen gas.
Degussa wet (50 wt% water) 10% Pd/C (1.663 g, 1.563 mmol) was added and the mixture was agitated on a Parr shaker under 70 psi hydrogen for 16 h. The mixture was filtered though
Celite® and concentrated. To the residue was added further 10% Pd/C and the hydrogenation was repeated at 70 psi for 6 h. The reaction mixture was filtered and concentrated to provide the title compound as an orange solid. LCMS m/z = 297.3 [M+H]+.
Step E: Preparation of te/t-Butyl 2-(7-(5-(3-Cyano-5-(trifluoromethoxy)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetate.
To a solution of tert-Buty\ 2-(7-cyano-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate
(4.95 g, 16.70 mmol) in ethanol (60 mL) was added a 50 wt% aqueous solution of
hydroxylamine (10.24 mL, 167 mmol). The resulting solution was stirred in a sealed flask at 70 0C for 4 h. The solution was concentrated under reduced pressure to provide the title compound as a yellow solid. LCMS m/z = 330.4 [M+Η]+.
Step F: Preparation of 3-Cyano-5-(trifluoromethoxy)benzoyl Chloride.
To a solution of 3-cyano-5-(trifluoromethoxy)benzoic acid (11.0 g, 47.6 mmol) in dichloromethane (100 mL) was added DMF (10 drops). The solution was cooled in an ice-bath and a 2.0 M dichloromethane solution of oxalyl chloride (29.7 mL, 59.5 mmol) was added over 5 min. After addition was complete, the cooling bath was removed and the mixture was stirred at 23 0C for 4 h. The solvent was removed under reduced pressure to provide the title
compound. 1H NMR (400 MHz, CDCl3) δ ppm 7.82 (s, IH), 8.17 (s, IH), 8.36 (s, IH).
Step G: Preparation of tert-Butyl 2-(7-(5-(3-Cyano-5-(trifluoromethoxy)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetate.
To a solution of tert-butyl 2-(7-(N'-hydroxycarbamimidoyl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetate (7.95 g, 24.14 mmol) in dioxane (175 mL) was added 3-cyano- 5-(trifiuoromethoxy)benzoyl chloride (6.02 g, 24.14 mmol) and triethylamine (11.77 mL, 84 mmol). The resulting suspension was stirred at 75 0C for 3.5 h. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in dichloromethane, washed with 0.5 M HCl (200 mL), and then brine. The organic layer was dried over MgSO4, filtered, and concentrated under reduced pressure to provide a beige solid. Crystallization from
dichloromethane/hexanes yielded the title compound as a white solid. LCMS m/z = 525.5
[M+Η]+; 1H ΝMR (400 MHz, CDCl3) δ ppm 1.50 (s, 9H), 2.29-2.38 (m, IH), 2.56 (dd, J = 15.7, 8.1 Hz, IH), 2.77 (dd, J= 15.9, 6.8 Hz, IH), 2.88-2.96 (m, IH), 3.76 (quintet, J= 7.5 Hz, IH), 4.06-4.12 (m, IH), 4.18-4.23 (m, IH), 6.29 (s, IH), 7.34 (d, J= 8.6 Hz, IH), 7.72 (s, IH), 7.93 (dd, J= 8.6, 1.5 Hz, IH), 8.31 (s, IH), 8.39 (s, IH), 8.47 (t, J= 1.3 Hz, IH).
Step H: Preparation of 2-(7-(5-(3-Cyano-5-(trifiuoromethoxy)phenyl)-l,2,4- oxadiazoI-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic Acid.
To a solution oftert-butyl 2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate (0.55 g, 1.049 mmol) and thioanisole (1.232 mL, 10.49 mmol) in dichloromethane (5 mL) was added trifluoroacetic acid (3.12 mL, 41.9 mmol). The resulting suspension was stirred at 23 °C for 3 h. The volatiles were removed under reduced pressure. The residue was taken up in hexanes and stirred overnight. The resulting solid was collected by filtration and rinsed with hexanes to provide a racemic mixture of the title compound. LCMS m/z = 469.3 [M+H]+; 1H NMR (400 MHz, DMSO-J6) δ ppm 2.23-2.32 (m, IH), 2.63 (dd, J = 16.4, 8.1 Hz, IH), 2.77 (dd, J = 16.7, 6.8 Hz, IH), 2.78- 2.87 (m, IH), 3.65 (quintet, J= 7.5 Hz, IH), 4.05-4.11 (m, IH), 4.21-4.27 (m, IH), 6.31 (s, IH), 5 7.50 (d, J= 8.8 Hz, IH), 7.80 (dd, J= 8.6, 1.5 Hz, IH), 8.27 (d, J= 1.0 Hz, IH), 8.43 (s, 2H), 8.66 (t, J= 1.3 Hz, IH), 12.39 (s, IH).
Resolution via Chiral HPLC.
Column: normal phase preparative ChiralPak AD-H, 250 x 20 mm ID, 5 μm particle size Eluent: 100% acetonitrile with 0.05% trifluoroacetic acid
10 Gradient: isocratic
Flow: 13 mL/min
Detector: 280 nm
Retention Times: 1st enantiomer: 7.9 min; 2nd enantiomer: 14.9 min
15 Example 1.2: Preparation of 2-(7-(5-(3-Cyano-4-isopropoxyphenyl)-l,2,4-oxadiazol-3-yl)-9- (2,2,2-trifluoroacetyl)-2,3-dihydro-Lf/-pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 1).
Step A: Preparation of Isopropyl 3-Bromo-4-isopropoxybenzoate.
A mixture of 3-bromo-4-hydroxybenzoic acid (20.00 g, 92 mmol), cesium carbonate (90 g, 276 mmol) 2-bromopropane (21.61 mL, 230 mmol) in DMF (200 mL) were stirred at 80 0C
™ for 16 h in a 500 mL round bottomed flask. After cooling down, the reaction mixture was
filtered by vacuum filtration though Celite® and washed with EtOAc (3 x 100 mL). The solvent was removed under reduced pressure. The residue was taken up in EtOAc, washed with water (3 x 100 mL), dried (MgSO4), filtered by vacuum filtration through a glass fiber paper and concentrated under reduced pressure. The residue was purified by column chromatography to
25 give the title compound (24.3 g) as a colorless oil. LCMS m/z = 301.3 [M+H]+; 1H NMR (400 MHz, DMSO-^6) δ ppm 1.34 (m, 12H), 4.76-4.85 (m, IH), 5.09-5.18 (m, IH), 7.22 (d, J= 8.97 Hz, IH), 7.92 (dd, J= 8.72, 2.15 Hz, IH), 8.09 (d, J= 2.15 Hz, IH).
Step B: Preparation of Isopropyl 3-Cyano-4-isopropoxybenzoate.
Isopropyl 3-bromo-4-isopropoxybenzoate (23.8 g, 79 mmol) and CuCN (9.20 g, 103
™ mmol) in anhydrous N-methyl-2-pyrrolidinone (158 mL) were heated to 200 0C for 16 h in a 500 mL round bottomed flask. After 16 h the reaction mixture was poured into 1 M HCl and extracted into EtOAc (3 x 200 mL). The organic layers were combined and washed with 1 M HCl (3 x 200 mL) and washed with saturated NaCl (1 x 200 mL), dried (MgSO4), filtered by vacuum filtration through a glass fiber paper and concentrated under reduced pressure. The residue was purified by column chromatography to give the title compound (17.46 g) as a tan solid. LCMS m/z = 248.1 [M+H]+; 1H ΝMR (400 MHz, DMSO-^6) δ ppm 1.34 (d, J= 6.19 Hz, 6H), 1 38 (d, J= 6.06 Hz, 6H), 4.88-4.96 (m, IH), 5.10-5.18 (m, IH), 7.39 (d, J= 8.84 Hz, IH), 8.12-8.21 (m, 2H).
Step C: Preparation of 3-Cyano-4-isopropoxybenzoic Acid.
To a solution of isopropyl 3-cyano-4-isopropoxybenzoate (17.46 g, 70.6 mmol) in dioxane (100 mL) was added LiOH (1.0 M, 212 mL, 212 mmol). The reaction was stirred at 30 0C for 1.5 h in a 1 L round bottomed flask. After 1.5 h, the reaction was cooled down to room temperature and poured into 1 M HCl and extracted with EtOAc (3 x 200 mL). The organic layers were combined, washed with saturated NaCl (I x 200 mL), dried (MgSO4), and filtered by vacuum filtration through a glass fiber paper. The solvent was removed under reduced pressure to give the title compound (14 g) as a solid. LCMS m/z = 206.1 [M+H]+; 1H NMR (400 MHz, DMSO-d6) δ ppm 1.36 (d, J = 6.06 Hz, 6H), 4.85-4.96 (m, IH), 7.37 (d, J= 8.72 Hz, IH), 8.12-8.22 (m, 2H), 13.17 (s, IH).
Step D: Preparation of terf-Butyl 2-(7-(5-(3-Cyano-5-isopropoxyphenyl)-l,2,4- oxadiazoI-3-yl)-2,3-dihydro-li/-pyrrolo[l,2-a]indol-l-yl)acetate.
To a solution of tert-buty\ 2-(7-(Nl-hydroxycarbamimidoyl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetate (0.148 g, 0.45 mmol) in dioxane (2.71 mL) and 3-cyano-4- lsopropoxybenzoic acid (0.120 g, 0.585 mmol) was added TEA (0.627 mL, 4.50 mmol) followed by 1-propylphosphonic acid cyclic anhydride (0.292 mL, 0.495 mmol). The reaction was heated to reflux for 2 h. The mixture was diluted with EtOAc, washed with water (3x) and bπne. The organics were dried over MgSO4 and concentrated. The residue was purified by column chromatography to give a white solid. LCMS m/z = 499.8 [M+Η]+.
Step E: Preparation of 2-(7-(5-(3-Cyano-4-isopropoxyphenyl)-l,2,4-oxadiazol-3-yl)- 9-(2,2,2-trifluoroacetyl)-23-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic Acid.
To a solution of tert-buty\ 2-(7-(5-(3-cyano-4-isopropoxyphenyl)-l,2,4-oxadiazol-3-yl)- 2,3-dihydro-lH-pyrrolo[l,2-a]mdol-l-yl)acetate (0.465 g, 0.933 mmol) in CH2Cl2 (3.11 mL) was added tπisopropylsilane (1.918 mL, 9.33 mmol) and TFA (3.59 mL, 46.6 mmol). The reaction was stirred overnight and concentrated under reduced pressure. The residue was purified via preparative LCMS, to give the title compound as a pink solid. LCMS m/z = 539.2 [M+H]+; 1H ΝMR (400 MHz, DMSO-J6) δ ppm 1.39 (d, J= 6.06 Hz, 6H), 2.40-2.47 (m, IH), 2.70-2.83 (m, 2H), 2.87-3.00 (m, IH), 3.91-4.01 (m, IH), 4.38 (t, J= 8.40 Hz, 2H), 4.92-5.03 (m, IH), 7.56 (d, J= 9.22 Hz, IH), 7.79 (d, J= 8.46 Hz, IH), 8.07 (dd, J= 8.53, 1.58 Hz, IH), 8.43 (dd, J= 9.03, 2.21 Hz, IH), 8.52 (d, J= 2.27 Hz, IH), 8.79 (s, IH), 12.45 (s, IH).
Example 1.3: Preparation of an Enantiomer of 2-(9-Chloro-7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yI)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetic Acid (Compound 4).
The 2nd enantiomer of Intermediate 1 (as descπbed m Example 1.1, eluting at 14.9 min from chiral ΗPLC) (0.117 g, 0.25 mmol) was dissolved in anhydrous TΗF (4.0 mL). The reaction mixture was cooled to 0 0C and added l-chloropyrrolidine-2,5-dione (0.035 g, 0.263 mmol). The reaction mixture was warmed to 25 0C and stirred for 18 h. The mixture was diluted with MTBE (100 mL), washed with 2 M sodium thiosulfate (20 mL), water (20 mL), and brine (20 mL). The organics were dried over MgSO4 and concentrated under reduced pressure to give the title compound (0.119 g) as a yellow solid. LCMS m/z = 502.8 [M+H]+; 1H NMR (400 MHz, DMSO-40 δ ppm 2.30-2.43 (m, IH), 2.63 (dd, J= 16.48, 9.54 Hz, IH), 2.82-2.94 (m, IH), 2.99 (dd, J= 16.42, 4.17 Hz, IH), 3.69-3.84 (m, IH), 4.07-4.19 (m, IH), 4.24-4.33 (m, IH), 7.58 (d, J= 8.59 Hz, IH), 7.90 (dd, J= 8.59, 1.52 Hz, IH), 8.18 (d, J= 1.14 Hz, IH), 8.40 (s, IH), 8.45 (s, IH), 8.69 (t, J= 1.33 Hz, IH), 12.40 (bs, IH).
Example 1.4: Preparation of 2-(9-Chloro-7-(5-(3-cyano-4-cyclohexyIphenyI)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 5).
Step A: Preparation of tert-Butyl 2-(7-(5-(3-Cyano-4-cyclohexylphenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetate.
3-Cyano-4-cyclohexylbenzoic acid (33 mg, 0.14 mmol) was dissolved in DMA (1.0 mL) and CDI (46.7 mg, 0.29 mmol) was added. The reaction was warmed to 60 0C and stirred for 1 h. tert-Buty\ 2-(7-(N'-hydroxycarbamimidoyl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate (47.4 g, 144 mmol) was added and the reaction was stirred at 90 0C for 3 h. The reaction was concentrated to an oil and purified by column chromatography to give the title compound (22.6 mg). LCMS m/z = 523.3 [M+Η]+.
Step B: Preparation of 2-(7-(5-(3-Cyano-4-cyclohexylphenyl)-l,2,4-oxadiazol-3-yl)- 2,3-dihydro-l/f-pyrroIo[l,2-a]indol-l-y.)acetic Acid.
tert-Butyl 2-(7-(5-(3-cyano-4-cyclohexylphenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetate (22.6 mg, 0.043 mmol) was dissolved in DCM (0.5 mL) and D/L-cysteine (16 mg, 0.130 mmol) was added. The reaction mixture was cooled to 0 0C and TFA (0.223 mL, 2.90 mmol) was added. The reaction was stirred at room temperature for 6 h. The DCM was removed under reduced pressure and the remaining solution was diluted with water causing a white precipitate to form. The solids were collected by filtration, rinsed with water, and dried to give the title compound (18 mg). LCMS m/z = 467.4 [M+Η]+; 1H ΝMR (400 MHz, DMSO-40 δ ppm 1.08-1.65 (m, 5H), 1.71-1.80 (m, IH), 1.81-1.92 (m, 4H), 2.22-2.34 (m, IH), 2.62 (dd, J= 16.4, 7.70, IH), 2.72-2.87 (m, 2H), 2.92-3.02 (m, IH), 3.65 (quintet, J= 7.5 Hz, IH), 4.04-4.13 (m, IH), 4.20-4.27 (m, IH), 6.31 (s, IH), 7.49 (d, J= 8.6 Hz, IH), 7.78-7.85 (m, 2H), 8.28 (s, IH), 8.42 (d, J= 8.3 Hz, IH), 8.53 (s, IH), 12.3 (bs, IH).
Step C: Preparation of 2-(9-Chloro-7-(5-(3-cyano-4-cyclohexylphenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indoI-l-yl)acetic Acid.
2-(7-(5-(3-Cyano-4-cyclohexylphenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetic acid (12.8 mg, 0.027 mmol) was dissolved in DCM (1.0 mL), and cooled to 0 0C. NCS (3.66 mg, 0.027 mmol) was added and the reaction was stirred for 30 minutes. The reaction was diluted with water and the DCM layer was washed two additional times with water. The organic layer was dried over sodium sulfate, filtered, and concentrated to give the title compound (13.0 mg). LCMS m/z = 501.4 [M+H]+; 1H NMR (400 MHz, DMSO-^5) δ ppm 1.10-1.63 (m, 5H), 1.80-1.86 (m, IH), 1.89-2.01 (m, 4H), 2.38-2.48 (m, IH), 2.67 (dd, J = 16.8, 10.2 Hz, IH), 2.98-3.13 (m, 2H), 3.35 (dd, J = 16.8, 3.8 Hz, IH), 3.83-3.92 (m, IH), 4.06-4.14 (m, IH), 4.20-4.27 (m, IH), 7.33 (d, J = 8.5 Hz, IH), 7.58 (d, J = 8.3 Hz, IH), 8.00 (dd, J= 8.6, 1.6Hz, IH), 8.37 (dd, J = 8.2, 1.8 Hz, IH), 8.41 (d, J= 1.3Hz, IH), 8.50 (d, J= 1.8 Hz, IH).
Example 1.5: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-fluoro-2,3-dihydro-lH-pyrrolo[l,2- a]indol-l-yl)acetic Acid (Compound 2).
To a solution of the 1st enantiomer of Intermediate 1 (as described in Example 1.1, eluting at 7.9 min from chiral HPLC) (50 mg, 0.107 mmol) in DMF (1 mL) was added 1- fluoropyridinium trifluoromethanesulfonate (23.8 mg, 0.096 mmol). The reaction was stirred at 23 0C for 4 h then at 40 0C for 18 h. The mixture was purified by preparative HPLC to give the title compound (7 mg) as a white solid. LCMS m/z = 487.2 [M+H]+; 1H NMR (400 MHz, DMSCwZ6) δ ppm 2.28-2.36 (m, IH), 2.67 (dd, J= 16.2, 8.2 Hz, IH), 2.78-2.87 (m, 2H), 3.77- 3.84 (m, IH), 4.06-4.13 (m, IH), 4.22-4.28 (m, IH), 7.53 (dd, J = 8.7, 2.0 Hz, IH), 7.86 (dd, J= 8.6, 1.6 Hz, IH), 8.24 (d, J = 1.0 Hz, IH), 8.40-8.41 (m, IH), 8.44-8.45 (m, IH), 8.69 (t, J= 1.4 Hz, IH), 11.54 (bs, IH).
