Deuterated compounds as immunomodulators

文档序号:1131533 发布日期:2020-10-02 浏览:26次 中文

阅读说明:本技术 作为免疫调节剂的氘代化合物 (Deuterated compounds as immunomodulators ) 是由 樊平臣 R·M·路易 V·R·马里 R·辛格 曾一斌 张朋烈 于 2018-10-29 设计创作,主要内容包括:提供化合物来调节C5a受体。这些化合物具有下式(I),包括其立体异构体及药学上可接受的盐,其中R<Sup>1</Sup>,R<Sup>2</Sup>和R<Sup>3</Sup>如本文所定义。还公开了与此类化合物的制备和使用有关的方法,以及包含此类化合物的药物组合物。<Image he="629" wi="669" file="DDA0002474404000000011.GIF" imgContent="drawing" imgFormat="GIF" orientation="portrait" inline="no"></Image>(Compounds are provided for modulating the C5a receptor. These compounds have the following formula (I), including stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1 ,R 2 And R 3 As defined herein. Also disclosed are methods relating to the preparation and use of such compounds, as well as pharmaceutical compositions comprising such compounds.)

1. A compound of formula (I)

Figure FDA0002474403980000011

Or a pharmaceutically acceptable salt thereof, wherein:

R1is selected from CH3,CH2D,CHD2,CD3Or CD2OH;

R2Is selected from CH3,CH2D,CHD2,CD3,CH2OH, CDHOH or CD2OH;

R3Is H or D;

R4is H or D;

m is an integer of 0 to 6;

n is an integer of 0 to 3;

o is an integer from 0 to 4;

p is an integer between 0 and 9; and

q is an integer of 0 to 3;

wherein said compound has at least one deuterium atom in an amount of at least 80%.

2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R1Is selected from CH3Or CD3

3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R2Is selected from CH3,CD3,CH2OH or CD2OH。

4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R3Is D.

5. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R3And R4Each is H.

6. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein subscript n is 0, 1, or 2.

7. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein subscript o is 0 or 1.

8. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein subscript q is 0 or 1.

9. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein subscript p is 0, 1, 4, or 9.

10. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein subscript m is 0.

11. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein subscript m is 0; subscript n is 0, 1, or 2; subscript o is 0 or 1; subscript p is 0, 1, 4, or 9; subscript q is 0 or 1.

12. A compound according to any one of claims 1 to 11 in the form of a pharmaceutically acceptable salt.

13. A pharmaceutical composition comprising a compound of any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

14. The pharmaceutical composition of claim 13, formulated for oral, intravenous, transdermal or subcutaneous administration.

15. The pharmaceutical composition of claim 13 or 14, further comprising one or more additional therapeutic agents.

16. The pharmaceutical composition of claim 15, wherein the one or more additional therapeutic agents are selected from the group consisting of: corticosteroids, steroids, immunosuppressive agents, immunoglobulin G agonists, dipeptidyl peptidase IV inhibitors, lymphocyte function anti-3 receptor antagonists, interleukin-2 ligands, interleukin-1 beta ligand inhibitors, IL-2 receptor alpha subunit inhibitors, HGF gene stimulators, IL-6 antagonists, IL-5 antagonists, alpha 1 antitrypsin stimulators, cannabinoid receptor antagonists, histone deacetylase inhibitors, AKT protein kinase inhibitors, CD20 inhibitors, Abl tyrosine kinase inhibitors, JAK tyrosine kinase inhibitors, TNF alpha ligand inhibitors, hemoglobin modulators, TNF antagonists, proteasome inhibitors, CD3 modulators, Hsp 70 family inhibitors, immunoglobulin agonists, CD30 antagonists, tubulin antagonists, sphingosine-1-phosphate receptor-1 agonists, connective tissue growth factor ligand inhibitors, caspase inhibitors, corticotropin ligands, Btk tyrosine kinase inhibitors, complement C1s subcomponent inhibitors, erythropoietin receptor agonists, B lymphocyte stimulator ligand inhibitors, cyclin-dependent kinase-2 inhibitors, P-selectin glycoprotein ligand 1 stimulators, mTOR inhibitors, elongation factor 2 inhibitors, cell adhesion molecule inhibitors, factor XIII agonists, calcineurin inhibitors, immunoglobulin G1 agonists, inosine monophosphate dehydrogenase inhibitors, complement C1s subcomponent inhibitors, thymidine kinase modulators, cytotoxic T-lymphocyte protein 4 modulators, angiotensin II receptor antagonists, angiotensin II receptor modulators, TNF superfamily receptor 12A antagonists, CD52 antagonists, adenosine deaminase inhibitors, t cell differentiation antigen CD6 inhibitors, FGF-7 ligands, dihydroorotate dehydrogenase inhibitors, Syk tyrosine kinase inhibitors, type I interferon receptor antagonists, interferon alpha ligand inhibitors, macrophage migration inhibitory factor inhibitors, integrin alpha-V/beta-6 antagonists, cysteine protease stimulators, p38MAP kinase inhibitors, TP53 gene inhibitors, shiga-like toxin I inhibitors, fucosyltransferase 6 stimulators, interleukin 22 ligands, IRS1 gene inhibitors, protein kinase alpha stimulators, protein kinase C inhibitors, CD74 antagonists, immunoglobulin gamma Fc receptor IIB antagonists, T cell antigen CD7 inhibitors, CD95 antagonists, N-acetyl mannosamine kinase stimulators, cardiotrophin 1 ligands, leukocyte elastase inhibitors, CD40 ligand receptor antagonists, CD40 ligand modulators, IL-17 antagonists, TLR-2 antagonists, mannan-binding lectin serine protein-2 (MASP-2) inhibitors, factor B inhibitors, factor D inhibitors, C3aR modulators, C5aR2 modulators, T cell receptor antagonists, chemokine receptors, in particular antagonists of CXCR1, CXCR2, CXCR3, CXCR4, CXCR7, CCR1, CCR2, CCR3, CCR4, CCR5, CCR7, CCR7, CCR9, CX3CR1 and CXCR6, and combinations thereof.