Example 1.6: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-l,9-difluoro-2β-dihydro-lH-pyrrolo[l,2- a]indol-l-yl)acetic Acid (Compound 3).
To a suspension of the 1st enantiomer of Intermediate 1 (as described in Example 1.1, eluting at 7.9 min from chiral HPLC) (50 mg, 0.107 mmol) in dimethoxyethane (1 mL) was added 1 -fluoropyridinium trifluoromethanesulfonate (33 mg, 0.134 mmol). The reaction was stirred at 55 0C for 4 h. The mixture was purified by preparative HPLC to give the title compound (10.8 mg) as a pink solid. LCMS m/z = 505.3 [M+H]+; 1H NMR (400 MHz, DMSO- d6) δ ppm 1.94-2.02 (m, IH), 2.24-2.33 (m, IH), 2.79 (dd, J= 18.6, 3.4 Hz, IH), 3.09 (dd, J = 18.6, 9.4 Hz, IH), 3.33-3.41 (m, IH), 3.68-3.74 (m, IH), 7.22 (d, J= 8.5 Hz, 1H),8.18 (d, J = 1.3 Hz, IH), 8.26 (dd, J= 8.5, 1.3 Hz, IH), 8.42-8.46 (m, 2H), 8.71 (t, J= 1.4 Hz, IH). Example 1.7: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-iodo-23-dihydro-lH-pyrrolo[l,2- a]indol-l-yl)acetic Acid (Compound 6).
Step A: Preparation of an Enantiomer of Methyl 2-(7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate.
The 2nd enantiomer of Intermediate 1 (as described in Example 1.1, eluting at 14.9 min from chiral HPLC) (0.5 g, 1.067 mmol) was suspended in MeOH (3 mL) and toluene (3 mL) solution. TMS-diazomethane (2 M solution in hexanes) (0.854 mL, 1.708 mmol) was slowly added to the mixture at room temperature. It slowly became a clear brown solution during addition. The reaction mixture was stirred for another 30 min. The solvent was removed under reduced pressure to give the title compound as a light pink solid (475 mg). LCMS m/z = 483.1 [M+H]+; 1H NMR (400 MHz, CDCl3) 2.30-2.38 (m, IH), 2.65 (dd, J = 16.2, 8.4 Hz, IH), 2.88 (dd, J= 16.1, 6.4 Hz, IH), 2.92-2.98 (m, IH), 3.76 (s, 3H), 3.75-3.85 (m, IH), 4.06-4.14 (m, IH), 4.18-4.27 (m, IH), 6.29 (s, IH), 7.34 (d, J = 8.5 Hz, IH), 7.72 (s, IH), 7.94 (dd, J= 8.5 and 1.6 Hz, IH), 8.31 (s, IH), 8.39 (d, J = 1.4 Hz, IH), 8.46 (t, J= 1.3 Hz, IH).
Step B: Preparation of an Enantiomer of Methyl 2-(7-(5-(3-Cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-iodo-2,3-dihydro-l//-pyrrolo[l,2- a] indol-l-yl)acetate.
To a solution of the above-mentioned enantiomer of methyl 2-(7-(5-(3-cyano-5-
(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate (470 mg, 0.974 mmol) in DCM (8 mL) was slowly added l-iodopyrrolidine-2,5-dione (230 mg, 1.023 mmol) at 0 0C. The reaction mixture was stirred at that temperature for 30 min, then diluted with DCM. The organic layer was washed with aqueous sodium thiosulfate solution, water and dried. The solvent was evaporated to give the title compound as light brown solid (548 mg). LCMS m/z = 609.4 [M+Ηf; 1H NMR (400 MHz, CDCl3) 2.39-2.48 (m, IH), 2.60 (dd, J= 16.5 and 10.6 Hz, IH), 2.96-3.05 (m, IH), 3.32 (dd, J= 16.4 and 3.6 Hz, IH), 3.75 (s, 3H), 3.75-3.82 (m, IH), 4.12-4.18 (m, IH), 4.22-4.30 (m, IH), 7.32 (d, J= 8.5 Hz, IH), 7.72 (s, IH), 7.90 (dd, J= 8.5 and 1.6 Hz, IH), 8.21 (d, J= 1.5 Hz, IH), 8.32 (s, IH), 8.48 (t, J= 1.3 Hz, IH).
Step C: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5-
(trifluoromethoxy)phenyl)-l,2,4-oxadiazoI-3-yl)-9-iodo-2,3-dihydro-l//-pyrrolo[l,2- a]indol-l-yl)acetic Acid.
The above-mentioned enantiomer of methyl 2-(7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-iodo-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate (75 mg, 0.123 mmol) in dioxane (1 mL) was added 1 M LiOH aqueous solution (0.493 mmol, 0.493 mL) at room temperature. The reaction mixture was stirred at the temperature for 3 h, diluted with water, and acidified with 0.5 M citric acid aqueous solution to pH 4. The solid collected was purified by preparative HPLC to give the title compound (50 mg). LCMS m/z = 594.9 [M+H]+; 1H NMR (400 MHz, DMSO-J6) 2.35-2.43 (m, IH), 2.60 (dd, J = 16.5, 10.3 Hz, IH), 2.85-2.94 (m, IH), 3.05 (dd, J= 16.5, 3.4 Hz, IH), 3.60-3.68 (m, IH), 4.14- 4.20 (m, IH), 4.25-4.34 (m, IH), 7.55 (d, J= 8.5 Hz, IH), 7.90 (dd, J= 8.5, 1.5 Hz, IH), 8.00 (d, J= 1.3 Hz, IH), 8.40 (s, IH), 8.44 (s, IH), 8.69 (t, J= 1.3 Hz, IH), 12.40 (bs, IH).
Example 1.8: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-methyl-2,3-dihydro-l//-pyrrolo[l,2- a]indol-l-yl)acetic Acid (Compound 7).
Step A: Preparation of an Enantiomer of Methyl 2-(7-(5-(3-Cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-methyl-2,3-dihydro-l//-pyrrolo[l,2- a]indol-l-yl)acetate.
To a mixture of the above-mentioned enantiomer of methyl 2-(7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)- 1 ,2,4-oxadiazol-3 -yl)-9-iodo-2,3-dihydro- lH-pyrrolo[ 1 ,2-a]indol- 1 - yl)acetate (464 mg, 0.763 mmol) and bis(tri-/-butylphosphine)palladium(0) (39.0 mg, 0.076 mmol) in TΗF was added methylzinc(II) chloride in TΗF solution (2 M) (1.144 mL, 2.288 mmol) at room temperature under nitrogen protection. The reaction mixture was heated at 70 0C for 2 h, cooled down, quenched with saturated NaHCO3 solution, and filtered through Celite®. The filtrate was extracted with ethyl acetate. The combined organics were dried then concentrated. The residue was purified by column chromatography to give the title compound as an off-white solid (255 mg). LCMS m/z = 497.2 [M+H]+; 1H NMR (400 MHz, CDCl3) 2.34 (s, 3H), 2.34-2.42 (m, IH), 2.56 (dd, J= 15.9 and 9.8 Hz, IH), 2.88-2.97 (m, IH), 2.98 (dd, J = 15.9 and 4.8 Hz, IH), 3.75 (s, 3H), 3.75-3.85 (m, IH), 4.02-4.08 (m, IH), 4.12-4.18 (m, IH), 7.29 (d, J= 8.5 Hz, IH), 7.72 (s, IH), 7.94 (dd, J= 8.5 and 1.5 Hz, IH), 8.31 (s, IH), 8.33 (d, J = 1.5 Hz, IH), 8.48 (t, J= 1.3 Hz, IH).
Step B: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5-
(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-methyl-2,3-dihydro-lH-pyrrolo[l,2- a]indol-l-yl)acetic Acid.
The above-mentioned enantiomer of methyl 2-(7-(5-(3-cyano-5-
(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-methyl-2,3-dihydro-lH-pyrrolo[l,2-a]indol- l-yl)acetate (200 mg, 0.403 mmol) in dioxane (2 mL) was added 1 M LiOH aqueous solution (1.61 mmol, 1.61 mL) at room temperature. The reaction mixture was stirred at the temperature for 3 h, diluted with water, acidified with 0.5 M citric acid aqueous solution to pΗ 4. The solid was collected and dried to give the title compound as a light yellow solid (182 mg). LCMS m/z = 483.3 [M+Η]+; 1H NMR (400 MHz, DMSO-^6) 2.29 (s, 3H), 2.29-2.35 (m, IH), 2.56 (dd, J = 16.0, 9.0 Hz, IH), 2.78-2.86 (m, 2H), 3.65-3.72 (m, IH), 4.03-4.08 (m, IH), 4.14-4.20 (m, IH), 7.45 (d, J= 8.5 Hz, IH), 7.80 (dd, 7= 8.5, 1.3 Hz, IH), 8.21 (d, J = 1.3 Hz, IH), 8.40 (s, IH), 8.44 (s, IH), 8.68 (t, J = 1.3 Hz, IH), 12.30 (bs, IH).
Example 1.9: Preparation of an Enantiomer of 2-(9-Cyano-7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2r3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetic Acid (Compound 9).
Step A: Preparation of an Enantiomer of Methyl 2-(9-cyano-7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l- yl)acetate.
To a mixture of the above-mentioned enantiomer of methyl 2-(7-(5-(3-cyano-5-
(trifluoromethoxy)phenyl)-l ,2,4-oxadiazol-3-yl)-9-iodo-2,3-dihydro-lH-pyrrolo[l ,2-a]indol-l - yl)acetate and dicyanozinc (17.5 mg, 0.148 mmol) in DMA (0.5 mL) under a nitrogen atmosphere was added tetrakis(triphenylphosphine)palladium(0) (8.5 mg, 0.1 eq). The reaction mixture was heated in a heavy-walled sealed tube under microwave irradiation at 140 0C for 1.5 h. The reaction was diluted with EtOAc, washed with H2O and 1 N HCl, dried, and
concentrated. The residue was purified by preparative TLC to give the title compound (8.5 mg). LCMS m/z = 508.2 [M+H]+.
Step B: Preparation of an Enantiomer of 2-(9-Cyano-7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazoI-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetic Acid.
To a solution of the above-mentioned enantiomer of methyl 2-(9-cyano-7-(5-(3-cyano- 5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate (8.2 mg, 0.016 mmol) in acetonitrile/Η2O (0.2 mL/4 μL, 2%) was added LiBr (21 mg, 0.24 mmol), and followed by Et3N (6.5 μL, 0.048 mmol). The reaction was stirred at 85 0C for 3 h. The reaction was diluted with DCM (5 mL), washed with H2O (twice) and 1 N HCl, dried with anhydrous MgSO4, and concentrated. The solid residue was washed with IPA to the title compound (2.1 mg). LCMS m/z = 494.3 [M+H]+; 1H NMR (400 MHz, CDCl3) δ 2.44-2.54 (m, IH), 2.69 (dd, J = 16.5 and 9.4 Hz, IH), 3.01-3.10 (m, IH), 3.19 (dd, /= 16.3 and 4.2 Hz, IH), 3.94-4.02 (m, IH), 4.11-4.18 (m, IH), 4.26-4.33 (m, IH), 7.40 (d, J= 8.6, IH), 7.74 (s, IH), 8.05 (dd, /= 8.5 and 1.4 Hz, IH), 8.33 (s, IH), 8.48 (s, IH), 8.53 (s, IH).
Example 1.10: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-(trifluoromethyl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 8).
Step A: Preparation of an Enantiomer of Methyl 2-(7-(5-(3-Cyano-5-
(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-(trifluoromethyl)-2,3-dihydro-l/-r- pyrrolo[l,2-a]indol-l-yl)acetate. To a mixture of potassium fluoride (196 mg, 3.37 mmol) and copper(I) iodide (626 mg, 3.29 mmol) in 5 mL DMA, trimethyl(trifluoromethyl)silane (514 μL, 3.29 mmol) was added. After stirring at 50 0C for 40 min, the above-mentioned enantiomer of methyl 2-(7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)- 1 ,2,4-oxadiazol-3 -yl)-9-iodo-2,3 -dihydro- lH-pyrrolo[ 1 ,2-a]indol- 1 - yl)acetate (200 mg, 0.329 mmol) was added and the mixture was continue to be stirred at 80 °C. After 30 min, the mixture was quenched with ice and purified by preparative ΗPLC. Fractions containing the title compound were partially concentrated and the residue was extracted with 1 M NaHCO3 solution and CH2Cl2. The organics were dried over MgSO4, filtered, and concentrated to give the title compound (28.0 mg) as a white solid. LCMS m/z = 551.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ ppm 2.47-2.53 (m, IH), 2.62-2.69 (m, IH), 2.96-3.06 (m, 2H), 3.73 (s, 3H), 3.90-
3.99 (m, IH), 4.15-4.28 (m, 2H), 7.40 (d, J= 8.6 Hz, IH), 7.35 (d, J = 0.9 Hz, IH), 8.05 (dd, J= 8.6, 1.6 Hz, IH), 8.33 (d, J= 0.9 Hz, IH), 8.48-8.52 (m, 2H).
Step B: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-(trifIuoroniethyI)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetic Acid.
To a solution of the above-mentioned enantiomer of methyl 2-(7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-(trifluoromethyl)-2,3-dihydro-lH-pyrrolo[l,2- a]indol-l-yl)acetate (3.7 mg, 6.72 μmol) in CH3CN (0.5 mL, with 2% H2O) were added lithium bromide (50 mg, 0.576 mmol) and triethylamine (10 μL, 0.072 mmol). After stirring at 80 0C (oil bath) for 1 h, the mixture was purified by preparative HPLC. Appropriate fractions were partly concentrated and the residue was extracted with 1 M HCl and CH2Cl2. The organics were dried over. MgSO4, filtered, and concentrated to give the title compound (0.7 mg) as a white solid. LCMS m/z = 537.2 [M+H]+. Example 1.11: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5-
(trifluoromethoxy)phenyI)-l,2,4-oxadiazol-3-yl)-9-(methylsulfonyl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 10).
Step A: Preparation of an Enantiomer of Methyl 2-(7-(5-(3-Cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yI)-9-(methylsulfonyl)-2,3-dihydro-l//- pyrrolo[l,2-a]indol-l-yl)acetate.
A mixture of the above-mentioned enantiomer of methyl 2-(7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-iodo-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate (105 mg, 0.173 mmol), sodium methanesulfmate (176 mg, 1.724 mmol), copper (I) trifluoromethanesulfonate benzene complex (10 mg, 0.020 mmol), and N'.Λ^-dimethylethane-l^- diamine (5 μL, 0.046 mmol) in DMSO (3 mL) was stirred at 80 0C (oil bath) overnight. The mixture was purified by preparative ΗPLC. Appropriate fractions were partly concentrated and the residue was extracted with 1 M NaHCO3 and CH2Cl2. The organics were dried over MgSO4, filtered, and concentrated to give the title compound (7.1 mg) as a white solid. LCMS m/z = 561.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ ppm 2.52-2.59 (m, IH), 2.88-2.94 (m, IH), 2.98-3.05 (m, IH), 3.21 (s, 3H), 3.21-3.29 (m, IH), 3.68 (s, 3H), 4.07-4.13 (m, IH), 4.19-4.33 (m, 2H), 7.43 (d, J = 8.6 Hz, IH), 7.74 (s, IH), 8.11 (dd, J = 8.6, 1.5 Hz, IH), 8.34 (s, IH), 8.50 (m, IH), 8.73 (m, IH).
Step B: Preparation of an Enantiomer of 2-(7-(5-(3-Cyano-5-
(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-(methylsulfonyl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetic Acid.
A mixture of the above-mentioned enantiomer of methyl 2-(7-(5-(3-cyano-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-(methylsulfonyl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetate (5.4 mg, 9.63 μmol), lithium bromide (25 mg, 0.288 mmol), and triethylamine (5 μL, 0.036 mmol) in 1 mL CH3CN with 2% H2O was stirred at 80 0C for 30 min. The mixture was purified by preparative HPLC. Appropriate fractions were partly concentrated and the residue was extracted with 1 M HCl and CH2Cl2. The organics were dried over MgSO4, filtered, and concentrated to give the title compound (3.8 mg) as a white solid. LCMS m/z = 547.3 [M+H]+; 1H NMR (400 MHz, CDCl3) δ ppm 2.56-2.63 (m, IH), 2.93-3.07 (m, 2H), 3.22 (s, 3H), 3.31-3.36 (m, IH), 4.08-4.13 (m, IH), 4.20-4.33 (m, 2H), 7.44 (d, J= 8.6 Hz, IH), 7.74 (s, IH), 8.11 (dd, J= 8.5, 1.5 Hz, IH), 8.34 (s, IH), 8.50 (d, J = 1.2 Hz, IH), 8.72 (s, IH). Example 1.12: Preparation of 2-(9-Chloro-7-(5-(3-cyano-5-methoxyphenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 14).