17. The pharmaceutical composition of any one of claims 15 or 16, wherein the one or more additional therapeutic agents are selected from the group consisting of: obituzumab, rituximab, oclizumab, cyclophosphamide, prednisone, hydrocortisone acetate, cortisone acetate, teicok polyphenol pivalate, prednisolone, methylprednisolone, triamcinolone, pyruvic acid, triamcinolone acetonide, methasone, fluocinolone acetonide, betamethasone sodium phosphate, dexamethasone sodium phosphate, fluocortolone, hydrocortisone 17-valerate, halometone, amitriptone, dipropionate, beclomethasone, betamethasone-17-propionate, fluocinolone hexanoate, fluocinolone pivalate, fluorobutadiene acetate, hydrocortisone 17-butyrate, hydrocortisone 17-acetoacetate, hydrocortisone 17-butyrate, ciclesonide and Poncinerate, GB-0998, immunoglobulins, Begolizumab, Afacit, leucocyte, Gevacizumab, daclizumab, Baliximab, Illinomab, pemoline, Serlukumab, tositulizumab, Clarazlizumab, Mapelizumab, fingolimod, Potiramistat, Triciclabine, nilotinib, Imatinib, tofacitinib, Mobilatinib, Africancitinib, Italiancitinib, Infosiximab, PEG-bHb-CO, Enscipril, Isazozomib, bortezomib, Mulitumumab, Oxicizumab, guanrititumus, Bentuximab, Pelamitocimox, KRP-203, FG-3019, Enlicarsyn, corticotropin, Imutamide, Lutissu, Luzertin, Tatarin, beta-cell polyethylene glycol-erythropolides, belimumab, bricibimod, asexicept, celecoxib, eculizumab, everolimus, sirolimus, denileukin-toxin linker, LMB-2, natalizumab, catrocco, cyclosporine, tacrolimus, woosporin, kanamynuumab, mycophenolate, imidazolinine, CE-1145, TK-DLI, abacavir, belicept, olmesartan medoxomil, slasonetanuma, TXA-127, BIIB-023, alilimumab, pentostatin, eritizumab, palivizumab, leflunomide, PRO-140, seirivierlotinib, fostamatinib, aninovumab, simuzumab, BAX-069, BG-00011, rockimab, QPI-1002, tzumt-101, tzu-652, raparisin, ladanisin, PTX-9908, acannil, APH-703, soterelin, milatuzumab, SM-101, Twei, APG-101, DEX-M74, cardiotrophin-1, Diprustat, ASKP-1240, BMS-986004, HPH-116, KD-025, OPN-305, TOL-101, defibrillation peptide, pomamine, thymocyte protein, laquinimod, ramistem cell-L, anti-thymocyte immunoglobulin, Stanpill, LIV-gamma, otta 10%, T2c-001, 99 mTc-methoxyisobutylisonitrile, krameride, pesobulba, pridopamonit, laramuranumab peptide, ATIR-101, BPX-501, BPX-01, ALLO-ASC-DFACP, irbesartan + propaggermanium, Apo cells, cannabidiol, RGI-2001, orotic acid, anti-CD 3 diabody-diphtheria toxin conjugate, NOX-100, LT-1951, OMS721, ALN-CC5, ACH-4471, AMY-101, Acthar gel and CD4+ CD25+ regulatory T cells, MEDI7814, P32, P59, CCX354, CCX721, CCX9588, CCX140, CCX872, CCX598, CCX6239, CCX587, CCX624, CCX282, CCX025, CCX507, CCX430, CCX765, CCX758, CCX771, CCX662, CCX650, and combinations thereof.

18. A method of treating a human suffering from or susceptible to a disease or condition involving pathological activation of the C5a receptor, the method comprising administering to a mammal a therapeutically effective amount of a compound according to any one of claims 1 to 16 or a pharmaceutical composition according to any one of claims 13 to 17.

19. The method of claim 18, wherein the disease or disorder is an inflammatory disease or disorder, an autoimmune disease or a neoplastic disease or disorder.

20. The method of claim 18, wherein the disease or disorder is a cardiovascular or cerebrovascular disorder.

21. The method of claim 18, wherein the disease or disorder is selected from the group consisting of: neutropenia, neutrophilia, Wegener's granulomatosis, microscopic polyangiitis, C3-glomerulopathy, C3-glomerulonephritis, dense deposit disease, membranoproliferative glomerulonephritis, Kawasaki disease, sepsis, septic shock, hemolytic uremic syndrome, atypical hemolytic uremic syndrome (aHUS), Alzheimer's disease, multiple sclerosis, stroke, inflammatory bowel disease, chronic obstructive pulmonary disease, inflammation associated with burns, lung injury, osteoarthritis, atopic dermatitis, chronic urticaria, ischemia reperfusion injury, acute respiratory distress syndrome, systemic inflammatory response syndrome, multiple organ dysfunction syndrome, uveitis, tissue transplant rejection, hyperacute rejection of transplanted organs, myocardial infarction, coronary thrombosis, vascular occlusion, post-operative vascular reocclusion, arteriosclerosis, polypoid choroidal vasculopathy, traumatic blindgut central nervous system injury, ischemic heart disease, rheumatoid arthritis, systemic lupus erythematosus, guillain-barre syndrome, pancreatitis, lupus nephritis, lupus glomerulonephritis, psoriasis, crohn's disease, vasculitis, ANCA vasculitis, irritable bowel syndrome, multiple bowel syndrome, dermatitis, pemphigus, scleroderma, myasthenia gravis, autoimmune hemolysis and thrombocytopenia, goodpasture's syndrome, immune vasculitis, graft-versus-host disease, paroxysmal nocturnal hemoglobinuria, sjogren's syndrome, insulin-dependent diabetes mellitus, lupus nephritis, glomerulonephritis, IGA nephropathy, membranoproliferative glomerulonephritis, antiphospholipid syndrome, age-related macular degeneration; dry age related macular degeneration, wet age related macular degeneration, motor neuron disease, contact sensitivity reactions, hidradenitis suppurativa or inflammation caused by blood contact with artificial surfaces.

22. The method of claim 18, wherein the disease or disorder is selected from the group consisting of: neutropenia, neutrophilia, wegener's granulomatosis, microscopic polyangiitis, C3-glomerulopathy, C3-glomerulonephritis, dense deposit disease, membranoproliferative glomerulonephritis, kawasaki disease, hemolytic uremia, atypical pneumolytic uremia syndrome (aHUS), tissue transplant rejection, hyperacute rejection of transplanted organs, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, lupus glomerulonephritis, vasculitis, ANCA vasculitis, autoimmune hemolytic and thrombocytopenic states, immune vasculitis, graft versus host disease nephropathy, haman nephritis, membranous nephritis, glomerulonephritis, IGA nephropathy, suppurative hidradenitis, or membranoproliferative glomerulonephritis.

23. The method of claim 18, wherein the disease or disorder is selected from the group consisting of: melanoma, lung cancer, lymphoma, sarcoma, carcinoma, fibrosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, angiosarcoma, lymphangiosarcoma, synovioma, mesothelioma, meningioma, leukemia, lymphoma smooth muscle sarcoma, rhabdomyosarcoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, papillary carcinoma, cystadenoadenocarcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatocellular carcinoma, transitional cell carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, wild-type cell carcinoma, polymorphic papilloma, renal tubular adenoma, bladder adenoma, papilloma, adenoma, leiomyoma, rhabdomyoma, hemangioma, lymphangioma, osteoma, chondroma, lipoma, or fibroma.

24. The method of any one of claims 18 to 23, further comprising administering to the human a therapeutically effective amount of one or more additional therapeutic agents.