Step A: Preparation of tert-Butyl 2-(7-(5-(3-Cyano-5-methoxyphenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetate.
3-Cyano-5-methoxybenzoic acid (0.027 g, 0.152 mmol) was dissolved in DMA (1.0 mL). HATU (0.075 g, 0.197 mmol) and triethylamine (0.042 mL, 0.304 mmol) were added. After stirring for 30 minutes, tert-butyl 2-(7-(N'-hydroxycarbamimidoyl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetate (0.05 g, 0.152 mmol) was added and the reaction was stirred at room temperature for 1 h. The reaction was warmed to 90 0C and stirred for 48 h. The reaction mixture was concentrated and the residue was purified by column chromatography to give the title compound (47.7 mg). LCMS m/z = 471.4 [M+Η]+.
Step B: Preparation of 2-(9-Chloro-7-(5-(3-cyano-5-methoxyphenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic Acid.
/er/-Butyl 2-(7-(5-(3-cyano-5-methoxyphenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetate (47.7 mg, 0.101 mmol) was taken up in DCM (1.0 mL) and cooled to 0 0C. ΝCS (13.5 mg, 0.101 mmol) was added and the reaction was stirred for 30 minutes and then diluted with saturated aqueous sodium thiosulfate. The DCM layer was separated, dried over sodium sulfate, and concentrated. The residue was purified by column chromatography and the resultant material was taken up into a mixture of D/L-cysteine (120 mg) and TFA (2.0 mL). After 10 minutes at room temperature, the reaction was diluted with water and the resulting solid was isolated by filtration to give the title compound (11.2 mg). LCMS m/z = 449.2 [M+H]+; 1H NMR (400 MHz, DMSO-^) δ 2.31-2.42 (m, IH), 2.63 (dd, J= 16.4 , 9.6 Hz, IH), 2.82-2.92 (m, IH), 2.95-3.04 (m, IH), 3.69-3.81 (m, IH), 3.96 (s, 3H), 4.08- 4.17 (m, IH), 4.23-4.33 (m, IH), 7.57 (d, J = 8.6 Hz, IH), 7.81 (s, IH), 7.89 (d, J= 8.6 Hz, IH), 7.95 (s, IH), 8.17 (s, 2H), 12.3 (bs, IH).
Example 1.13: Preparation of 2-(9-Chloro-7-(5-(2-chloro-6-methyIpyridin-4-yl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrroIo[l,2-a]indol-l-yl)acetic Acid (Compound 13).
Step A: Preparation of te/f-Butyl 2-(7-(5-(2-Chloro-6-methylpyridin-4-yl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate.
To a solution of tert-butyl 2-(7-(N'-hydroxycarbamimidoyl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetate (0.075 g, 0.228 mmol) and triethylamine (0.111 mL, 0.797 mmol) in TΗF was added 2-chloro-6-methylisonicotinoyl chloride (0.032 mL, 0.228 mmol). The resulting suspension was stirred overnight. The mixture was concentrated and dissolved in DMF, then heated at 90 0C for 4 h. The solvent was removed and the residue was taken up in EtOAc. The organics were washed with 1 M HCl followed by brine (twice). The organic layer was dried over MgSO4, filtered, and concentrated under reduced pressure. The residue was purified by silica gel to yield the title compound as a white solid (0.078 g). LCMS m/z = 465.3 [M+Η]+; 1H ΝMR (400 MHz, CDCl3) δ ppm 1.51 (s, 9H), 2.29-2.39 (m, IH), 2.57 (dd, J = 15.79, 8.21 Hz, IH), 2.69 (s, 3H), 2.78 (dd, J= 15.92, 6.82 Hz, IH), 2.93 (m, IH), 3.77 (m, IH), 4.09 (dt, J= 10.10, 7.58 Hz, IH), 4.21 (m, IH), 6.29 (s, IH), 7.34 (d, J= 8.34 Hz, IH), 7.87 (s, IH), 7.91-7.97 (m, 2H), 8.40 (d, J= 1.52 Hz, IH).
Step B: Preparation of tert-Butyl 2-(9-Chloro-7-(5-(2-chloro-6-methylpyridin-4-yl)- l,2,4-oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetate.
To a solution of tert-butyl 2-(7-(5-(2-chloro-6-methylpyridin-4-yl)-l,2,4-oxadiazol-3- yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate (0.075 g, 0.161 mmol) in anhydrous DCM (3.23 mL) cooled to 0 0C was added l-chloropyrrolidine-2,5-dione (0.023 g, 0.169 mmol). After stirring the reaction mixture at 0 0C for 1 h, the reaction mixture was warmed to room temperature for about 15 min, after which the reaction mixture was quenched with 2 M sodium thiosulfate (4.03 mL, 8.07 mmol). The mixture was extracted with MTBE (50 mL). The organic layer was washed with 2 M sodium thiosulfate (2 x 10 mL), water (10 mL), brine (10 mL), dried over MgSO4, and concentrated under reduced pressure. The residue was purified by silica gel chromatography to give the title compound as a yellow solid (48 mg). LCMS m/z = 499.1
[M+Η]+; 1H ΝMR (400 MHz, Acetone-rf6) δ ppm 1.44 (s, 9H), 2.50 (m, IH), 2.65 (d, J= 6.57 Hz, IH), 2.68 (s, 3H), 3.01 (dd, J= 12.88, 4.67 Hz, IH), 3.07 (dd, J= 16.04, 4.04 Hz, IH), 3.84 (m, IH), 4.21 (ddd, J= 10.17, 8.02, 6.95 Hz, IH), 4.35 (ddd, J= 10.20, 8.56, 4.74 Hz, IH), 7.53 (d, J= 8.59 Hz, IH), 7.94 - 7.99 (m, 2H), 8.02 (s, IH), 8.31 (d, J= 1.39 Hz, IH).
Step C: Preparation of 2-(9-Chloro-7-(5-(2-chloro-6-methylpyridin-4-yl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetic Acid.
tert-Butyl 2-(9-chloro-7-(5-(2-chloro-6-methylpyridin-4-yl)-l ,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate (0.044 g, 0.088 mmol) was dissolved in a pre- mixed solution of DCM (2 mL, 31.1 mmol), TFA (1.0 mL, 12.98 mmol), and thioanisole (0.104 mL, 0.881 mmol) and stirred at room temperature for 1 h. Toluene (20 mL) was added and the solvent was evaporated under reduced pressure at 25 0C to give a residue which was coevaporated with toluene (20 mL) again at 25 0C to give a solid residue. The solid residue was suspended in DCM (10 mL) and hexane (40 mL) was added to give a grey suspension. The solvent was evaporated under reduced pressure at 25 0C to a volume of 10 mL. The solid was filtered, washed with hexanes (3 times), and dried under reduced pressure to give the title compound as a grey solid (0.038 g). LCMS m/z = 443.3 [M+Η]+; 1H NMR (400 MHz, DMSO- d6) δ ppm 2.36 (m, IH), 2.63 (s, 3H), 2.65 (d, J= 9.60 Hz, IH), 2.82-2.92 (m, IH), 2.99 (dd, J = 16.48, 4.23 Hz, IH), 3.70-3.82 (m, 2H), 4.28 (m, IH), 7.59 (d, J= 8.46 Hz, IH), 7.90 (dd, J = 8.59, 1.64 Hz, IH), 8.00 (s, IH), 8.05 (s, IH), 8.19 (d, J= 1.26 Hz, IH), 12.40 (bs, IH).
Example 1.14: Preparation of 2-(9-Chloro-7-(5-(4-cyclopentyl-3-(trifluoromethyl)phenyl)- l,2,4-oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 12).
Step A: Preparation of tert-Butyl 2-(7-(5-(4-CyclopentyI-3-
(trifluoromethyl)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate.
4-Cyclopentyl-3-(trifluoromethyl)benzoic acid (58.8 mg, 0.228 mmol), oxalyl chloride (0.040 ml, 0.455 mmol) and DMF (1.175 μL, 0.015 mmol) in DCM (1.00 mL) were stirred at room temperature for 1 h in a 20 mL sealed scintillation vial. After 1 h, the solvent was removed under reduced pressure to give 4-cyclopentyl-3-(trifluoromethyl)benzoyl chloride, which was immediately added to a solution of tert-buty\ 2-(7-(N-hydroxycarbamimidoyl)-2,3-dihydro-lH- pyrrolo[l,2-a]indol-l-yl)acetate (50 mg, 0.152 mmol) and TEA (0.023 ml, 0.167 mmol) in TΗF (1.00 mL). The reaction was stirred at room temperature for 1 h. After removal of the solvent under reduced pressure, the residue was dissolved in DMF (1.00 mL). The reaction was then heated to 75 0C for 16 h. The reaction was then taken up in EtOAc (5 mL) and washed with water (2 x 5 mL), dried over MgSO4, filtered by vacuum filtration through a glass fiber paper and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as an off-white solid (35 mg). LCMS m/z = 552.5 [M+Η]+. Step B: Preparation of terf-Butyl 2-(9-ChIoro-7-(5-(4-cyclopentyl-3- (trifluoromethyI)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-l//-pyrroIo[l,2-a]indol-l- yl)acetate.
To a solution of tert-butyl 2-(7-(5-(4-cyclopentyl-3-(trifluoromethyl)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate (23.4 mg, 0.042 mmol) in DCM (1.00 mL) at 0 0C in a 20 mL sealed scintillation vial, NCS (5.66 mg, 0.042 mmol) was added and the reaction was allowed to continue at 0 0C for 1 h. The mixture was taken up in DCM (5 mL) and washed with water (3 x 5 mL) then saturated sodium thiosulfate (aq) (I x 5mL), dried over MgSO4, and filtered by vacuum filtration through a glass fiber paper. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound as a yellow oil (18.3 mg). LCMS m/z = 586.2
[M+Η]+.
Step C: Preparation 2-(9-Chloro-7-(5-(4-cyclopentyI-3-(trifluoromethyl)phenyl)- l,2,4-oxadiazol-3-yl)-2,3-dihydro-l//-pyrroIo[l,2-a]indol-l-yl)acetic Acid.
A solution of 2-amino-3-mercaptopropanoic acid (11.35 mg, 0.094 mmol) in TFA (1 mL) was cooled down to 0 0C. The cold solution was added to tert-huty\ 2-(9-chloro-7-(5-(4- cyclopentyl-3-(trifluoromethyl)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2- a]indol-l-yl)acetate (18.3 mg, 0.031 mmol) in a 20 mL sealed scintillation vial and stirred at 0 0C for 1 h. The mixture was poured into ice water and stirred for 30 minutes. The resulting precipitate was collected by vacuum filtration, washed with water (2 x 5 mL), n-hexane (3 x 5 mL), and dried (vacuum oven) to give the title compound as a tan solid (14.0mg). LCMS m/z = 530.3 [M+Η]+; 1H NMR (400 MHz, DMSCW6) δ ppm 1.63-1.79 (m, 4H), 1.84-1.96 (m, 2H), 2.03-2.14 (m, 2H), 2.31-2.42 (m, IH), 2.63 (dd, J = 16.55, 9.60 Hz, IH), 2.83-2.93 (m, IH), 2.99 (dd, J= 16.42, 4.29 Hz, IH), 3.29-3.42 (m, IH), 3.70-3.81 (m, IH), 4.09-4.17 (m, IH), 4.24-4.32 (m, IH), 7.58 (d, J= 8.59 Hz, IH), 7.88-7.96 (m, 2H), 8.18 (d, J= 1.01 Hz, IH), 8.36 (d, J= 1.26 Hz, IH), 8.42 (d, J= 8.34 Hz, IH), 12.40 (s, IH).
Example 1.15: Preparation of 2-(7-(5-(3,5-Bis(trifluoromethyl)phenyl)-l,2,4-oxadiazol-3- yl^-chloro^^-dihydro-lH-pyrroloIl^-aJindol-l-yOacetic Acid (Compound 11).
Step A: Preparation of terf-Butyl 2-(7-(5-(3,5-Bis(trifluoromethyl)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate.
To a stirred solution of 3,5-bis(trifluoromethyl)benzoic acid (39.2 mg, 0.152 mmol) and HATU (69.3 mg, 0.182 mmol) in DMF (1 mL) was added DIEA (52.9 μL, 0.304 mmol). After 15 min, tert-butyl 2-(7-(N'-hydroxycarbamimidoyl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate (50 mg, 0.152 mmol) was added. The reaction mixture was stirred at room temperature for 30 min, and then heated at 65 0C for 1 h. The solvent was evaporated, and the residue was purified by column chromatography to give the title compound (53 mg). LCMS m/z = 552.6 [M+H]+; 1H NMR (400 MHz, CDCl3) δ ppm 1.51 (s, 9H), 2.28-2.38 (m, IH), 2.55 (dd, J = 15.8 and 8.2 Hz, IH), 2.78 (dd, 7= 15.9 and 6.6 Hz, IH), 2.88-2.96 (m, IH), 3.72-3.80 (m, IH), 4.02-4.10 (m, IH), 4.15-4.23 (m, IH), 6.26 (s, IH), 7.32 {ά, J = 8.5 Hz, IH), 7.94 (dd, 7 = 8.5 and 1.5 Hz, IH), 8.09 (s, IH), 8.40 (s, IH), 8.67 (s, 2H).
Step B: Preparation of tert-Butyl 2-(7-(5-(3,5-Bis(trifluoromethyl)phenyl)-l,2,4- oxadiazoIO-ylJ-P-chloro^jS-dihydro-lH-pyrrolofl^-alindol-l-ylJacetate.
To a stirred solution of tert-butyl 2-(7-(5-(3,5-bis(trifluoromethyl)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate (53 mg, 0.096 mmol) in DCM (2 mL) was added NCS (13.5 mg, 0.1 mmol) at 0 0C. The reaction mixture was stirred at this temperature for 30 min, diluted with ethyl acetate, washed with aqueous Na2S2O3 solution and water, and dried over anhydrous Na2SO4. The solvent was evaporated to give the title compound (40 mg). LCMS m/z = 586.3 [M+Η]+.
Step C: Preparation of 2-(7-(5-(3,5-Bis(trifluoromethyl)phenyl)-l,2,4-oxadiazol-3- yl)-9-ch!oro-2T3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetic Acid.
D/L-Cysteine (41.4 mg, 0.341 mmol) was dissolved in TFA (1 mL) then cooled to 0 0C.
The solution was added to a solution of ter/-butyl 2-(7-(5-(3,5-bis(trifluoromethyl)phenyl)- l,2,4-oxadiazol-3-yl)-9-chloro-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate (40 mg, 0.068 mmol) in DCM (1 mL) at 0 0C. The reaction mixture was warmed to room temperature and stirred for 30 min. Water was added, then ethyl acetate was added. The organic layer was separated, washed with water, brine and dried over anhydrous Na2SO4. The solvent was evaporated, and the residue was passed through a silica gel column with 10% MeOΗ/DCM to give the title compound as light yellow solid (23 mg). LCMS m/z = 530.0 [M+Η]+; 1H NMR (400 MHz, DMSO-^6) δ ppm 2.30-2.41 (m, IH), 2.63 (dd, J = 16.4 and 9.6 Hz, IH), 2.84-2.94 (m, IH), 3.00 (dd, J = 16.4 and 4.2 Hz, IH), 3.72-3.80 (m, IH), 4.10-4.18 (m, IH), 4.25-4.32 (m, IH), 7.60 (d, J= 8.5 Hz, IH), 7.93 (dd, 7= 8.5 and 1.5 Hz, IH), 8.20 (s, IH), 8.54 (s, IH), 8.76 (s, 2H), 12.3 (br, IH).
Example 1.16: Preparation of 2-(6-(5-(3-Cyano-5-(trifluoromethoxy)phenyI)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-benzo[d]pyrrolo[l,2-a]imidazol-3-yl)acetic Acid
(Compound 15).
Step A: Preparation of Ethyl 2-(l-(4-Cyano-2-nitrophenyl)-2-oxopyrrolidin-3- yl)acetate.
Ethyl 2-(2-oxopyrrolidin-3-yl)acetate (0.792 g, 4.63 mmol) was taken up in DMA (15 mL) and the resulting mixture was cooled to 0 0C. Sodium hydride (0.185 g, 4.63 mmol) was added and the reaction was warmed to room temperature and stirred for 1 h. 4-Fluoro-3- nitrobenzonitrile (0.768 g, 4.63 mmol) was added and the reaction was stirred at room temperature for 2 days. The reaction was diluted with water and extracted with EtOAc. The organic extract was dried over sodium sulfate, filtered, and concentrated. The residue was purified by silica gel column chromatography to give the title compound (620 mg). LCMS m/z = 318.2 [H+M]+.
Step B: Preparation of Ethyl 2-(6-Cyano-2,3-dihydro-l//-benzo[d]pyrrolo[l,2- a]imidazol-3-yl)acetate.