25. The method of claim 24, wherein one or more additional therapeutic agents are selected from the group consisting of: corticosteroids, steroids, immunosuppressive agents, immunoglobulin G agonists, dipeptidyl peptidase IV inhibitors, lymphocyte function anti-3 receptor antagonists, interleukin-2 ligands, interleukin-1 beta ligand inhibitors, IL-2 receptor alpha subunit inhibitors, HGF gene stimulators, IL-6 antagonists, IL-5 antagonists, alpha 1 antitrypsin stimulators, cannabinoid receptor antagonists, histone deacetylase inhibitors, AKT protein kinase inhibitors, CD20 inhibitors, Abl tyrosine kinase inhibitors, JAK tyrosine kinase inhibitors, TNF alpha ligand inhibitors, hemoglobin modulators, TNF antagonists, proteasome inhibitors, CD3 modulators, Hsp 70 family inhibitors, immunoglobulin agonists, CD30 antagonists, tubulin antagonists, sphingosine-1-phosphate receptor-1 agonists, connective tissue growth factor ligand inhibitors, caspase inhibitors, corticotropin ligands, Btk tyrosine kinase inhibitors, complement C1s subcomponent inhibitors, erythropoietin receptor agonists, B lymphocyte stimulator ligand inhibitors, cyclin-dependent kinase-2 inhibitors, P-selectin glycoprotein ligand 1 stimulators, mTOR inhibitors, elongation factor 2 inhibitors, cell adhesion molecule inhibitors, factor XIII agonists, calcineurin inhibitors, immunoglobulin G1 agonists, inosine monophosphate dehydrogenase inhibitors, complement C1s subcomponent inhibitors, thymidine kinase modulators, cytotoxic T-lymphocyte protein 4 modulators, angiotensin II receptor antagonists, angiotensin II receptor modulators, TNF superfamily receptor 12A antagonists, CD52 antagonists, adenosine deaminase inhibitors, t cell differentiation antigen CD6 inhibitors, FGF-7 ligands, dihydroorotate dehydrogenase inhibitors, Syk tyrosine kinase inhibitors, type I interferon receptor antagonists, interferon alpha ligand inhibitors, macrophage migration inhibitory factor inhibitors, integrin alpha-V/beta-6 antagonists, cysteine protease stimulators, p38MAP kinase inhibitors, TP53 gene inhibitors, shiga-like toxin I inhibitors, fucosyltransferase 6 stimulators, interleukin 22 ligands, IRS1 gene inhibitors, protein kinase alpha stimulators, protein kinase C inhibitors, CD74 antagonists, immunoglobulin gamma Fc receptor IIB antagonists, T cell antigen CD7 inhibitors, CD95 antagonists, N-acetyl mannosamine kinase stimulators, cardiotrophin 1 ligands, leukocyte elastase inhibitors, CD40 ligand receptor antagonists, CD40 ligand modulators, IL-17 antagonists, TLR-2 antagonists, mannan-binding lectin serine protein-2 (MASP-2) inhibitors, factor B inhibitors, factor D inhibitors, C3aR modulators, C5aR2 modulators, T cell receptor antagonists, chemokine receptors, in particular antagonists of CXCR1, CXCR2, CXCR3, CXCR4, CXCR7, CCR1, CCR2, CCR3, CCR4, CCR5, CCR7, CCR7, CCR9, CX3CR1 and CXCR6, and combinations thereof.

26. The method of claim 24 or 25, wherein the one or more additional therapeutic agents are selected from the group consisting of: obituzumab, rituximab, oclizumab, cyclophosphamide, prednisone, hydrocortisone acetate, cortisone acetate, teicok polyphenol pivalate, prednisolone, methylprednisolone, triamcinolone, pyruvic acid, triamcinolone acetonide, methasone, fluocinolone acetonide, betamethasone sodium phosphate, dexamethasone sodium phosphate, fluocortolone, hydrocortisone 17-valerate, halometone, amitriptone, dipropionate, beclomethasone, betamethasone-17-propionate, fluocinolone hexanoate, fluocinolone pivalate, fluorobutadiene acetate, hydrocortisone 17-butyrate, hydrocortisone 17-acetoacetate, hydrocortisone 17-butyrate, ciclesonide and Poncinerate, GB-0998, immunoglobulins, Begolizumab, Afacit, leucocyte, Gevacizumab, daclizumab, Baliximab, Illinomab, pemoline, Serlukumab, tositulizumab, Clarazlizumab, Mapelizumab, fingolimod, Potiramistat, Triciclabine, nilotinib, Imatinib, tofacitinib, Mobilatinib, Africancitinib, Italiancitinib, Infosiximab, PEG-bHb-CO, Enscipril, Isazozomib, bortezomib, Mulitumumab, Oxicizumab, guanrititumus, Bentuximab, Pelamitocimox, KRP-203, FG-3019, Enlicarsyn, corticotropin, Imutamide, Lutissu, Luzertin, Tatarin, beta-cell polyethylene glycol-erythropolides, belimumab, bricibimod, asexicept, celecoxib, eculizumab, everolimus, sirolimus, denileukin-toxin linker, LMB-2, natalizumab, catrocco, cyclosporine, tacrolimus, woosporin, kanamynuumab, mycophenolate, imidazolinine, CE-1145, TK-DLI, abacavir, belicept, olmesartan medoxomil, slasonetanuma, TXA-127, BIIB-023, alilimumab, pentostatin, eritizumab, palivizumab, leflunomide, PRO-140, seirivierlotinib, fostamatinib, aninovumab, simuzumab, BAX-069, BG-00011, rockimab, QPI-1002, tzumt-101, tzu-652, raparisin, ladanisin, PTX-9908, acarinil, APH-703, Sotelastelin, Miratuzumab, SM-101, Twei, APG-101, DEX-M74, cardiotrophin-1, Tipralatte, ASKP-1240, BMS-986004, HPH-116, KD-025, OPN-305, TOL-101, defibrillation peptide, Bomaliamine, Thymoglobulin, Laquinmod, Ramiphragma cells, anti-thymocyte immunoglobulin, Stanpal, LIV-gamma, Autagar 10%, T2c-001, 99 mTc-methoxyisobutylisonitrile, Clariti, Prossoba, Podoprilipinab, Laprilizumab, Furamuzumab, ATIR-101, BPX-501, ACP-01, ALLO-ASC-U, Irbesartan + Geranium, apo cells, cannabidiol, RGI-2001, orotic acid, anti-CD 3 bivalent antibody-diphtheria toxin conjugate, NOX-100, LT-1951, OMS721, ALN-CC5, ACH-4471, AMY-101, Acthar gel and CD4+ CD25+ regulatory T cells, MEDI7814, P32, P59, CCX354, CCX721, CCX9588, CCX140, CCX872, CCX598, CCX6239, CCX587, CCX624, CCX282, CCX025, CCX507, CCX430, CCX765, CCX758, CCX771, CCX662, CCX650 and combinations thereof.

Background

The complement system plays a central role in the clearance of immune complexes and immune responses to infectious agents, foreign antigens, virus-infected cells and tumor cells. Inappropriate or excessive reaction of the complement system can lead to deleterious, and possibly even life-threatening, consequences and tissue destruction due to severe inflammation. These consequences are manifested clinically in a variety of diseases, including septic shock; myocardial and intestinal ischemia/reperfusion injury; transplant rejection; organ failure; nephritis; pathological inflammation and autoimmune diseases.

The complement system consists of a group of proteins that are normally present in the serum in an inactive state. Activation of the complement system mainly involves three different pathways, namely the classical pathway, the alternative pathway and the lectin pathway (v.m. holers, clinical immunology: principles and practice, 1 st edition, r.r. rich code, mossbi press; 1996, 363-: 1) the classical pathway is a calcium/magnesium dependent cascade, usually activated by the formation of an antigen-antibody complex. Activation can also be by antibody-independent means, i.e., complexing ligands by binding to C-reactive protein, and binding to many pathogens including gram-negative bacteria. 2) An alternative pathway is a magnesium-dependent cascade that can be activated by deposition and activation of C3 on certain sensitive surfaces (e.g., cell wall polysaccharides of yeast and bacteria, as well as certain biopolymer materials). 3) The lectin pathway involves initial binding of mannose-binding lectin followed by activation of C2 and C4, which is common in the classical pathway (Matsushita, M. et al., J.Exp.Med.176:1497-1502 (1992); C.Suankratay et al, J.Immunol.160:3006-3013 (1998)).

Activation of the complement pathway produces biologically active fragments of complement proteins, such as C3a, C4a, and C5a anaphylatoxins, and C5b-9 Membrane Attack Complex (MAC), all by affecting leukocyte chemotaxis; activating macrophages, neutrophils, platelets, mast cells and endothelial cells; and increase vascular permeability, cytolysis and tissue damage to mediate inflammatory responses.

Complement C5a is one of the most potent pro-inflammatory mediators in the complement system. (the inflammatory response elicited by the allergic C5a peptide was 100-fold stronger than C3a on a molar basis.) C5a is the activated form of C5 (190kD, molecular weight). The content of C5a in human serum was about 80. mu.g/ml (Kohler, P.F., et al, J.Immunol.99:1211-1216(1967)). it consists of two polypeptide chains, α and β, with molecular weights of about 115kD and 75kD, respectively (Tack, B.F., et al, Biochemistry 18: 1490-1497 (1979)). As a single-stranded pre-molecule, biosynthetic C5 is cleaved enzymatically into a double-stranded structure during processing and secretion. After cleavage, the two chains are held together by at least one disulfide bond and non-covalent interactions (Ooi, Y.M. et al, J.Immunol.124:2494-2498 (1980)).

During activation of the complement pathway, C5 is cleaved into C5a and C5b fragments. For the classical pathway, the invertase responsible for C5 activation is the multi-subunit complex of C4b, C2a and C3b, and for the alternative pathway (C3b)2The multi-subunit complex of Bb and P (Goldust, M.B. et al, J.Immunol.113:998- & 1007 (1974); Schreiber, R.D. et al, Proc. Natl.Acad.Sci.75:3948- & 3952 (1978)). C5 is activated by cleavage at position 74-75 of the α -strand (Arg-Leu). after activation, 11.2kD, 7 kDThe 4 amino acid peptide C5a was released from the amino terminal portion of the α -chain C5a and C3a are both potent stimulators of neutrophils and monocytes (Schindeler, R. et al., Blood 76: 1631-.

In addition to being anaphylactotoxic, C5a also induced chemotactic migration of neutrophils (Ward, P.A. et al, J.Immunol.102:93-99(1969)), eosinophils (Kay, A.B. et al, Immunol.24: 969-252976 (1973)), basophils (Lett-Brown, M.A. et al, J.Immunol.117: 246-2521976)) and monocytes (Snyderman, R. et al, Proc.Soc.Exp.biol.Med.138: 387-3901971)). Both C5a and C5b-9 activate endothelial cells to express adhesion molecules essential for sequestering activated leukocytes that mediate tissue Inflammation and injury (Formaman, K.E. et al, J.Clin. invest.94: 1147-Bufonis 1155 (1994); Formaman, K.E. et al, Inflammation 20:1-9 (1996); Rollins, S.A. et al, Transplantation 69:1959-1967(2000)) C5a also mediate inflammatory responses by causing smooth muscle contraction, increasing vascular permeability, inducing basophil and mast cell degranulation and inducing release of lysosomal proteases and oxidative radicals (Gerard, C. et al, Ann. Rev. munol.12:775-808 (1994)). In addition, C5a regulates hepatic acute phase gene expression and enhances the overall immune response by increasing the production of TNF- α, IL-1- β, IL-6, IL-8, prostaglandins, and leukotrienes (Lambris, J.D., et al, journal of the human complement System in Health and Disease, "Volanakis, J.E., eds., MD Limited (Marcel Dekker), New York, pp.83-118).

The allergic and chemotactic effects of C5a are thought to be mediated through its interaction with the C5a receptor. The human C5a receptor (C5aR) is a 52kD membrane-bound G protein-coupled receptor expressed on neutrophils, monocytes, basophils, eosinophils, hepatocytes, lung smooth muscle and endothelial cells and glomerular tissue (Van-Epps, D.E. et al, J.Immunol.132: 2862. cndot. 2867 (1984); Haviland, D.L. et al, J.Immunol.154: 1861. cndot. 1869 (1995); Wetsel, R.A., Immunol.Leff.44: 183. cndot. 187 (1995); Buchner, R.R. et al, J.Immunol.155: 308. cndot. 315 (1995); Chenoweth, D.E. J.Natl. Acad. cndot. Sci.75:3943 (1978); Zwirner, J.877. cndot. J.884: 884)). The ligand binding site of C5aR is complex and consists of at least two physically separable binding domains. One bound the C5a amino terminus (amino acids 1-20) and disulfide-linked core (amino acids 21-61), while the second bound the C5a carboxy terminus (amino acids 62-74) (Wetsel, R.A., curr. Opin. Immunol.7:48-53 (1995)).

C5a plays an important role in inflammation and tissue injury. In cardiopulmonary bypass and hemodialysis, activation of the alternative complement pathway results in C5a formation when human blood is brought into contact with artificial surfaces of the heart-lung machine or kidney dialysis machine (Howard, R.J. et al, Arch.Surg.123: 1496-. C5a causes capillary permeability and edema, bronchoconstriction, pulmonary vasoconstriction, leukocyte and platelet activation and increased penetration of tissues, particularly lung tissues (Czermak, b.j. et al, j.leukc.biol.64: 40-48 (1998)). Administration of an anti-C5 a monoclonal antibody has been shown to reduce coronary endothelial dysfunction caused by cardiopulmonary bypass and cardiac arrest (Tofukuji, M. et al., J.Thorac.Cardiovasc.Surg.116: 1060-.

C5a is also implicated in Acute Respiratory Distress Syndrome (ARDS), Chronic Obstructive Pulmonary Disease (COPD) and Multiple Organ Failure (MOF) (Hack, C.E. et al, am.J.Med.1989:86: 20-26; Hammerschmidt DE et al Lancet 1980; 1: 947-949; Heideman M. et al J.Trauma 1984; 4: 1038-1043; Marc, MM et al, am.J.Respir.celland mol. biol.,2004:31: 216-219). C5a enhances the production of two important proinflammatory cytokines, TNF- α and IL-1, by monocytes. C5a also has been shown to play an important role in the development of tissue damage, particularly lung damage, in animal models of septic shock (Smedegard G et al, am. J. Pathol. 1989; 135: 489-497; Markus, S et al, FASEB Journal (2001),15: 568-570). anti-C5 a antibody administered to animals prior to endotoxin or E.coli treatment reduces tissue damage and reduces IL-6 production in models of sepsis using rats, pigs and non-human primates (Smedegard, G et al, am. J. Pathol.135:489-497 (1989); Hopken, U et al, Eur. J. Immunol.26:1103-1109 (1996); Stevens, J.H et al, J.Clin. invest.77:1812-1816 (1986)). More importantly, blocking C5a with the polyclonal anti-C5 a antibody has been shown to significantly improve survival in the rat caecal ligation/puncture model (Czermak, B.J., et al, nat. Med.5:788-792 (1999)). This model shares many aspects of the clinical manifestations of sepsis in humans. (Parker, S.J et al, Br. J. Surg.88:22-30(2001)) in the same sepsis model, anti-C5 a antibody was shown to inhibit thymocyte apoptosis (Guo, R.F et al, J.Clin. invest.106: 1271-Bus1280 (2000)) and prevent MOF (Huber-Lang, M et al, J.Immunol.166: 1193-Bus1199 (2001)). anti-C5 a antibody also had protective effects in the cobra venom factor model of rat lung injury and in immune complex-induced lung injury (Mulligan, M.S. et al, J.Clin. invest.98:503-512 (1996)). The importance of C5a in immune complex-mediated lung injury was later demonstrated in mice (Bozic, C.R., et al, Science 26:1103-1109 (1996)).