Ethyl 2-(l-(4-cyano-2-nitrophenyl)-2-oxopyrrolidin-3-yl)acetate (0.62 g, 1.954 mmol) was taken up in THF (10 mL) and palladium on carbon (0.042 g, 0.391 mmol) was added. The reaction vessel was purged with nitrogen followed by hydrogen. A hydrogen balloon was attached and the reaction was stirred at room temperature for 5 h. The reaction mixture was filtered through Celite®, and the filtrate was concentrated under reduced pressure. The residue was dissolved in acetic acid (5 mL, 87 mmol) and the solution was warmed to 75 0C. After stirring for 16 h, the reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound (426 mg). LCMS m/z = 270.2 [M+H]+; 1H NMR (400 MHz, CDCl3) δ ppm 1.25 (t, J= 7.2 Hz, 3H), 2.45-2.55 (m, IH), 2.73 (dd, J= 16.8, 9.0 Hz, IH), 3.04-3.19 (m, 2H), 3.70-3.80 (m, IH), 4.07-4.28 (m, 4H), 7.37 (d, J= 8.3 Hz, IH), 7.49 (dd, J= 8.3, 1.4 Hz, IH), 8.02 (s, IH).
Step C: Preparation of Ethyl 2-(6-(iV-Hydroxycarbamimidoyl)-2,3-dihydro-lH- benzo [d] pyrrolo [ 1 ,2-a] imidazol-3-yl)acetate.
Ethyl 2-(6-cyano-2,3-dihydro-lH-benzo[d]pyrrolo[l,2-a]imidazol-3-yl)acetate (0.213 g, 0.791 mmol) was dissolved in ethanol (10 mL) and 50% hydroxylamine in water (0.261 g, 7.91 mmol) was added. The reaction was warmed to 60 0C and stirred for 16 h. The mixture was concentrated under reduced pressure to give the title compound (213 mg) as a tan solid. LCMS Wz = 303.4 [M+Η]+.
Step D: Preparation of Ethyl 2-(6-(5-(3-Cyano-5-(trifluoromethoxy)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-benzo[d]pyrrolo[l,2-a]imidazol-3-yl)acetate.
3-Cyano-5-(trifluoromethoxy)benzoic acid (0.163 g, 0.705 mmol) was dissolved in DMA (3 mL). HATU (0.348 g, 0.916 mmol) and triethylamine (0.196 mL, 1.409 mmol) were added. The reaction was stirred for 30 min at room temperature. Ethyl 2-(6-(N'- hydroxycarbamimidoyl^^-dihydro-lH-benzofdJpyrrolotl^-a^midazol-S-yOacetate (0.213 g, 0.705 mmol) was added. The reaction was stirred for 1 h, warmed to 90 0C, and stirred for an additional hour. The mixture was extracted with EtOAc and water. The EtOAc layer was separated, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound (82 mg). LCMS m/z = 498.5 [M+Η]+.
Step E: Preparation of 2-(6-(5-(3-Cyano-5-(trifluoromethoxy)phenyl)-l,2,4- oxadiazol-S-yO-Z^-dihydro-lH-benzoIdlpyrrololl^-alimidazol-S-y^acetic Acid. Ethyl 2-(6-(5 -(3 -cyano-5 -(trifluoromethoxy)phenyl)- 1 ,2,4-oxadiazol-3 -yl)-2,3 -dihydro- lH-benzo[d]pyrrolo[l,2-a]imidazol-3-yl)acetate (82 mg, 0.165 mmol) was dissolved in 2% water in acetonitrile solution (1.0 mL). LiBr (12.0 mg, 1.65 mmol) and triethylamine (69 μL, 0.495 mmol) were added. The reaction was warmed to 70 0C and stirred for 16 h. The reaction mixture was concentrated under reduced pressure and purified by preparatory TLC to give the title compound. LCMS m/z = 470.1 [M+Η]+.
Example 1.17: Preparation of 2-(9-Chloro-7-(5-(3-chloro-5-(trifluoromethoxy) phenyl)- l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 16).
Step A: Preparation of tø/t-Butyl 2-(7-(5-(3-Chloro-5-(trifluoromethoxy)phenyl)-
1 ,2,4-oxadiazol-3-yl)-2,3-dihydro-l/-r-pyrrolo [1 ,2-a] indol-l-yl)acetate.
To a stirred solution of 3-chloro-5-(trifluoromethoxy)benzoic acid (51 mg, 0.213 mmol) and HATU (105 mg, 0.276 mmol) in DMF (1 mL) was added DIEA (74.0 μL, 0.425 mmol). After 15 min, tert-butyl 2-(7-(N-hydroxycarbamimidoyl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate (70 mg, 0.213 mmol) was added. The reaction mixture was stirred at room temperature for 30 min, then heated at 70 0C for 1 h, and cooled down. The solvent was evaporated, and the residue was purified by silica gel column chromatography to give the title compound as a white solid (80 mg). LCMS m/z = 534.4 [M+Η]+; 1H ΝMR (400 MHz, CDCl3) 1.51 (s, 9H), 2.28-2.38 (m, IH), 2.55 (dd, J = 15.8 and 8.2 Hz, IH), 2.78 (dd, J= 15.8 and 6.6 Hz, IH), 2.88-2.96 (m, IH), 3.72-3.80 (m, IH), 4.02-4.11 (m, IH), 4.15-4.23 (m, IH), 6.28 (s, IH), 7.32 (d, J= 8.5 Hz, IH), 7.45 (s, IH), 7.93 (dd, J= 8.5 and 1.5 Hz, IH), 7.99 (s, IH), 8.17 (t, 7= 1.3 Hz, IH), 8.39 (d, 7= 1.3 Hz, IH).
Step B: Preparation of tert-Butyl 2-(9-Chloro-7-(5-(3-chloro-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2^-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate.
To a stirred solution of ter/-butyl 2-(7-(5-(3-chloro-5-(trifluoromethoxy)phenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate (80 mg, 0.150 mmol) in DCM (1 mL) was added ΝCS (21 mg, 0.157 mmol) at 0 0C. The reaction mixture was stirred at this temperature for 30 min, diluted with ethyl acetate, washed with aqueous Na2S2O3 solution and water, dried over anhydrous Na2SO4. The solvent was evaporated and the residue was purified by silica gel column chromatography to give the title compound as a yellowish white solid (62 mg). LCMS m/z = 568.3 [M+Η]+.
Step C: Preparation of 2-(9-ChIoro-7-(5-(3-chloro-5-(trifluoromethoxy)phenyl)- l,2,4-oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indoI-l-yl)acetic Acid.
D/L-Cysteine (66 mg, 0.545 mmol) was dissolved in TFA (1.5 mL) then cooled to 0 0C.
The solution was added to a solution of terf-butyl 2-(9-chloro-7-(5-(3-chloro-5- (trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l- yl)acetate (62 mg, 0.109 mmol) in DCM (1 mL) at O 0C. The reaction mixture was warmed to room temperature, stirred for 30 min, and water was added, followed by ethyl acetate. The organic layer was separated, washed with water and brine, and dried over anhydrous Na2SO4. The solvent was evaporated, and the residue was passed through a silica gel column with 10% MeOH/DCM to give the title compound as a solid (48 mg). LCMS m/z = 512.2 [M+H]+; 1H NMR (400 MHz, CDCl3) 2.38-2.48 (m, IH), 2.66 (dd, J= 16.8, 10.2 Hz, IH), 2.98-3.08 (m, IH), 3.35 (dd, J = 16.8 and 3.9 Hz, IH), 3.82-3.92 (m, IH), 4.06-4.15 (m, IH), 4.21-4.28 (m, IH), 7.33 (d, J= 8.5 Hz, IH), 7.48 (s, IH), 7.98-8.03 (m, 2H), 8.20 (t, J = 1.6 Hz, IH), 8.41 (d, J= 1.3 Hz, IH).
Example 1.18: Preparation of 2-(9-Chloro-7-(5-(3-fluoro-4-methoxyphenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 17).
Step A: Preparation of te/t-Butyl 2-(7-(5-(3-Fluoro-4-methoxyphenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-pyrroIo[l,2-a]indol-l-yl)acetate.
3-Fluoro-4-methoxybenzoyl chloride (42.9 mg, 0.228 mmol) was added to a solution of 2-(7-(N-hydroxycarbamimidoyl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate (50.0 mg, 0.152 mmol) and TEA (0.032 mL, 0.228 mmol) in TΗF (1.0 mL) in a 20 mL sealed scintillation vial. The reaction was stirred at room temperature for 1 h. The solvent was removed under reduced pressure. The residue was dissolved in DMA (1.0 mL) and heated to 70 0C for 16 h. The solvent was again removed under reduced pressure and the residue was purified by silica gel column chromatography to give the title compound (35.6 mg) as a white solid. LCMS m/z = 464.3 [M+Η]+.
Step B: Preparation of te/t-Butyl 2-(9-Chloro-7-(5-(3-fluoro-4-methoxyphenyl)- l,2,4-oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yI)acetate.
To a solution of tert-butyl 2-(7-(5-(3-fluoro-4-methoxyphenyl)-l,2,4-oxadiazol-3-yl)-
2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetate (35.6 mg, 0.077 mmol) in DCM (1.00 mL) at 0 0C in a 20 mL sealed scintillation vial. ΝCS (10.26 mg, 0.077 mmol) was added and the reaction was stirred at 0 0C for 1 h. The mixture was taken up in DCM (5 mL) and washed with water (3 x 5 mL), then saturated sodium thiosulfate (aq) (1 x 5 mL), dried over MgSO4, and filtered by vacuum filtration through a glass fiber paper. The solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography to give the title compound (22.9 mg) as a light yellow solid. LCMS m/z = 498.4 [M+Η]+.
Step C: Preparation of 2-(9-Chloro-7-(5-(3-fluoro-4-methoxyphenyl)-l,2,4- oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetic Acid.
A solution of 2-amino-3-mercaptopropanoic acid (16.72 mg, 0.138 mmol) in TFA (1 mL) was made and cooled down to 0 0C. The cold solution was added to /er/-butyl 2-(9-chloro- 7-(5 -(3-fluoro-4-methoxyphenyl)- 1 ,2,4-oxadiazol-3 -yl)-2,3 -dihydro- lH-pyrrolo[ 1 ,2-a]indol- 1 - yl)acetate (22.9 mg, 0.046 mmol) in a 20 mL sealed scintillation vial at 0 °C and stirred at 0 0C for 5 mm. The reaction mixture was warmed up to room temperature and stirred at 23 0C for 15 mm. The mixture was poured into ice water and stirred for 30 mm. The resulting precipitate was collected by vacuum filtration, washed with water (2 x 5 mL), /i-hexanes (3 x 5 mL) and dried (vacuum oven) to give the title compound as a tan solid (18.6 mg). LCMS m/z = 442.6 [M+H]+; 1H NMR (400 MHz, DMSO-J6) δ ppm 2.31-2.42 (m, IH), 2.63 (dd, J = 16.42, 9.60 Hz, IH), 2.82-2.93 (m, IH), 2.99 (dd, J = 16.42, 4.17 Hz, IH), 3.72-3.81 (m, IH), 3.98 (s, 3H), 4.08-4.17 (m, IH), 4.23-4.32 (m, IH), 7.44 (t, J = 8.59 Hz, IH), 7.57 (d, J= 8.46 Hz, IH), 7.88 (dd, J = 8.59, 1.52 Hz, IH), 7.98-8.08 (m, 2H), 8.17 (d, J= 1.26 Hz, IH), 12.40 (s, IH).
Example 1.19: Preparation of 2-(9-Chloro-7-(5-(4-isopropoxy-3-methoxyphenyI)-l,2,4- oxadiazoI-3-yl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 18).
Step A: Preparation of tert-Butyl 2-(9-Chloro-7-(5-(4-isopropoxy-3- methoxyphenyl)-l,2,4-oxadiazol-3-yl)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetate. tert-Butyl 2-(7-(5-(4-isopropoxy-3 -methoxyphenyl)- 1 ,2,4-oxadiazol-3 -yl)-2,3 -dihydro- lH-pyrrolo[l,2-a]indol-l-yl)acetate (0.049 g, 0.097 mmol) was dissolved in anhydrous dichloromethane (1.95 mL) to give a solution which was cooled to 5 0C after which 1- chloropyrrohdine-2,5-dione (0.014 g, 0.107 mmol) was added. After stirring at 0 0C for 1.5 h the reaction mixture was allowed to warm to 24 0C for about 15 mm, then the reaction was quenched with 2 M sodium sulfothioate (2.4 mL, 4.9 mmol). The reaction mixture was extracted with MTBE (50 mL), the organic layer was washed with 2 M sodium sulfothioate (10 mL), water (10 mL), bπne (10 mL), dried with MgSO4, and the solvent was concentrated to give a yellow solid which was purified by silica gel flash chromatography to give the title compound as a beige solid (0.035 g). LCMS m/z = 538.1 [M+Η]+; 1H NMR (400 MHz, DMSO-J6) δ ppm 1.33 (d, J= 6.06 Hz, 6H), 1.39 (s, 9H), 2.29 - 2.43 (m, IH), 2.61 - 2.70 (m, IH), 2.81 - 2.90 (m, IH), 2.93 (dd, J= 16.04, 4.29 Hz, IH), 3.68 - 3.84 (m, IH), 3.91 (s, 3H), 4.05 - 4.19 (m, IH), 4.21 - 4.32 (m, IH), 4.66 - 4.84 (m, IH), 7.23 (d, J= 8.72 Hz, IH), 7.57 (d, J= 8.46 Hz, IH), 7.67 (d, J= 2.02 Hz, IH), 7.79 (dd, J= 8.34, 2.02 Hz, IH), 7.89 (dd, J= 8.59, 1.52 Hz, IH), 8.17 (d, J= 1.39 Hz, IH).
Step B: Preparation of 2-(9-Chloro-7-(5-(4-isopropoxy-3-methoxyphenyI)-l,2,4- oxadiazol-3-yI)-2,3-dihydro-l//-pyrrolo[l,2-a]indol-l-yl)acetic Acid (Compound 18).
L(+)-Cysteine (0.037 g, 0.307 mmol) was dissolved in TFA (0.613 mL) to give a solution which was cooled to 0 0C. This solution was added to tert-butyl 2-(9-chloro-7-(5-(4- isopropoxy-3-methoxyphenyl)- 1 ,2,4-oxadiazol-3 -yl)-2,3 -dihydro- 1 H-pyrrolo[ 1 ,2-a]indol- 1 - yl)acetate (0.033 g, 0.061 mmol) which was contained in a flask pre-cooled to 00C. The reaction was stirred at 00C for 45 mm then at 240C for 10 min. Cold water (20 mL) was added to the reaction mixture while stirring to give a suspension to which Et2O (50 mL) was added to give dissolution of the suspension. The mixture was shaken, the organic layer was washed with water (3 x 20 mL), brine (20 mL), dried with MgSO4, and the solvent was evaporated in the presence of toluene under reduced pressure at 250C to afford the title compound as a grey solid (17 mg). LCMS m/z = 482.2 [M+H]+; 1H NMR (400 MHz, DMSO-^6) δ ppm 1.33 (d, J = 6.06 Hz, 6H), 2.31 - 2.42 (m, IH), 2.63 (dd, J = 16.42, 9.73 Hz, IH), 2.81 - 2.92 (m, IH), 2.99 (dd, J= 16.48, 4.23 Hz, IH), 3.70 - 3.82 (m, IH), 3.91 (s, 3H), 4.07 - 4.18 (m, IH), 4.23 - 4.33 (m, IH), 4.69 - 4.82 (m, IH), 7.23 (d, J = 8.72 Hz, IH), 7.57 (d, J = 8.59 Hz, IH), 7.67 (d, J= 2.02 Hz, IH), 7.79 (dd, J= 8.46, 2.02 Hz, IH), 7.89 (dd, J= 8.59, 1.52 Hz, IH), 8.17 (d, J= 1.39 Hz, IH), 12.39 (bs, IH).
Example 2: Homogeneous Time-Resolved Fluorescence (HTRF®) Assay for Direct cAMP Measurement.
Compounds of the present invention were screened for agonists of the SlPl receptor (e.g., human SlPl receptor) using the HTRF® assay for direct cAMP measurement (Gabriel et al., Assay and Drug Development Technologies, 1:291-303, 2003) and recombinant CHO-Kl cells stably transfected with SlPl. CHO-Kl cells were obtained from ATCC® (Manassas, VA; Catalog # CCL-61). An agonist of the SlPl receptor was detected in the HTRF® assay for direct cAMP measurement as a compound which decreased cAMP concentration. HTRF® assay also was used to determine EC50 values for SlPl receptor agonists.
Principle of the assay: HTRF® assay kit was purchased from Cisbio-US, Inc. (Bedford,
MA; Catalog # 62AM4PEC). The HTRF® assay supported by the kit is a competitive immunoassay between endogenous cAMP produced by the CHO-Kl cells and tracer cAMP labeled with the dye d2. The tracer binding is visualized by a monoclonal anti-cAMP antibody labeled with Cryptate. The specific signal (i.e., fluorescence resonance energy transfer, FRET) is inversely proportional to the concentration of unlabeled cAMP in the standard or sample.
Standard curve: The fluorescence ratio (665 nm/620 nm) of the standards (0.17 to 712 nM cAMP) included in the assay was calculated and used to generate a cAMP standard curve according to the kit manufacturer's instructions. The fluorescence ratio of the samples (test compound or compound buffer) was calculated and used to deduce respective cAMP concentrations by reference to the cAMP standard curve.