C5a was found to be the primary mediator of myocardial ischemia-reperfusion injury. Complement depletion reduces myocardial infarct size in mice (Weisman, H.F et al, Science 249: 146-151(1990)) and treatment with anti-C5 a antibody reduces injury in the hindlimb ischemia-reperfusion rat model (Bless, N.M et al, am. J. physiol.276: L57-L63 (1999)). Reperfusion injury during myocardial infarction was also significantly reduced in pigs treated with monoclonal anti-C5 a IgG (Amsterdam, E.A., et al, am.J.Physiol.268: H448-H457 (1995)). Recombinant human C5aR antagonists reduced infarct size in a porcine model of surgical revascularization (Riley, R.D et al, J.Thorac.Cardiovasc.Surg.120:350-358 (2000)).

C5 a-driven neutrophils also cause a number of bullous diseases (e.g., bullous pemphigus, pemphigus vulgaris and pemphigus foliaceus). These are chronic and recurrent inflammatory diseases, clinically characterized by the presence of sterile blisters in the sub-epidermal space of the skin and mucosa. While autoantibodies to keratinocytes located on the basal membrane of the skin are believed to be the basis for the separation of epidermal basal keratinocytes from the underlying basal membrane, the blebs are also characterized by accumulation of neutrophils in the upper layers of the dermis and in the vacuolar cavity. In experimental models, the reduction of neutrophils or the absence of complement (total or C5 selective) can inhibit the formation of subcutaneous blisters, even in the presence of high autoantibody titers.

Patients with rheumatoid arthritis (Jose, P.J et al, Ann. Rheum. Dis.49:747-752 (1990); Grant, E.P et al, J.of exp. Med.,196(11):1461-1471, (2002)), lupus nephritis (Bao, L et al, Eur. J.of Immunol.,35(8),2496-2506, (2005)) and Systemic Lupus Erythematosus (SLE) (Porcel, J.M et al, Clin. Immunol. Immunopathol.74:283-288(1995)) have elevated levels of complement. C5a levels correlate with the severity of the disease state. Collagen-induced arthritis in mice and rats resembles human rheumatoid arthritis disease. Mice lacking the C5a receptor showed complete protection against arthritis induced by injection of monoclonal anti-collagen Abs (Banda, N.K et al, J.of Immunol.,2003,171: 2109-2115). Thus, inhibition of the C5a and/or C5a receptor (C5aR) may be useful in the treatment of these chronic diseases.

The complement system is thought to be activated in patients with Inflammatory Bowel Disease (IBD) and is thought to play a role in the pathogenesis of the disease. Activated complement products are found on the surface epithelial luminal surface as well as in the muscularis mucosae and submucosal vessels of IBD patients (Woodruff, T.M et al., J. of Immunol.,2003,171: 5514-one 5520)).

C5aR is up-regulated on reactive astrocytes, microglia and endothelial cells in the inflamed human central nervous system (Gasque, P et al, am. J. Pathol.150:31-41 (1997)). C5a may be involved in neurodegenerative diseases such as Alzheimer' S disease (Mukherjee, P et al, J.Neurommunol.105: 124-. Activation of neuronal C5aR may induce apoptosis (FarkasI et al J. physiol.1998; 507: 679-687). Thus, inhibition of C5a and/or C5aR may also be useful for treating neurodegenerative diseases.

There is evidence that the production of C5a exacerbates inflammation associated with atopic dermatitis (Neuber, K et al, Immunology 73:83-87, (1991)) and chronic urticaria (Kaplan, A.P., J.allergy Clin. immunol.114; 465-474, (2004)).

Psoriasis is now known to be a T cell mediated disease (Gottlieb, E.L et al, nat. Med.1:442-447 (1995)). However, neutrophils and mast cells may also be involved in the pathogenesis of the disease (Terui, T et al, exp. Dermatol.9: 1-10; 2000); werfel, T et al, Arch.Dermatol.Res.289:83-86 (1997)). Accumulation of neutrophils under the stratum corneum was observed in highly inflamed areas of psoriatic plaques, and psoriatic lesion (scale) extracts contained high levels of C5a and had strong chemotactic activity for neutrophils, which could be inhibited by the addition of C5a antibody. T cells and neutrophils are chemotactic and attracted by C5a (Nataf, S et al, J.Immunol.162: 4018-. Furthermore, C5aR expression has been demonstrated on plasmacytoid dendritic cells (pdcs) isolated from cutaneous lupus erythematosus lesions, and these cells show chemotactic behavior for C5a, suggesting that blocking C5aR on pdcs may be effective in reducing pDC infiltration into inflamed skin in SLE and psoriasis. Thus, C5a may be an important therapeutic target for the treatment of psoriasis.

Immunoglobulin G-containing Immune Complexes (IC) play pathophysiological roles in a number of autoimmune diseases, such as systemic lupus erythematosus, rheumatoid arthritis, Sjogren's disease, Goodpasture's syndrome and hypersensitivity pneumonitis (Madaio, M.P., Semin. Nephrol.19:48-56 (1999); Korganow, A.S et al, Immunity 10:451-459 (1999); Bolten, W.K., Kidney Int.50:1754-1760 (1996); Ando, M et al, Current. Opin. Pulm. Med.3:391-399 (1997)). These diseases are highly heterogeneous and often affect one or more of the following organs: skin, blood vessels, joints, kidneys, heart, lungs, nervous system and liver (including cirrhosis and liver fibrosis). The classical animal model of inflammatory response in these IC diseases is the Arthus response, which is characterized by polymorphonuclear cell infiltration, hemorrhage and plasma exudation (Arthus, M., C.R.Soc.biol.55:817-824 (1903)). Recent studies have shown that C5aR deficient mice are protected from IC-induced tissue damage (Kohl, J et al, mol. Immunol.36: 893-1070 (1999); Baumann, U et al, J. Immunol.164:1065-1070 (2000)). The results are consistent with those observed that small peptide anti-C5 aR antagonists inhibit the inflammatory response caused by IC deposition (Strachan, A.J et al, J.Immunol.164: 6560-. C5a and its receptor together play an important role in the pathogenesis of IC disease. Inhibitors of C5a and C5aR may be useful in the treatment of these diseases.

Disclosure of Invention

In one aspect, provided herein are compounds having formula (I):

or a pharmaceutically acceptable salt thereof, wherein:

R1is selected from CH3,CH2D,CHD2Or CD3Or CD2OH。

R2Is selected from CH3,CH2D,CHD2,CD3,CH2OH, CDHOH or CD2OH。

R3Is H or D;

R4is H or D;

m is an integer of 0 to 6;

n is an integer of 0 to 3;

o is an integer from 0 to 4;

p is an integer between 0 and 9; and

q is an integer of 0 to 3;

wherein said compound has at least one deuterium atom present in an amount of at least 80%.

In addition to the compounds provided herein, the invention also provides pharmaceutical compositions comprising one or more of these compounds, as well as methods of using the compounds in therapy, primarily in the treatment of diseases associated with C5a signaling activity.

Brief description of the drawings

Not applicable to

Detailed description of the invention

I. Abbreviations and Definitions

The terms "a", "an" or "the" as used herein include not only aspects of one member but also aspects of a plurality of members. For example, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a cell" also includes a plurality of such cells, and reference to "the agent" also includes one or more agents known to those skilled in the art, and so forth.

The terms "about" and "approximately" generally refer to a range of acceptable measurement errors due to the nature or accuracy of a given measurement method. Exemplary degrees of error are typically within 20%, preferably within 10%, and more preferably within 5% of a given value or range of values. Alternatively, particularly in biological systems, the terms "about" and "approximately" may denote a value within one order of magnitude, preferably within 5-fold, more preferably within 2-fold, of a given value. Unless otherwise indicated, the numerical values set forth herein are approximate, meaning that when not explicitly stated, the reference is to "about" or "approximately".

The term "pharmaceutically acceptable salt" is intended to include salts of the active compounds prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When the compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid in pure or suitable inert solvents. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids such as hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, monohydrogencarbonic acid, phosphoric acid, monohydrogenphosphoric acid, dihydrogenphosphoric acid, sulfuric acid, monohydrogensulfuric acid, hydrofluoric acid, or phosphorous acid, and the like, as well as those derived from relatively nontoxic organic acids such as acetic acid, propionic acid, isobutyric acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, methanesulfonic acid, and the like. Also included are Salts derived from amino acids, such as arginine Salts and the like, and Salts of organic acids, such as glucuronic acid or galacturonic acid and the like (see, e.g., Berge, SM, and the like, "drug Salts" (Pharmaceutical Salts), Journal of Pharmaceutical science, 1977, 66, 1-19).

The neutral form of the compound may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties (e.g. solubility in polar solvents), but for the purposes of the present invention, the salts are equivalent to the parent form of the compound.

Certain compounds of the invention may exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the present invention may exist in a variety of crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.

Certain compounds of the present invention have asymmetric carbon atoms (optical centers) or double bonds; racemates, diastereomers, geometric isomers, regioisomers and individual isomers (e.g., individual enantiomers) are all encompassed within the scope of the present invention.

As used herein, a line drawn to the inside of a ring refers to a connection at any available location on the ring.

II. Compound

In one aspect, the invention provides compounds having formula (I):

or a pharmaceutically acceptable salt thereof, wherein:

R1is selected from CH3,CH2D,CHD2Or CD3Or CD2OH;

R2Is selected from CH3,CH2D,CHD2,CD3,CH2OH, CDHOH or CD2OH;

R3Is H or D;

R4is H or D;

m is an integer of 0 to 6;

n is an integer of 0 to 3;

o is an integer from 0 to 4;

p is an integer from 0 to 9; and

q is an integer of 0 to 3;

wherein said compound has at least one deuterium atom in an amount of at least 80%.

In some embodiments, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein R is1Is selected from CH3Or CD3

In some embodiments, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein R is2Is selected from CH3,CD3,CH2OH or CD2OH。

In some embodiments, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein R is3Is D.

In some embodiments, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein R is3And R4Each is H.

In some embodiments, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein subscript n is 0, 1, or 2.

In some embodiments, compounds of formula (I) or a pharmaceutically acceptable salt thereof are provided wherein subscript o is 0 or 1.

In some embodiments, compounds of formula (I) or a pharmaceutically acceptable salt thereof are provided wherein subscript q is 0 or 1.

In some embodiments, compounds of formula (I) or a pharmaceutically acceptable salt thereof are provided wherein subscript p is 0, 1, 4, or 9.

In some embodiments, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein subscript m is 0.

In some embodiments, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein subscript m is 0; subscript n is 0, 1, or 2; subscript o is 0 or 1; subscript p is 0, 1, 4, or 9; subscript q is 0 or 1.

In some embodiments, there is provided a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein said compound has at least two deuterium atoms, said deuterium atoms being present in an amount of at least 80% for each deuterated position. In some embodiments, there is provided a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein the compound has at least three deuterium atoms present in an amount of at least 80% for each deuterated position. In some embodiments, there is provided a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein the compound has at least four deuterium atoms present in an amount of at least 80% for each deuterium position. In some embodiments, there is provided a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein the compound has at least five deuterium atoms, for each deuterium position, present in an amount of at least 80%.

The relative amount of deuterium at each deuterated position can vary. In some embodiments, for each deuterated position, the deuterium atoms are present in an amount of at least 80%, 85%, 90%, 95%, 97%, or 99%. In some embodiments, for each deuterated position, the deuterium atoms are present in an amount of at least 85%. In some embodiments, for each deuterated position, the deuterium atoms are present in an amount of at least 90%. In some embodiments, for each deuterated position, the deuterium atoms are present in an amount of at least 95%. In some embodiments, for each deuterated position, the deuterium atoms are present in an amount of at least 97%. In some embodiments, for each deuterated position, the deuterium atoms are present in an amount of at least 99%.

In addition to the compounds provided above, pharmaceutically acceptable salts of the compounds are also provided. In some embodiments, the pharmaceutically acceptable salt is selected from hydrochloric acid, carbonic acid, monohydrogencarbonic acid, phosphoric acid, monohydrogenphosphoric acid, dihydrogenphosphoric acid, acetic acid, propionic acid, isobutyric acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, tartaric acid, methanesulfonic acid, arginine salts, glucuronic acid or galacturonic acid (salts).

Pharmaceutical composition

In addition to the compounds provided above, compositions for modulating C5a activity in humans and animals typically comprise a pharmaceutical carrier or diluent.

As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. By "pharmaceutically acceptable" is meant that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.

Pharmaceutical compositions for administering the compounds of the present invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy and drug delivery. All methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. Generally, pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, bringing the product into the desired form. In the pharmaceutical composition, the active object compound is included in an amount sufficient to produce the desired effect during the course of the disease.

The pharmaceutical compositions comprising the active ingredient may be in a form suitable for oral use, for example, as described in U.S. patent application 2002-0012680, tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions and self-emulsifying agents, hard or soft capsules, syrups, elixirs, solutions, buccal patches, oral gels, chewing gums, chewable tablets, effervescent powders and effervescent tablets. Compositions for oral administration may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more ingredients selected from the group consisting of sweetening agents, flavoring agents, coloring agents, antioxidants and preserving agents in order to produce pharmaceutically elegant and palatable preparations. The tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of pharmaceutical tablets. These excipients may be, for example, inert diluents, such as cellulose, silicon dioxide, alumina, calcium carbonate, sodium carbonate, glucose, mannitol, sorbitol, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as corn starch or alginic acid; binding agents, for example PVP, cellulose, PEG, starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. Tablets may be uncoated or they may be enterically or otherwise coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be identified by U.S. Pat. nos. 4,256,108; 4,166,452 and 4,265,874 to form osmotic therapeutic tablets for controlled release.

Oral formulations may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil. In addition, non-water miscible ingredients such as oils may be used to prepare and stabilize the emulsion with surfactants such as monoglycerides, PEG esters, and the like.

Aqueous suspensions contain the active substance in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadiene-ethyleneoxycetanol, or condensation products of ethylene oxide with esters derived in part from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene glycol sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl or n-propyl p-hydroxybenzoate, one or more colouring agents, one or more flavouring agents and one or more sweetening agents, such as sucrose or saccharin.

Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.

Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are illustrated by those examples mentioned above. Other excipients, for example sweetening, flavouring and colouring agents, are also present.

The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these oils. Suitable emulsifying agents may be naturally-occurring gums (for example gum acacia or gum tragacanth), naturally-occurring phosphatides (for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate), and condensation products of the said partial esters with ethylene oxide (for example polyoxyethylene sorbitan monooleate). The emulsion may also contain sweetening and flavoring agents.

Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavouring and colouring agents. Oral solutions can be prepared by mixing with, for example, cyclodextrin, PEG, and a surfactant.