Setup of the assay: The HTRF® assay was carried out using a two-step protocol essentially according to the kit manufacturer's instructions, in 20 μL total volume per well in 384-well plate format (ProxiPlates; PerkinElmer, Fremont, CA; catalog # 6008280). To each of the experimental wells was transferred 1500 recombinant CHO-Kl cells in 5 μL phosphate buffered saline containing calcium chloride and magnesium chloride ("PBS+"; Invitrogen, Carlsbad, CA; catalog # 14040) supplemented with IBMX (250 μM) and rolipram (20 μM) (phosphodiesterase inhibitors; Sigma-Aldrich, St. Louis, MO; catalog # 15879 and catalog # R6520, respectively), followed by test compound in 5 μL compound buffer (PBS+
supplemented with 10 μL NKH477 (water-soluble forskolin derivative; SignaGen Laboratories, Gaithersburg, MD; catalog # PKI-NKH477-010)) or 5 μL compound buffer. The plate was then incubated at room temperature for 1 h. To each well was then added 5 μL cAMP-d2 conjugate in lysis buffer and 5 μL Cryptate conjugate in lysis buffer according to the kit manufacturer's instructions. The plate was then further incubated at room temperature for 1 hour, after which the assay plate was read.
Assay readout: HTRF® readout was accomplished using a PHERAstar (BMG
LABTECH Inc., Durham, NC) or EnVision™ (PerkinElmer, Fremont CA) microplate reader.
Certain compounds of the present invention and their corresponding activity values are shown in TABLE B.
TABLE B
Figure imgf000071_0001
Certain other compounds of the invention had EC50 SlPl (HTRF®) values ranging from about 5 nM to about 0.1 nM in this assay.
Example 3: Cellular/Functional Ca2+ Assay for Agonist Activity on S1P3 Receptor.
A compound of the present invention can be shown to have no or substantially no agonist activity on the S1P3 receptor by using in assay a human neuroblastoma cell line which endogenously expresses S1P3 (predominantly), S1P2 and S1P5 receptors, but not SlPl or S1P4 receptors, based on mRNA analysis (Villullas et al, J. Neurosci. Res., 73:215-226, 2003). Of these, S1P3 and S1P2 receptors respond to agonists, such as SlP, with an intracellular calcium increase. No or substantially no increase of intracellular calcium in response to a test compound is indicative of the test compound exhibiting no or substantially no agonist activity on the S1P3 receptor. Such an assay can be performed commercially, e.g. by Caliper LifeSciences
(Hopkinton, MA).
Assay: The human neuroblastoma cells are washed and resuspended in physiological buffer. The cells are then loaded with dye that measures intracellular calcium. SlP is used as a reference agonist. After addition of SlP or a test compound, fluorescence is measured at 485 nm excitation / 525 nm emission every 2 s for at least 60 s. Calcium ionophore A23187 is then added as an internal positive control. Example 4: Effect of Compounds in Peripheral Lymphocyte Lowering (PLL) Assay.
A compound of the present invention can be shown to induce peripheral lymphocyte lowering (PLL).
A. Mouse PLL Assay.
Animals: Male BALB/c mice (Charles River Laboratories, Wilmington, MA) are housed four per cage and maintained in a humidity-controlled (40 to 60%) and temperature- controlled (68 to 72 0F) facility on a 12 h:12 h light/dark cycle (lights on at 6:30 am) with free access to food (Harlan Teklad, Orange, CA, Rodent Diet 8604) and water. Mice are allowed one week of habituation to the animal facility before testing.
PLL Assay: Mice are given an oral dose of a compound of the present invention or dosing vehicle (0.5% methylcellulose) in a total volume of 10 mL/kg. Peripheral blood samples are collected at 5 hours post-dose. The mice are anesthetized with isoflurane and blood is collected via cardiac puncture. A complete cell count (CBC), including lymphocyte count, is obtained using a CELL-DYN® 3700 (Abbott Laboratories, Abbott Park, IL) instrument.
Reduction of the PBL count by the test compounds in comparison with vehicle is indicative of the test compounds exhibiting activity or inducing peripheral lymphocyte lowering.
B. Rat PLL Assay.
Animals: Male Sprague-Dawley rats (Charles River Laboratories, Hollister, CA) were housed and maintained in humidity (40 to 60%) and temperature (68 to 72 0F) controlled facility on a 12 h:12 h light/dark cycle (lights on at 6:30 am) with free access to food (Harlan Teklad, Orange, CA, Rodent Diet 8604) and water. Rats were allowed (approximately) one week of habituation to the animal facility before testing.
PLL Assay: Rats were given a 1 mg/kg intravenous dose of the enantiomer of compound 4 (as reported in Example 1.3), or dosing vehicle (20% hydroxypropyl-cyclodextrin (HPCD)) in a total volume of 1.00 mL/kg. Peripheral blood samples were collected at 5 h post- dose. Blood was collected via indwelling catheter. A complete cell count (CBC), including lymphocyte count, was obtained using a CELL-DYN® 3700 (Abbott Laboratories, Abbott Park, IL) instrument. Results are presented in Figure 1, in which peripheral blood lymphocyte (PBL) count is shown for the 5 hour group. Reduction of the PBL count by the test compound in comparison with vehicle is indicative of the test compound exhibiting activity or inducing peripheral lymphocyte lowering. It is apparent from inspection of Figure 1 that the exemplified enantiomer of compound 4 (as reported in Example 1.3) exhibited activity for inducing PBL lowering (lymphopenia) in the rat.
Example 5: Effect of Compounds on Experimental Autoimmune Encephalomyelitis (EAE). A compound of the present invention can be shown to have therapeutic efficacy in multiple sclerosis by showing it to have therapeutic efficacy in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. In certain exemplary well- established models, EAE is induced in rodents by injection of myelin oligodendrocyte glycoprotein (MOG) peptide, by injection of myelin basic protein (MBP) or by injection of proteolipid protein (PLP) peptide.
A. MOG-induced EAE in Mice.
Animals: Female C57BL/6 mice (8 to 10 weeks of age at start of study) (Jackson Laboratory, Bar Harbor, ME) are housed four per cage and maintained in a humidity-controlled (40-60%) and temperature-controlled (68-72 0F) facility on a 12 h: 12 h light/dark cycle (lights on at 6:30 am) with free access to food (Harlan Teklad, Orange, CA, Rodent Diet 8604) and water. Mice are allowed one week of habituation to the animal facility before testing.
Induction of EAE: Mice are immunized subcutaneously, 50 μL per hind flank, with a total of 100 μg MOG3S-Ss peptide emulsified 1: 1 with complete Freund's adjuvant containing 4 mg/mL heat-killed Mycobacterium tuberculosis. Mice also receive 200 ng pertussis toxin intraperitoneally on the day of immunization and 48 h later.
Clinical scoring: Severity of disease symptoms is scored as follows (in increasing order of severity): 0 = normal; 1 = limp tail OR hind limb weakness; 2 = limp tail AND limb weakness / weakness of 2 or more limbs; 3 = severe limb weakness or single limb paralysis; 4 = paralysis of 2 or more limbs; 5 = death.
Drug treatment: Mice are dosed orally, with vehicle or a test compound, once a day from day 3 until day 21. Dosing volume is 5 mL/kg. The test compound is dosed at, e.g., 1 mg/kg, 3 mg/kg, 10 mg/kg or 30 mg/kg. Mice are weighed daily. Mice are monitored daily from day 7 onward for disease symptoms. After the last dose on day 21, disease progression is monitored daily for 2 more weeks. Reduction of the severity of disease symptoms by the test compound in comparison with vehicle is indicative of the test compound exhibiting therapeutic efficacy in EAE.
B. PLP-induced EAE in Mice.
Animals: Female SJL/J mice (8 to 10 weeks of age at start of study) (Jackson
Laboratory, Bar Harbor, ME) are housed four per cage and maintained in a humidity-controlled (40-60%) and temperature-controlled (68-72 0F) facility on a 12 h: 12 h light/dark cycle (lights on at 6:30 am) with free access to food (Harlan-Teklad Western Res, Orange, CA, Rodent Diet 8604) and water. Mice are allowed one week of habituation to the animal facility before testing.
Induction of EAE: Mice are immunized subcutaneously with 100 μg
PLP139.151 peptide emulsified 1 : 1 with complete Freund's adjuvant containing 4 mg/mL heat- killed Mycobacterium tuberculosis. Mice also receive 200 ng pertussis toxin intravenously on the day of immunization. Clinical scoring: Severity of disease symptoms is scored as follows (in increasing order of severity): 0 = normal; 1 = limp tail OR hind limb weakness; 2 = limp tail AND limb weakness / weakness of 2 or more limbs; 3 = severe limb weakness or single limb paralysis; 4 = paralysis of 2 or more limbs; 5 = death.
Drug treatment: Mice are dosed orally, with vehicle or a test compound, once a day from day 3 until day 21. Dosing volume is 5 ml/kg. The test compound is dosed at, e.g., 1 mg/kg, 3 mg/kg, 10 mg/kg or 30 mg/kg. Mice are weighed daily. Mice are monitored daily from day 7 onward for disease symptoms. After the last dose on day 21, disease progression is monitored daily for two more weeks.
C. MBP-induced EAE in Rats.
Animals: Male Lewis rats (325-375 g at start of study) (Harlan, San Diego, CA) are housed two per cage and maintained in a humidity-controlled (30-70%) and temperature- controlled (20-22 0C) facility on a 12 h:12 h light/dark cycle (lights on at 6:30 A.M.) with free access to food (Harlan-Teklad Western Res., Orange, CA, Rodent Diet 8604) and water. Rats are allowed one week of habituation to the animal facility before testing. During the study, rats are weighed daily prior to clinical scoring at 11 am.
Induction of EAE: Myelin basic protein (MBP; guinea pig) is dissolved in sterile saline at a concentration of 1 mg/ml, and then emulsified 1:1 with complete Freund's adjuvant (1 mg/mL). 50 μL of this emulsion is administered by intraplantar (ipl) injection into both hind paws of each rat, for a total injected volume of 100 μL per rat and a total dose of 50 μg of MBP per rat.
Clinical scoring: Severity of disease symptoms is scored daily after body weighing and before drug dosing. Severity of disease symptoms is scored as follows (in increasing order of severity): 0 = normal; 1 = tail OR limb weakness; 2 = tail AND limb weakness; 3 = severe hind limb weakness or single limb paralysis; 4 = loss of tail tone and paralysis of 2 or more limbs; 5 = death.
Drug treatment: Rats are dosed orally, with vehicle or a test compound, 1 hour prior to MBP injection on day 0 and daily thereafter, after clinical scoring, for the duration of the study. Dosing volume is 5 mL/kg. The test compound is dosed at, e.g., 1 mg/kg, 3 mg/kg, 10 mg/kg or 30 mg/kg. Reduction of the severity of disease symptoms by the test compound in comparison with vehicle is indicative of the test compound exhibiting therapeutic efficacy in EAE.
Example 6: Effect of Compounds on Type I Diabetes.
A compound of the present invention can be shown to have therapeutic efficacy in type I diabetes using an animal model for type I diabetes, such as cyclophosphamide-induced type I diabetes in mice. Animals: Baseline blood glucose measurements are taken from 9-10 week old female NOD/Ltj mice (Jackson Laboratory, Bar Harbor, ME) to ensure that they are normoglycemic (blood glucose is 80-120 mg/dL) prior to initiation of the experiment. Blood glucose is measured from tail bleeds using a OneTouch® Ultra® meter and test strips (LifeScan, Milpitas, CA).
Cyclophosphamide induction of type I diabetes: On day 0 and day 14,
normoglycemic NOD mice are injected intraperitoneally with 4 mg cyclophosphamide monohydrate (200 mg/kg) dissolved in 0.9% saline. If mice are diabetic (blood glucose is >250 mg/dL), they are not given a booster dose of cyclophosphamide on day 14.
Drug Treatment: Mice are dosed orally, with vehicle or test compound, once a day from day 0 until day 25. Compounds are suspended in 0.5% methyl cellulose vehicle using a sonicator to ensure uniform suspension. Mice are weighed twice weekly and are dosed according to weight. Dosing volume is 5 mL/kg. The test compound is dosed at, e.g. , 1 mg/kg, 3 mg/kg, 10 mg/kg or 30 mg/kg. Blood glucose is measured twice weekly. After dosing is completed at day 25, the mice continue to be monitored and blood glucose measurements are taken once a week for 3 weeks. Promotion of normoglycemia by the test compound in comparison with vehicle is indicative of the test compound exhibiting therapeutic efficacy in type I diabetes.
Example 7: Allograft Survival.
A compound of the present invention can be shown to have therapeutic efficacy in prolonging allograft survival by showing it to have therapeutic efficacy in prolonging, e.g., survival of a skin allograft in an animal model.
Animals: Female Balbc/J mice (6 to 7 weeks of age at start of study) (Jackson Laboratory, Bar Harbor, ME) are housed four per cage and maintained in a humidity-controlled (40-60%) and temperature-controlled (68-72 0F) facility on a 12 h: 12 h light/dark cycle (lights on at 6:30 am) with free access to food (Harlan Teklad, Orange, CA, Rodent Diet 8604) and water. Female C57BL/6 mice (8 to 10 weeks of age at start of study) (Jackson Laboratory, Bar Harbor, ME) are similarly housed and maintained. Mice are allowed one week of habituation to the animal facility before testing.
Skin allograft: Balbc/J and C57BL/6 mice are used as donors and recipients, respectively, in a model of skin allograft transplantation. Donor Balbc/J mice are anesthetized, and 0.5 cm - diameter full thickness areas of abdominal skin are surgically removed. Skin grafts harvested from the Balbc/J mice are sutured onto the dorsum of anesthetized recipient C57BL/6 mice. Sutured allografts are covered with Vaseline gauze and Bolster dressing for 7 days. The allografted mice are divided into 8 groups of 8 mice each.
Clinical scoring: Skin allografts are inspected and digital images recorded daily until rejection, which is defined as the first day on which more than 80% of the graft is necrotic. Histological analysis of the rejected graft is carried out on hematoxylin and eosin (H&E)-stained sections. In an optional related study, on post-transplantation day 5 isolated lymphocytes from peripheral lymph nodes and spleen are counted and characterized for activation markers (e.g., T-cell activation markers) by flow cytometry. Also on day 5, grafts are removed from transplanted recipients, cut into small fragments, digested with collagenase and sedimented over Ficoll-Paque (Pharmacia Biotech, Uppsala, Sweden) to isolate graft-infiltrating lymphocytes, which are counted and characterized for activation markers (e.g., T-cell activation markers) by flow cytometry.
Histological analysis of the graft on day 5 can be carried out on hematoxylin and eosin (H&E)- stained sections.
Drug treatment: Mice are dosed orally, with vehicle or test compound, once a day from the day of transplantation until the end of the study, e.g. until day 14, 21 or 28. Dosing volume is 5 mL/kg. The test compound is dosed at, e.g., 1 mg/kg, 3 mg/kg, 10 mg/kg or 30 mg/kg. Delay of time of rejection of the skin allograft by the test compound in comparison with vehicle is indicative of the test compound exhibiting therapeutic efficacy in prolonging skin allograft survival .
Example 8: Effect of Compounds on Colitis.
A compound of the present invention can be shown to have therapeutic efficacy in colitis using an animal model for colitis. Suitable animal models are known in the art (Boismenu et al, J. Leukoc. Biol, 67:267-278, 2000). A first exemplary animal model for colitis is trinitrobenzenesulfonic acid (TNBS)-induced colitis, which presents clinical and
histopathological findings that resemble those in Crohn's disease (Neurath et al., J. Exp. Med., 182: 1281-1290, 1995; Boismenu et al, J. Leukoc. Biol, 67:267-278, 2000). A second exemplary animal model for colitis is dextran sulfate sodium (DSS)-induced colitis, which presents clinical and histopathological findings that resemble those in ulcerative colitis
(Okayasu et al, Gastroenterology, 98:694-702, 1990; Boismenu et al, J. Leukoc. Biol, 67:267- 278, 2000). Compounds can be commercially tested for efficacy in at least DSS-induced colitis and TNBS-induced colitis, e.g. by the Jackson Laboratory (Bar Harbor, ME).
A. Mouse Model for Colitis.
Animals: Male BALB/c mice (6 weeks of age at start of study) (Jackson Laboratory,
Bar Harbor, ME) are housed four per cage and maintained in a humidity-controlled (40-60%) and temperature-controlled (68-72 0F) facility on a 12 h:12 h light/dark cycle (lights on at 6:30 am) with free access to food (Harlan Teklad, Orange CA, Rodent Diet 8604) and water. Mice are allowed one week of habituation to the animal facility before testing.
TNBS induction of colitis: Mice are weighed for baseline body weights and fasted later that day beginning at 6:15 pm just prior to lights-out (day 0). Body weights are taken again the following morning (day 1) at approximately 7:30 am. Mice are anesthetized with isoflurane prior to induction of colitis. Colitis is induced in the mice by intracolonic injection of about 150 mg/kg TNBS in 50% ethanol (in a volume of 150 μL) using an intubation needle (22 g, 1.5 in) inserted completely into the anus with the mouse held by the tail in a vertical position. The mouse is held vertically for 30 additional seconds to allow thorough absorption and minimize leakage, after which the mouse is returned to its cage. Mice are then fed, following the preceding approximately 14 hour of fasting. Each morning thereafter, the mice are weighed. In control experiments, mice receive 50% ethanol alone using the same protocol.
Drug treatment: Drug treatment begins on day 2. Mice are dosed orally, with vehicle or a test compound, once a day from day 2 until the conclusion of the experiment on, e.g., day 7, 14 or 21. Dosing volume is 5 mL/kg. The test compound is dosed at, e.g., 1 mg/kg, 3 mg/kg, 10 mg/kg or 30 mg/kg.
Clinical scoring: Upon conclusion of the experiment, colons are extracted and measured. Mice are euthanized with CO2 and colon is removed from anus to cecum. Excised colon is measured for entire length, length from anus to end of inflamed area and length of inflamed (affected) area. After measurements, colon is cleared of excrement by flushing with saline and then cut open to clear more thoroughly. Colon is then weighed and preserved in neutral buffered formalin (NBF; 10% formalin, pH 6.7-7.0). The colon tissue is embedded in paraffin and processed for hematoxylin and eosin (H & E)-stained sections. Severity of disease symptoms is scored histologically from the stained sections as follows: 0 = no evidence of inflammation; 1 = low level of leukocyte infiltration with infiltration seen in <10% of high-power fields AND no structural changes; 2 = moderate leukocyte infiltration with infiltration seen in 10% to 25% of high-power fields AND crypt elongation AND bowel wall thickening that does not extend beyond the mucosal layer AND no ulcerations; 3 = high level of leukocyte infiltration seen in 25% to 50% of high- power fields AND crypt elongation AND infiltration beyond the mucosal layer AND thickening of the bowel wall AND superficial ulcerations; 4 = marked degree of transmural leukocyte infiltration seen in >50% of high-power fields AND elongated and distorted crypts AND bowel wall thickening AND extensive ulcerations. Reduction of the severity of the disease symptoms by the test compound in comparison with vehicle is indicative of the test compound exhibiting therapeutic efficacy in colitis.
B. Rat Model for Colitis.
Animals: Male Wistar rats (175-200 g at start of study) (Charles River Laboratories, Wilmington, MA) are housed two per cage and maintained in a humidity-controlled (40-60%) and temperature-controlled (68-72 0F) facility on a 12 h:12 h light/dark cycle (lights on at 6:30am) with free access to food (Harlan Teklad, Orange CA, Rodent Diet 8604) and water. Rats are allowed one week of habituation to the animal facility before testing.
TNBS induction of colitis: Rats are weighed for baseline body weights and fasted later that day beginning at 6: 15 pm just prior to lights-out (day 0). Body weights are taken again the following morning (day 1) at approximately 7:30 am. Rats are anesthetized with isoflurane pπor to induction of colitis Colitis is induced in the rats by intracolonic injection of about 60 mg/kg TNBS m 50% ethanol (m a volume of 500 μL) using a fabπcated intubation needle (7 5 Fr umbilical catheter and 14 g hub) inserted 8 cm into the anus with the rat held by the tail m a vertical position. The rat is held vertically for 30 additional s to allow thorough absorption and minimize leakage, after which the rat is returned to its cage. Rats are then fed, following the preceding approximately 14 h of fasting. Each morning thereafter, the rats are weighed. In control expeπments, rats receive 50% ethanol alone using the same protocol.
Drug treatment: Drug treatment begins on day 2. Rats are dosed orally, with vehicle or test compound, once a day from day 2 until the conclusion of the expeπment on, e g , day 7, 14 or 21. Dosing volume is 5 mL/kg. Test compound is dosed at, e g , 1 mg/kg, 3 mg/kg, 10 mg/kg or 30 mg/kg.
Clinical scoring: Upon conclusion of the expeπment, colons are extracted and measured Rats are euthanized with CO2 and colon is removed from anus to cecum. Excised colon is measured for entire length, length from anus to end of inflamed area, and length of inflamed (affected) area. After measurements, colon is cleared of excrement by flushing with saline and then cut open to clear more thoroughly. Colon is then weighed and preserved in neutral buffered formalin (NBF, 10% formalin, pH 6.7-7.0). The colon tissue is embedded in paraffin and processed for hematoxylin and eosm (H & E)-stamed sections. Seventy of disease symptoms is scored histologically from the stained sections as follows: 0 = no evidence of inflammation; 1 = low level of leukocyte infiltration with infiltration seen m <10% of high-power fields AND no structural changes; 2 = moderate leukocyte infiltration with infiltration seen in 10% to 25% of high-power fields AND crypt elongation AND bowel wall thickening that does not extend beyond the mucosal layer AND no ulcerations; 3 = high level of leukocyte infiltration seen in 25% to 50% of high-power fields AND crypt elongation AND infiltration beyond the mucosal layer AND thickening of the bowel wall
AND superficial ulcerations; 4 = marked degree of transmural leukocyte infiltration seen in >50% of high-power fields AND elongated and distorted crypts AND bowel wall thickening AND extensive ulcerations. Reduction of the seventy of the disease symptoms by the test compound in companson with vehicle is indicative of the test compound exhibiting therapeutic efficacy in colitis.
Example 9: Effects of Compounds on Cardiac Telemetry in the Rat.
Animals: Male Sprague-Dawley rats (250-300 g at time of surgery) are implanted by Charles River Laboratones (Wilmington, MA) with cardiac transmitting devices (Data Sciences PhysioTel C50-PXT) into the pentoneal space, with a pressure-sensing catheter inserted into the descending aorta. Rats are allowed at least one week to recover. Rats are housed in individual cages and maintained in a humidity-controlled (30-70%) and temperature-controlled (20-22 0C) facility on a 12 h:12 h light/dark cycle (lights on at 7:00 am) with free access to food (Harlan- Teklad, Orange, CA, Rodent Diet 8604) and water. Rats are allowed one week of habituation to the animal facility before testing.
Measurement of cardiovascular parameters: The implanted transmitting devices transmit continuous measurements of blood pressure (systolic, diastolic, mean arterial, pulse), heart rate, body temperature, and motor activity in freely moving conscious animals. These data are transmitted via radiofrequency to a computer which bin the data into 1 min averages using DataSciences ART software. Telemetry recording takes place over a 21-h period, starting at noon and continuing until 9:00 am the following day. A maximum of eight rats are tested at a time, and the same eight rats are utilized for all treatment groups in a within-subject design.
Drug treatment: Rats are injected orally with vehicle or compound at 1 :00 pm. A full study (vehicle + 3 doses) requires four separate testing sessions, which occur on Mondays- Tuesdays and Thursdays-Fridays. During each of the testing sessions, the eight rats are divided into four treatment groups such that each group comprises N = 2 for any given session. Rats are re-tested in subsequent testing sessions in a crossover design such that by the end of the four sessions, all animals receive all treatments in a pseudo-random order, and each group comprises N = 8.
Exemplary bradycardia assay: It is expressly contemplated that the rats can be used to show that a compound of the invention has no or substantially no activity for bradycardia. By way of illustration and not limitation, the rats are administered vehicle or a test compound and heart rate is then measured over a 120 min period. No or substantially no reduction of heart rate in response to the test compound in comparison with vehicle is indicative of the test compound exhibiting no or substantially no activity for bradycardia.
Example 10: Effect of Compounds on Arthritis.
Female Lewis rats can be used in this study to determine that a compound of the invention has an effect on arthritis. Acclimated animals are anesthetized with isoflurane and given the first collagen injection (day 0). On day 6, the animals are anesthetized again for the second collagen injection. Collagen is prepared by making a 4 mg/mL solution in 0.01 N acetic acid. Equal volumes of collagen and incomplete Freund's adjuvant are emulsified by hand mixing until a bead of this material holds its form when placed in water. Each animal receives 300 μL of the mixture each time, spread over 3 subcutaneous sites on the back.
Treatment (p.o., q.d., 5 mL/kg dosing volume) begins on day 0 and continued through day 16 with vehicle or compounds given at 24 h intervals. Rats are weighed on days 0, 3, 6 and 9 through 17 and caliper measurements of the ankles taken on days 9 through 17. A reduction of mean ankle diameter in the treated animals compared to untreated animals is indicative of the compound exhibiting a positive effect on arthritis as described by this animal model. Those skilled in the art will recognize that various modifications, additions, substitutions and variations to the illustrative examples set forth herein can be made without departing from the spirit of the invention and are, therefore, considered within the scope of the invention.