The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. Such suspensions may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Acceptable vehicles and solvents that may be used 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 injectables.

The compounds of the present invention may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols. In addition, the compounds may be administered by ophthalmic administration via a solution or ointment. Still further, transdermal delivery of the target compound may be accomplished by means of ionophoresis or the like. For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention may be used. As used herein, topical application is also intended to include the use of mouthwashes and gargles.

The compounds of the invention may also be combined with a carrier, which is a suitable polymer as a carrier for targeted drugs. Such polymers may include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropyl-methacrylamide-phenol, polyhydroxyethyl-asparagine-phenol or polyethylene oxide-polylysine substituted with palmitoyl residues. In addition, the compounds of the invention may be conjugated to a carrier, which is a class of biodegradable polymers useful for achieving controlled release of drugs, such as polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, poly-caprolactone, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphiphilic block copolymers of hydrogels. The polymers and semi-permeable polymer matrices can be formed into shaped articles, such as valves, stents, tubing, prostheses, and the like. In one embodiment of the invention, the compounds of the invention are coupled to a polymer or semipermeable polymer matrix used to make a stent or stent graft device.

The pharmaceutical compositions of the present disclosure may be formulated with one or more other therapeutic agents. The one or more additional therapeutic agents may include a corticosteroid, a steroid, an immunosuppressant, or a CD20 inhibitor. In some embodiments, the one or more additional therapeutic agents include obituzumab, rituximab, oclizumab, cyclophosphamide, prednisone, hydrocortisone acetate, cortisone acetate, teichok polyphenol pivalate, prednisolone, methylprednisolone, triamcinolone acetonide, pyruvic acid, triamcinolone acetonide, methasone, fluocinolone acetonide, betamethasone sodium phosphate, dexamethasone sodium phosphate, fluocortolone, hydrocortisone 17-valerate, halomirsone, amitriptonide, dipropionate propionate, beclomethasone, betamethasone-17-propionate, fluocinolone hexanoate, fluocinolone pivalate, fluorobutadiene acetate, hydrocortisone 17-butyrate, hydrocortisone 17-acetoacetate, hydrocortisone 17-butyrate, ciclesonide and pointenoformate, GB-0998, immunoglobulins, begrezumab, alfacaept (alefacept), leucocyte, gavacizumab, daclizumab, basiliximab, ilomumab, pemirolast (perminigene perplexid), serluzumab (sirukumab), tosiluzumab (tocilizumab), clarizumab (tocilizumab), clavam (clazakizumab), meperizumab (mepolizumab), fingolimod (fingolimod), panobinostat (panobit), triciribine (tricicibine), nilotinib (nilotinib), imatinib (imatinib), tofacitinib (tofacitinib), tofacitinib (mometab), mometamicin (moleptinib), morulitinib (mometamicin), peicitinib (peicitinib), bemisinficinib (PEG-bHb), tezomib (PEG-tezomib), oxycolizumab (otelixizumab), guanipimox (gusperimus), Bentuximab (brentuximab vedotin), Ponesimod (Ponesimod), KRP-203, FG-3019, Enlicarban (emricasan), adrenocorticotropic hormone, ibrutinib (ibrutinib), Venez (cinryze), cotitastat, methoxypolyethylene glycol-erythropoietin beta, belimumab (belimumab), bribimod (blisibimod), asecept (atacept), celecoxib (seliciclib), Nerilizumab (neihulizumab), Everolimus (Everolimus), sirolimus (sirolimus), Denil interleukin-toxin linker (denitifiukin diftimox), LMB-2, nacreolimus, tacrolimus, Votacrolimus (DLvosporine, Volvin-5), Volvox-vosporine (DLvosporine, Volvox-5, Volvorinacorinacin (R), abacavir, belicept, olmesartan medoxomil, sparsin tanuma (sparstanum), TXA-127, BIIB-023, alilimumab (alemtuzumab), pentostatin (pentostatin), rituzumab (itolizumab), palimin (palifermin), leflunomide, PRO-140, seniviroc (cenicriviroc), fostamatinib (stokes), anifrovatuzumab (aniflorumab), sivalimab (sifolimumab), BAX-069, BG-00011, los mamod (losbamimaod), QPI-1002, shigabs, TZ-101 maf-652, ritralin (reaprisxin), ladarisin (ladarixin), pti-9908, apsaintg-703), apirin (apsartori-T), tagasarin (APG-101, T-101 mah-652, griffonia-652, trastussin (reasonasolin (reasongarin), ladirisin (ladirisin), saratin-997, pti-703), tsunamine (aprine (aparin), DEX-M74, cardiotrophin-1, Diprelistat (tiprelestat), ASKP-1240, BMS-986004, HPH-116, KD-025, OPN-305, TOL-101, defibrillation peptide, Bomamide, Thymoglobulin, Laquinmod, Ramiprami Stem cells (remestcel-L), equine anti-thymocyte immunoglobulin, Stanpril (Stempecel), LIV-gamma, Ottago (Octagam) 10%, T2c-001, 99 mTc-methoxyisobutylisonitrile (sestamibi), Clariy (Clairyg), Prusobab (Prosorba), Primodopa (pomubide), Lasuxofenacin, framumab (formalumab), ATIR-101, BPX-501, ACP-01, ALLO-ASC-PADU, Irbesartan + Propoxadiol, ApoE-D-C-P-D, ApoE-C-D-M, ApoE-D-II, ApoE-C-D-M-D-52, ApoE-C-D-C-D, ApoE-C-D-S-C3, NOX-100, LT-1951, OMS721, ALN-CC5, ACH-4471, AMY-101, Acthar gel and CD4+ CD25+ regulatory T cells, MEDI7814, P32, P59, CCX354, CCX721, CCX9588, CCX140, CCX872, CCX598, CCX6239, CCX587, CCX624, CCX282, CCX025, CCX507, CCX430, CCX765, CCX758, CCX771, CCX662, CCX650, and combinations thereof. In certain selected embodiments, the additional agent is selected from obitumumab, rituximab, ocrelizumab, cyclophosphamide, prednisone, hydrocortisone acetate, cortisone acetate, teichoc polyphenol pivalate, prednisolone, methylprednisolone, triamcinolone acetonide, amcinolone acetonide, mometasone fluoroquinolone, fluocinolone, betamethasone sodium phosphate, dexamethasone sodium phosphate, fluocortolone, hydrocortisone 17-valerate, halomirsone, amiloride, dipropionate, beclomethasone, betamethasone ketone, betamethasone 17-propionate, fluorometholone hexanoate, fluorometholone pivalate, fluorobutadiene acetate, hydrocortisone 17-butyrate, hydrocortisone 17-acetoacetate, hydrocortisone 17-butyrate valerate, ciclesonide and prednisolone formate. Further discussion of combination therapies is contained in the "methods of use" section of the present application. In some embodiments, additional agents are formulated alone and/or in a "kit" format with the deuterated compounds provided herein.

Method of use

The compounds of the present invention may be used as agonists of the C5a receptor, preferably antagonists, partial agonists, inverse agonists, in a variety of contexts, both in vitro and in vivo. In one embodiment, the compounds of the invention are C5aR antagonists, which may be used to inhibit the binding of C5a receptor ligands (e.g., C5a) to the C5a receptor in vitro or in vivo. Typically, such methods include the steps of: the C5a receptor is contacted with a sufficient amount of one or more C5a receptor modulators provided herein in the presence of an aqueous solution of a C5a receptor ligand and under conditions suitable for ligand binding to the C5a receptor. The C5a receptor may be present in suspension (e.g., in an isolated membrane or cell preparation), in cultured or isolated cells, or in a tissue or organ.