Claims

CLAIMS What is claimed is:
1. A compound selected from compounds of Formula (Ia) and pharmaceutically
acceptable salts, solvates, and hydrates thereof:
Figure imgf000081_0001
(Ia)
wherein:
E is N or CR1;
G is N or CR3;
R1 is selected from the group consisting Of C1-C6 alkyl, Ci-C6 alkylsulfonyl, cyano, C1-C6 haloalkyl, C1-C6 haloacyl, and halogen;
R2, R3, and R4 are each independently selected from the group consisting of H, C1-C6 alkoxy, C]-C6 alkyl, cyano, C3-C7 cycloalkyl, C3-C7 cycloalkyloxy, C1-C6 haloalkoxy, Ci-C6 haloalkyl, and halogen; and
R5 is H or halogen.
2. The compound according to claim 1, wherein E is N.
3. The compound according to claim 1, wherein E is CR1.
4. The compound according to claim 3, wherein R1 is selected from the group consisting of chloro, cyano, fluoro, iodo, methyl, methylsulfonyl, trifluoroacetyl, and trifluoromethyl.
5. The compound according to claim 3, wherein R1 is chloro or fluoro.
6. The compound according to any one of claims 1 to 5, wherein G is CR3.
7. The compound according to any one of claims 1 to 6, wherein R2, R3, and R4 are each independently selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, fluoro, iodo, isopropoxy, methoxy, methyl, trifluoromethoxy, and trifluoromethyl.
8. The compound according to any one of claims 1 to 7, wherein R2 is selected from the group consisting of H, chloro, cyano, methoxy, methyl, trifluoromethoxy, and trifluoromethyl .
9. The compound according to any one of claims 1 to 8, wherein R3 is selected from the group consisting of H, cyclohexyl, cyclopentyl, and isopropoxy.
10. The compound according to any one of claims 1 to 9, wherein R4 is selected from the group consisting of H, chloro, cyano, methoxy, methyl, trifluoromethoxy, and trifluoromethyl.
11. The compound according to any one of claims 1 to 5, wherein G is N.
12. The compound according to claim 11, wherein R2 and R4 are each independently Ci-C6 alkyl or halogen.
13. The compound according to claim 11 or 12, wherein R2 and R4 are each independently chloro or methyl.
14. The compound according to any one of claims 1 to 13, wherein R5 is H or F.
15. The compound according to any one of claims 1 to 13, wherein R5 is H.
16. The compound according to claim 1, wherein:
R1 is selected from the group consisting of chloro, cyano, fluoro, iodo, methyl, methylsulfonyl, trifluoroacetyl, and trifluoromethyl;
R2, R3, and R4 are each independently selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, fluoro, iodo, isopropoxy, methoxy, methyl, trifluoromethoxy, and trifluoromethyl; and
R5 is H or F.
17. The compound according to claim 1, selected from compounds of Formula (Ib) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000083_0001
wherein:
R1 is selected from the group consisting of CpCe alkyl, Ci-C6 alkylsulfonyl, Ci- C6 haloalkyl, Ci-C6 haloacyl, cyano, and halogen; and
R2, R3, and R4 are each independently selected from the group consisting of H, Ci-C6 alkoxy, cyano, C3-C7 cycloalkyl, CrC6 haloalkoxy, CrC6 haloalkyl, and halogen.
18. The compound according to claim 1, selected from compounds of Formula (Ib) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000083_0002
wherein:
R1 is selected from the group consisting of chloro, cyano, fluoro, iodo, methyl, methylsulfonyl, trifluoroacetyl, and trifluoromethyl; and
R2, R3, and R4 are each independently selected from the group consisting of H, chloro, cyano, cyclohexyl, cyclopentyl, fluoro, iodo, isopropoxy, methoxy,
trifluoromethoxy, and trifluoromethyl.
19. The compound according to claim 1, selected from compounds of Formula (Ib) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000083_0003
wherein:
R! is selected from the group consisting of chloro, cyano, fluoro, iodo, methyl, methylsulfonyl, trifluoroacetyl, and trifluoromethyl;
R2 is selected from the group consisting of H, chloro, cyano, fluoro, methoxy, trifluoromethoxy, and trifluoromethyl; R3 is selected from the group consisting of H, cyclohexyl, cyclopentyl, and isopropoxy; and
R4 is selected from the group consisting of H, chloro, cyano, fluoro, methoxy, trifluoromethoxy, and trifluoromethyl.
20. The compound according to claim 1, selected from compounds of Formula (Ic) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000084_0001
(Ic)
wherein:
R1 is selected from the group consisting Of Ci-C6 alkyl, Q-C6 alkylsulfonyl, Cr
C6 haloalkyl, cyano, and halogen; and
R2 and R4 are each independently selected from the group consisting of cyano, C1-C6 alkoxy, Ci-C6 haloalkoxy, Ci-C6 haloalkyl, and halogen.
21. The compound according to claim 1, selected from compounds of Formula (Ic) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000084_0002
(Ic)
wherein:
R1 is selected from the group consisting of methyl, trifluoromethyl, chloro, cyano, methylsulfonyl, chloro, iodo, and fluoro; and
R2 and R4 are each independently from the group consisting of cyano, methoxy, trifluoromethoxy, and trifluoromethyl.
22. The compound according to claim 1, selected from compounds of Formula (Ic) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000085_0001
wherein:
R1 is selected from the group consisting of methyl, trifluoromethyl, cyano, methylsulfonyl, chloro, iodo, and fluoro;
R2 is cyano; and
R4 is trifiuoromethoxy.
23. The compound according to claim 1 , selected from compounds of Formula (Ie) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000085_0002
wherein:
R1 is CrC6 haloacyl or halogen; and
R2 and R3 are each independently selected from the group consisting of Ci -C6 alkoxy, cyano, C3-C7 cycloalkyl, and halogen.
24. The compound according to claim 1, selected from compounds of Formula (Ie) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000085_0003
wherein:
R1 is Cj-C6 haloacyl or halogen;
R2 is cyano, Ci-C6 haloalkyl, or halogen; and
R3 is Ci-C6 alkoxy or C3-C7 cycloalkyl.
25. The compound according to claim 1, selected from compounds of Formula (Ie) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000086_0001
(Ie)
wherein:
R1 is chloro or trifluoroacetyl;
R2 is cyano, fluoro, or trifluoromethyl; and
R3 is cyclohexyl, cyclopentyl, isopropoxy, or methoxy.
26. A compound according to claim 1, selected from the following compounds and
pharmaceutically acceptable salts, solvates, and hydrates thereof:
2-(7-(5-(3-cyano-4-isopropoxyphenyl)-l,2,4-oxadiazol-3-yl)-9-(2,2,2- trifluoroacetyO^^-dihydro-lH-pyrrolofl^-aJindol-l-yOacetic acid;
2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-fluoro- 2 ,3 -dihydro- 1 H-pyrrolo [ 1 ,2-a]indol- 1 -yl)acetic acid;
2-(7-(5 -(3 -cyano-5 -(trifluoromethoxy)phenyl)- 1 ,2,4-oxadiazol-3-yl)- 1 ,9- difluoro-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid;
2-(9-chloro-7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-
2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid;
2-(9-chloro-7-(5-(3-cyano-4-cyclohexylphenyl)-l,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l ,2-a]indol-l -yl)acetic acid;
2-(7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-9-iodo-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid;
2-(7-(5-(3 -cyano-5 -(trifluoromethoxy)phenyl)- 1 ,2,4-oxadiazol-3 -yl)-9-methyl- 2,3 -dihydro- lH-pyrrolo[ 1 ,2-a]indol- 1 -yl)acetic acid;
2-(7-(5 -(3 -cyano-5 -(trifluoromethoxy)phenyl)- 1 ,2,4-oxadiazol-3 -yl)-9- (trifluoromethyl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid;
2-(9-cyano-7-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-
2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid;
2-(7-(5 -(3 -cyano-5 -(trifluoromethoxy)phenyl)- 1 ,2,4-oxadiazol-3 -yl)-9- (methylsulfonyl)-2,3-dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid;
2-(7-(5-(3,5-bis(trifluoromethyl)phenyl)-l,2,4-oxadiazol-3-yl)-9-chloro-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid;
2-(9-chloro-7-(5 -(4-cyclopentyl-3 -(trifluoromethyl)phenyl)- 1 ,2,4-oxadiazol-3 - yl^^-dihydro-lH-pyπolofl^-aJindol-l-yOacetic acid; 2-(9-chloro-7-(5-(2-chloro-6-methylpyridin-4-yl)-l,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid;
2-(9-chloro-7-(5-(3-cyano-5-methoxyphenyl)-l,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l ,2-a]indol-l -yl)acetic acid;
2-(6-(5-(3-cyano-5-(trifluoromethoxy)phenyl)-l,2,4-oxadiazol-3-yl)-2,3- dihydro- 1 H-benzo[d]pyrrolo[ 1 ,2-a]imidazol-3 -yl)acetic acid;
2-(9-chloro-7-(5-(3-chloro-5-(trifluoromethoxy) phenyl)-l,2,4-oxadiazol-3-yl)- 2,3-dihydro- lH-pyrrolo[ 1 ,2-a]indol-l -yl)acetic acid;
2-(9-chloro-7-(5-(3-fluoro-4-methoxyphenyl)-l,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid; and
2-(9-chloro-7-(5-(4-isopropoxy-3-methoxyphenyl)-l,2,4-oxadiazol-3-yl)-2,3- dihydro-lH-pyrrolo[l,2-a]indol-l-yl)acetic acid.
27. The compound according to any one of claims 1 to 26, wherein the stereochemistry of the C(I) ring carbon of said compound is R.
28. The compound according to any one of claims 1 to 26, wherein the stereochemistry of the C(I) ring carbon of said compound is S.
29. A pharmaceutical composition comprising a compound according to any one of claims 1 to 28 and a pharmaceutically acceptable carrier.
30. A method for treating an SlPl receptor-associated disorder in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound according to any one of claims 1 to 28, or a pharmaceutical composition according to claim 29.
31. A method for treating a disorder in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound according to any one of claims 1 to 28, or a pharmaceutical composition according to claim 29, wherein said disorder is selected from the group consisting of psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy, glomerulosclerosis, gastritis, polymyositis, thyroiditis, vitiligo, hepatitis, and biliary cirrhosis.
32. A method for treating psoriasis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound according to any one of claims 1 to 28, or a pharmaceutical composition according to claim 29.
33. A method for treating multiple sclerosis in an individual comprising administering to said individual in need thereof a therapeutically effective amount of a compound according to any one of claims 1 to 28, or a pharmaceutical composition according to claim 29.
34. Use of a compound according to any one of claims 1 to 28 in the manufacture of a medicament for the treatment of an SlPl receptor-associated disorder.
35. Use of a compound according to any one of claims 1 to 28 in the manufacture of a medicament for the treatment of an SlPl receptor-associated disorder selected from the group consisting of psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy,
glomerulosclerosis, gastritis, polymyositis, thyroiditis, vitiligo, hepatitis, and biliary cirrhosis.
36. Use of a compound according to any one of claims 1 to 28 in the manufacture of a medicament for the treatment of psoriasis.
37. Use of a compound according to any one of claims 1 to 28 in the manufacture of a medicament for the treatment of multiple sclerosis.
38. A compound according to any one of claims 1 to 28 for use in a method for the
treatment of the human or animal body by therapy.
39. A compound according to any one of claims 1 to 28 for use in a method for the
treatment of an SlPl receptor-associated disorder.
40. A compound according to any one of claims 1 to 28 for use in a method for the
treatment of an SlPl receptor-associated disorder selected from the group consisting of psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy, glomerulosclerosis, gastritis, polymyositis, thyroiditis, vitiligo, hepatitis, and biliary cirrhosis.
41. A compound according to any one of claims 1 to 28 for use in a method for the
treatment of psoriasis.
42. A compound according to any one of claims 1 to 28 for use in a method for the
treatment of multiple sclerosis.
43. A process for preparing a composition comprising admixing a compound according to any one of claims 1 to 28 and a pharmaceutically acceptable carrier.
PCT/US2010/001787 2009-06-23 2010-06-22 Disubstituted oxadiazole derivatives useful in the treatment of autoimmune and inflammatory disorders WO2011005290A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US26926909P 2009-06-23 2009-06-23
US61/269,269 2009-06-23

Publications (1)

Publication Number Publication Date
WO2011005290A1 true WO2011005290A1 (en) 2011-01-13

Family

ID=42785552

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/001787 WO2011005290A1 (en) 2009-06-23 2010-06-22 Disubstituted oxadiazole derivatives useful in the treatment of autoimmune and inflammatory disorders

Country Status (1)

Country Link
WO (1) WO2011005290A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8415484B2 (en) 2008-08-27 2013-04-09 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US8580841B2 (en) 2008-07-23 2013-11-12 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US8853419B2 (en) 2010-01-27 2014-10-07 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
WO2015066515A1 (en) * 2013-11-01 2015-05-07 Receptos, Inc. Selective sphingosine 1 phosphate receptor modulators and combination therapy therewith
US9085581B2 (en) 2010-03-03 2015-07-21 Arena Pharmaceuticals, Inc. Processes for the preparation of S1P1 receptor modulators and crystalline forms thereof
US9388147B2 (en) 2009-11-13 2016-07-12 Celgene International II Sárl Selective sphingosine 1 phosphate receptor modulators and methods of chiral synthesis
US9394264B2 (en) 2009-11-13 2016-07-19 Receptos, Inc. Sphingosine 1 phosphate receptor modulators and methods of chiral synthesis
US9481659B2 (en) 2011-05-13 2016-11-01 Celgene International Ii Sàrl Selective heterocyclic sphingosine 1 phosphate receptor modulators
WO2018028557A1 (en) * 2016-08-08 2018-02-15 南京明德新药研发股份有限公司 Tricyclic compound and use thereof
WO2018237379A3 (en) * 2017-06-23 2019-02-07 Enzo Biochem, Inc. Sphingosine pathway modulating compounds for the treatment of cancers
US10301262B2 (en) 2015-06-22 2019-05-28 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compund1) for use in SIPI receptor-associated disorders
EP3492465A4 (en) * 2016-07-22 2020-03-04 Shijiazhuang Sagacity New Drug Development Co., Ltd. S1p1 agonist and application thereof
US10660879B2 (en) 2017-06-23 2020-05-26 Enzo Biochem, Inc. Sphingosine pathway modulating compounds for the treatment of cancers
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11478448B2 (en) 2017-02-16 2022-10-25 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis
US11555015B2 (en) 2018-09-06 2023-01-17 Arena Pharmaceuticals, Inc. Compounds useful in the treatment of autoimmune and inflammatory disorders

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008076356A1 (en) * 2006-12-15 2008-06-26 Abbott Laboratories Novel oxadiazole compounds
EP2003132A1 (en) * 2006-04-03 2008-12-17 Astellas Pharma Inc. Hetero compound
WO2009094157A1 (en) * 2008-01-25 2009-07-30 Arena Pharmaceuticals, Inc. Dihydro- 1h- pyrrolo [1,2-a] indol-1-yl carboxylic derivatives which act as s1p1 agonists

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2003132A1 (en) * 2006-04-03 2008-12-17 Astellas Pharma Inc. Hetero compound
WO2008076356A1 (en) * 2006-12-15 2008-06-26 Abbott Laboratories Novel oxadiazole compounds
WO2009094157A1 (en) * 2008-01-25 2009-07-30 Arena Pharmaceuticals, Inc. Dihydro- 1h- pyrrolo [1,2-a] indol-1-yl carboxylic derivatives which act as s1p1 agonists

Non-Patent Citations (100)

* Cited by examiner, † Cited by third party
Title
BALATONI ET AL., BRAIN RESEARCH BULLETIN, vol. 74, 2007, pages 307 - 316
BAUMRUKER ET AL., EXPERT OPIN. INVESTIG. DRUGS, vol. 16, 2007, pages 283 - 289
BAUMRUKER ET AL., EXPERT. OPIN. INVESTIG. DRUGS, vol. 16, 2007, pages 283 - 289
BERGE ET AL., JOURNAL OFPHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
BOISMENU ET AL., J LEUKOC. BIOL., vol. 67, 2000, pages 267 - 278
BOISMENU ET AL., J. LEUKOC. BIOL., vol. 67, 2000, pages 267 - 278
BOLICK ET AL., ARTERIOSCLER. THROMB. VASC. BIOL., vol. 25, 2005, pages 976 - 981
BRINKMANN ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 21453 - 21457
BRINKMANN ET AL., PHARMACOLOGY & THERAPEUTICS, vol. 115, 2007, pages 84 - 105
BRINKMANN ET AL., TRANSPLANT PROC., vol. 33, 2001, pages 530 - 531
BRINKMANN ET AL., TRANSPLANTATION, vol. 72, 2001, pages 764 - 769
BRINKMANN, PHARMACOLOGY & THERAPEUTICS, vol. 115, 2007, pages 84 - 105
BUDDE ET AL., J. AM. SOC. NEPHROL., vol. 13, 2002, pages 1073 - 1083
CHIBA ET AL., CELL MOL. BIOL., vol. 3, 2006, pages 11 - 19
CHIBA, PHARMACOLOGY & THERAPEUTICS, vol. 108, 2005, pages 308 - 319
CHUN ET AL., PHARMACOLOGICAL REVIEWS, vol. 54, 2002, pages 265 - 269
COELHO ET AL., J. PHARMACOL. EXP. THER., vol. 323, 2007, pages 626 - 635
COLLIER, T. L., J. LABELLED COMPD. RADIOPHARM., vol. 42, 1999, pages S264 - S266
D. NAKASHIMA ET AL., J. INVESTIGATIVE DERMATOLOGY, vol. 128, no. 12, 2008, pages 2833 - 2841
DANIEL ET AL., J. IMMUNOL., vol. 178, 2007, pages 2458 - 2468
DEGUCHI ET AL., ONCOLOGY REPORTS, vol. 16, 2006, pages 699 - 703
DEV ET AL., PHARMACOLOGY AND THERAPEUTICS, vol. 117, 2008, pages 77 - 93
E. BAR-HAIM ET AL., PLOS PATHOGENS, vol. 4, no. 11, 2008
FU ET AL., TRANSPLANTATION, vol. 73, 2002, pages 1425 - 1430
FUJII ET AL., AM. J. PHYSIOL. GASTROINTEST. LIVER PHYSIOL., vol. 291, 2006, pages G267 - G274
FUJINO ET AL., J. PHARMACOL. EXP. THER., vol. 305, 2003, pages 70 - 77
FUJISHIRO ET AL., J. HEART LUNG TRANSPLANT, vol. 25, 2006, pages 825 - 833
G. P. STAHLY, CRYSTAL GROWTH & DESIGN, vol. 7, no. 6, 2007, pages 1007 - 1026
GABRIEL ET AL., ASSAY AND DRUG DEVELOPMENT TECHNOLOGIES, vol. 1, 2003, pages 291 - 303
HALE ET AL., BIOORG. MED. CHEM. LETT., vol. 14, 2004, pages 3351 - 3355
HERZINGER ET AL., AM. J. CLIN. DERMATOL., vol. 8, 2007, pages 329 - 336
HWANG ET AL., CIRCULATION, vol. 100, 1999, pages 1322 - 1329
IDZKO ET AL., J. CLIN. INVEST., vol. 116, 2006, pages 2935 - 2944
ISHII ET AL., NATURE, 8 February 2009 (2009-02-08)
JOHAN GROENEVELD, VASCUL. PHARMACOL., vol. 39, 2003, pages 247 - 256
JUNG ET AL., GLIA, vol. 55, 2007, pages 1656 - 1667
KANEIDER ET AL., FASEB J., vol. 18, 2004, pages 309 - 311
KAPPOS ET AL., N. ENGL. J. MED., vol. 355, 2006, pages 1124 - 1140
KATAOKA ET AL., CELLULAR & MOLECULAR IMMUNOLOGY, vol. 2, 2005, pages 439 - 448
KAUDEL ET AL., TRANSPLANT. PROC, vol. 39, 2007, pages 499 - 502
KEUL ET AL., ARTERIOSCLER. THROMB. VASC. BIOL., vol. 27, 2007, pages 607 - 613
KIM ET AL., CELL SIGNAL, vol. 16, 2004, pages 89 - 95
KIMURA ET AL., STEM CELLS, vol. 25, 2007, pages 115 - 124
KIYABAYASHI ET AL., J. CARDIOVASC. PHARMACOL., vol. 35, 2000, pages 410 - 416
KOHNO ET AL., BIOL. PHARM. BULL., vol. 27, 2004, pages 1392 - 1396
KORECK ET AL., DERMATOLOGY, vol. 206, 2003, pages 96 - 105
KUROSE ET AL., EXP. EYE RES., vol. 70, 2000, pages 7 - 15
LAMONTAGNE ET AL., CANCER RES., vol. 66, 2006, pages 221 - 231
LE BAS, M.-D., J. LABELLED COMPD. RADIOPHARM., vol. 44, 2001, pages S280 - S282
LEE ET AL., CLIN. CANCER RES., vol. 11, 2005, pages 84588466
LIMA ET AL., TRANSPLANT PROC., vol. 36, 2004, pages 1015 - 1017
LIU ET AL., MICROSURGERY, vol. 27, 2007, pages 300 - 304
MAKI ET AL., TRANSPLANTATION, vol. 74, 2002, pages 1684 - 1686
MAKI ET AL., TRANSPLANTATION, vol. 79, 2005, pages 1051 - 1055
MARTINI ET AL., AM. J. PHYSIOL. RENAL PHYSIOL., vol. 292, 2007, pages F1761 - F1770
MARTINI ET AL., EXPERT OPIN. INVESTIG. DRUGS, vol. 16, 2007, pages 505 - 518
MATLOUBIAN ET AL., NATURE, vol. 427, 2004, pages 355
MATLOUBIAN ET AL., NATURE, vol. 427, 2004, pages 355 - 360
MATSUURA ET AL., INFLAMM. RES., vol. 49, 2000, pages 404 - 410
MATSUURA ET AL., INT. J. IMMUNOPHARMACOL., vol. 22, 2000, pages 323 - 331
MIRON ET AL., ANN. NEUROL., vol. 63, 2008, pages 61 - 71
MIYAMOTO ET AL., J. AM. COLL. CARDIOL., vol. 37, 2001, pages 1713 - 1718
MIZUSHIMA ET AL., INFLAMM. BOWEL DIS., vol. 10, 2004, pages 182 - 192
NEURATH ET AL., J. EXP. MED., vol. 182, 1995, pages 1281 - 1290
NOFER ET AL., CIRCULATION, vol. 115, 2007, pages 501 - 508
O. COSTU ET AL., JOURNAL OF CELLULAR AND MOLECULAR MEDICINE, vol. 12, no. 3, 2008, pages 995 - 1004
OGAWA ET AL., BBRC, vol. 361, 2007, pages 621 - 628
OKAYASU ET AL., GASTROENTEROLOGY, vol. 98, 1990, pages 694 - 702
OKAZAKI ET AL., J. RHEUMATOL., vol. 29, 2002, pages 707 - 716
OO ET AL., J. BIOL. CHEM., vol. 282, 2007, pages 9082 - 9089
PAN ET AL., CHEMISTRY & BIOLOGY, vol. 13, 2006, pages 1227 - 1234
PREMENKO-LANIER ET AL., NATURE, vol. 454, 2008, pages 894
RAUSCH ET AL., J. MAGN. RESON. IMAGING, vol. 20, 2004, pages 16 - 24
RAVENEY ET AL., ARCH. OPHTHALMOL., vol. 126, no. 10, 2008, pages 1390
ROSEN ET AL., IMMUNOL. REV., vol. 195, 2003, pages 160 - 177
SAKAGAWA ET AL., TRANSPL. IMMUNOL., vol. 13, 2004, pages 161 - 168
SANCHEZ ET AL., J. BIOL. CHEM., vol. 278, no. 47, 2003, pages 47281 - 47290
SANNA ET AL., J. BIOL. CHEM., vol. 279, 2004, pages 13839 - 13848
SANNA ET AL., JBIOL CHEM., vol. 279, 2004, pages 13839 - 13848
SANNA ET AL., NAT CHEM BIOL., vol. 2, 2006, pages 434 - 441
SAUER ET AL., J. BIOL. CHEM., vol. 279, 2004, pages 38471 - 38479
SAWICKA ET AL., J. IMMUNOL., vol. 171, 2003, pages 6206 - 6214
SCHMID ET AL., J. CELL BIOCHEM., vol. 101, 2007, pages 259 - 270
SCHWAB ET AL., NATURE IMMUNOL., vol. 8, 2007, pages 1295 - 1301
SHIMIZU ET AL., CIRCULATION, vol. 111, 2005, pages 222 - 229
SUZUKI ET AL., J. HEART LUNG TRANSPLANT, vol. 25, 2006, pages 302 - 209
SUZUKI ET AL., TRANSPL. IMMUNOL., vol. 4, 1996, pages 252 - 255
T. KOHONO ET AL., BIOLOGICAL & PHARMACEUTICAL BULLETIN, vol. 28, no. 4, 2005, pages 736 - 739
TAYLOR ET AL., BLOOD, vol. 110, 2007, pages 3480 - 3488
TRUONG ET AL., AMERICAN JOURNAL OF TRANSPLANTATION, vol. 7, 2007, pages 2031 - 2038
VILLULLAS ET AL., J. NEUROSCI. RES., vol. 73, 2003, pages 215 - 226
WEBB ET AL., J. NEUROIMMUNOL., vol. 153, 2004, pages 108 - 121
WEBSTER, CUTIS, vol. 76, 2005, pages 4 - 7
WHETZEL ET AL., CIRC. RES., vol. 99, 2006, pages 731 - 739
YAN ET AL., BIOORG. & MED. CHEM. LETT., vol. 16, 2006, pages 3679 - 3683
YANAGAWA ET AL., J. IMMUNOL., vol. 160, 1998, pages 5493 - 5499
YANG ET AL., CLINICAL IMMUNOLOGY, vol. 107, 2003, pages 30 - 35
ZHANG ET AL., J. CELL. MOL. MED., vol. 11, 2007, pages 307 - 314
ZHANG ET AL., MINI-REVIEWS IN MEDICINAL CHEMISTRY, vol. 7, 2007, pages 845 - 850
ZHU, G-D., J ORG. CHEM., vol. 67, 2002, pages 943 - 948

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8580841B2 (en) 2008-07-23 2013-11-12 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US9522133B2 (en) 2008-07-23 2016-12-20 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US9126932B2 (en) 2008-07-23 2015-09-08 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US9108969B2 (en) 2008-08-27 2015-08-18 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US8415484B2 (en) 2008-08-27 2013-04-09 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US9388147B2 (en) 2009-11-13 2016-07-12 Celgene International II Sárl Selective sphingosine 1 phosphate receptor modulators and methods of chiral synthesis
US9394264B2 (en) 2009-11-13 2016-07-19 Receptos, Inc. Sphingosine 1 phosphate receptor modulators and methods of chiral synthesis
US10239846B2 (en) 2009-11-13 2019-03-26 Celgene International Ii Sàrl Selective sphingosine 1 phosphate receptor modulators and methods of chiral synthesis
US9175320B2 (en) 2010-01-27 2015-11-03 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US9447041B2 (en) 2010-01-27 2016-09-20 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US8853419B2 (en) 2010-01-27 2014-10-07 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US11674163B2 (en) 2010-01-27 2023-06-13 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US11149292B2 (en) 2010-01-27 2021-10-19 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US9085581B2 (en) 2010-03-03 2015-07-21 Arena Pharmaceuticals, Inc. Processes for the preparation of S1P1 receptor modulators and crystalline forms thereof
US9481659B2 (en) 2011-05-13 2016-11-01 Celgene International Ii Sàrl Selective heterocyclic sphingosine 1 phosphate receptor modulators
WO2015066515A1 (en) * 2013-11-01 2015-05-07 Receptos, Inc. Selective sphingosine 1 phosphate receptor modulators and combination therapy therewith
US11896578B2 (en) 2015-01-06 2024-02-13 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US10301262B2 (en) 2015-06-22 2019-05-28 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compund1) for use in SIPI receptor-associated disorders
US10676435B2 (en) 2015-06-22 2020-06-09 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound 1) for use in SIPI receptor-associated disorders
US11884626B2 (en) 2015-06-22 2024-01-30 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound1) for use in S1P1 receptor-associated disorders
US11091435B2 (en) 2015-06-22 2021-08-17 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3, 4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(compound1) for use in S1P1 receptor-associated disorders
RU2754845C2 (en) * 2016-07-22 2021-09-08 Медшайн Дискавери Инк. S1p1 agonist and its application
US11192886B2 (en) 2016-07-22 2021-12-07 Medshine Discovery Inc. S1P1 agonist and application thereof
EP3492465A4 (en) * 2016-07-22 2020-03-04 Shijiazhuang Sagacity New Drug Development Co., Ltd. S1p1 agonist and application thereof
WO2018028557A1 (en) * 2016-08-08 2018-02-15 南京明德新药研发股份有限公司 Tricyclic compound and use thereof
CN109071469A (en) * 2016-08-08 2018-12-21 南京明德新药研发股份有限公司 Tricyclic compounds and its application
CN109071469B (en) * 2016-08-08 2022-04-05 南京明德新药研发有限公司 Tricyclic compound and application thereof
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis
US11478448B2 (en) 2017-02-16 2022-10-25 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations
WO2018237379A3 (en) * 2017-06-23 2019-02-07 Enzo Biochem, Inc. Sphingosine pathway modulating compounds for the treatment of cancers
US10660879B2 (en) 2017-06-23 2020-05-26 Enzo Biochem, Inc. Sphingosine pathway modulating compounds for the treatment of cancers
US10675255B2 (en) 2017-06-23 2020-06-09 Enzo Bochem, Inc. Sphingosine pathway modulating compounds for the treatment of cancers
US10278960B2 (en) 2017-06-23 2019-05-07 Enzo Biochem, Inc. Sphingosine pathway modulating compounds for the treatment of cancers
US11865105B2 (en) 2017-06-23 2024-01-09 Enzo Biochem, Inc. Sphingosine pathway modulating compounds for the treatment of cancers
US10646459B2 (en) 2017-06-23 2020-05-12 Enzo Biochem, Inc. Sphingosine pathway modulating compounds for the treatment of cancers
US10881643B2 (en) 2017-06-23 2021-01-05 Enzo Biochem, Inc. Sphingosine pathway modulating compounds for the treatment of cancers
US11555015B2 (en) 2018-09-06 2023-01-17 Arena Pharmaceuticals, Inc. Compounds useful in the treatment of autoimmune and inflammatory disorders

Similar Documents

Publication Publication Date Title
US20200407316A1 (en) SUBSTITUTED (1,2,3,4-TETRAHYDROCYCLOPENTA[b]INDOL-3-YL)ACETIC ACID DERIVATIVES USEFUL IN THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISORDERS
WO2011005290A1 (en) Disubstituted oxadiazole derivatives useful in the treatment of autoimmune and inflammatory disorders
EP2342205B1 (en) Substituted tricyclic acid derivatives as s1p1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
WO2009094157A1 (en) Dihydro- 1h- pyrrolo [1,2-a] indol-1-yl carboxylic derivatives which act as s1p1 agonists
EP2542554B1 (en) Processes for the preparation of s1p1 receptor modulators and crystalline forms thereof
WO2009151621A1 (en) Substituted (1, 2, 4-0xadiaz0l-3-yl) indolin-1-yl carboxylic acid derivatives useful as s1p1 agonists
WO2011005295A1 (en) Modulators of the sphingosine-1-phosphate (s1p) receptor useful for the treatment of disorders related thereto
WO2009151626A1 (en) Substituted (1, 2, 4-0xadiaz0l-3-yl) indolin-1-yl carboxylic acid derivatives useful as s1p1 agonists

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: 10728458

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: 10728458

Country of ref document: EP

Kind code of ref document: A1