Preferably, the amount of C5a receptor modulator contacted with the receptor should be sufficient to inhibit binding of C5a to the C5a receptor in vitro as measured using the radioligand binding assay, calcium mobilization assay or chemotaxis assay described herein.

In one embodiment of the invention, the C5a modulators of the invention are used to modulate, preferably to inhibit, the signal transduction activity of the C5a receptor, for example by contacting (in vitro or in vivo) one or more compounds of the invention with the C5a receptor under conditions suitable for binding of the modulator to the receptor. The receptor may be present in solution or suspension, in a cultured or isolated cell preparation, or in the body of a patient. Any modulation of signal transduction activity can be assessed by measuring the effect on calcium mobilization of calcium ions or by measuring cell chemotaxis mediated by the C5a receptor. Typically, an effective amount of a C5a modulator is an amount sufficient to modulate C5a receptor signaling activity in a calcium mobilization assay in vitro or to modulate C5a receptor-mediated cellular chemotaxis in a migration assay.

When the compounds of the invention are used to inhibit C5a receptor-mediated cellular chemotaxis, preferably leukocyte (e.g., neutrophil) chemotaxis, such methods comprise contacting leukocytes (particularly primate leukocytes, particularly human leukocytes) with one or more compounds of the invention in an in vitro chemotaxis assay. Preferably, the concentration is sufficient to inhibit chemotaxis of leukocytes in an in vitro chemotaxis assay, such that the level of chemotaxis observed in a control assay is significantly higher than that observed in an assay to which a compound of the invention has been added, as described above.

In another embodiment, the compounds of the present invention may also be used to treat a patient suffering from a disorder responsive to modulation of the C5a receptor. As used herein, the terms "treatment" or "method of treatment" encompass both disease-modifying treatment and symptomatic treatment, either of which may be prophylactic (i.e., to prevent, delay or lessen the severity of symptoms prior to the onset of symptoms) or may be therapeutic (i.e., to lessen the severity and/or duration of symptoms after the onset of symptoms). As used herein, a condition is considered "responsive to modulation of C5a receptors" if modulation of C5a receptor activity results in a reduction in inappropriate activity of C5a receptors. As used herein, the term "patient" includes primates (particularly humans), domesticated companion animals (e.g., dogs, cats, horses, etc.) and livestock (e.g., cattle, pigs, sheep, etc.) utilizing dosages as described herein.

Diseases that can be treated by C5a modulation:

autoimmune diseases-for example, rheumatoid arthritis, systemic lupus erythematosus, guillain-barre syndrome, pancreatitis, lupus nephritis, lupus glomerulonephritis, psoriasis, crohn's disease, vasculitis, irritable bowel syndrome, dermatomyositis, multiple sclerosis, bronchitis, sedimentary disease, pemphigus, pemphigoid, scleroderma, myasthenia gravis, autoimmune hemolysis and thrombocytopenia, goodpasture's syndrome (and associated glomerulonephritis and pulmonary hemorrhage), C3 glomerulopathy, C3 glomerulonephritis, membranoproliferative glomerulonephritis, kawasaki disease, IGs nephropathy, immune vasculitis, tissue transplant rejection, graft-versus-host disease, transplant organ hyperacute rejection; and so on.

Inflammatory diseases and related disorders-for example, neutropenia, sepsis, septic shock, alzheimer's disease, multiple sclerosis, neutrophilia, stroke, Inflammatory Bowel Disease (IBD), inflammation associated with severe burns, lung injury and ischemia reperfusion injury, osteoarthritis and acute (adult) respiratory distress syndrome (ARDS), chronic lung obstructive disease (COPD), Systemic Inflammatory Response Syndrome (SIRS), atopic dermatitis, psoriasis, chronic urticaria and multi-organ dysfunction syndrome (MODS), hemolytic uremic syndrome, hidradenitis suppurativa and atypical hemolytic uremic syndrome (aHUS). Also included are pathological sequelae associated with: insulin-dependent diabetes mellitus (including diabetic retinopathy), lupus nephropathy, Heiman nephritis, membranous nephritis and other forms of glomerulonephritis, contact sensitivity responses and inflammation caused by contact of blood with artificial surfaces that can cause complement activation, such as occurs during extracorporeal blood circulation (e.g., during hemodialysis or by heart-lung machines, e.g., associated with vascular surgery such as coronary artery bypass grafting or heart valve replacement), or in association with contact with other artificial blood vessels or container surfaces (e.g., ventricular assist devices, artificial heart machines, blood transfusion vessels, blood storage bags, plasma replacement, platelet replacement, etc.). Also included are diseases associated with ischemia/reperfusion injury, for example, diseases caused by transplantation including solid organ transplantation, and syndromes such as ischemia reperfusion injury, ischemic colitis and cardiac ischemia. The compounds of the invention are also useful in the treatment of age-related macular degeneration (Hageman et al, P.N.A.S.102:7227-7232,2005)).

Cardiovascular and cerebrovascular diseasesFor example, myocardial infarction, coronary thrombosis, vascular occlusion, postoperative vascular reocclusion, atherosclerosis, traumatic central nervous system injury and ischemic heart disease. In one embodiment, an effective amount of a compound of the invention may be administered to a patient at risk of myocardial infarction or thrombosis (i.e., having one or more recognized risk factors for myocardial infarction or thrombosis, e.g., but not limited toRestricted to obesity, smoking, hypertension, hypercholesterolemia, past or genetic history of myocardial infarction or thrombosis) to reduce the risk of myocardial infarction or thrombosis.

Neoplastic disease or disorder-for example, melanoma, lung cancer, lymphoma, sarcoma, carcinoma, fibrosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, angiosarcoma, lymphangiosarcoma, synovioma, mesothelioma, meningioma, leukemia, lymphoma, leukemic squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, papillary carcinoma, cystadenocarcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatocellular carcinoma, transitional cell carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, wilms' tumor, polymorphic adenoma, hepatocellular papilloma, renal tubular adenoma, bladder adenoma, papilloma, adenoma, leiomyoma, rhabdomyoma, hemangioma, lymphangioma, osteoma, chondroma, lipoma and fibroma.

VasculitisVascular diseases are characterized by inflammation of the blood vessels. The infiltration of leukocytes leads to the destruction of the vessel wall, while the complement pathway is thought to play a major role in initiating leukocyte migration and eventual damage manifested at sites of inflammation (vasculitis, second edition, edited by Ball and Bridges, oxford university press, pages 47-53, 2008). The compounds provided by the invention can be used for treating leucocytic vasculitis, anti-neutrophil cytoplasmic antibody (ANCA) associated vasculitis, immune vasculitis Wegener granulomatosis, microscopic polyangiitis, allergic granulomatosis vasculitis, Henoch-Schonlein purpura nephritis, nodular multiple nephritis, Rapidly Progressive Glomerulonephritis (RPGN), cryoglobulinemia, Giant Cell Arteritis (GCA), Behcet's disease and TAK.

HIV infection and AIDSModulators of the C5a receptor provided herein are useful for inhibiting HIV infection, delaying the progression of AIDS or reducing the severity of symptoms or HIV infection and AIDS.

Neurodegenerative and related diseasesIn other aspects, the C5a antagonists provided herein are useful for treating alzheimer's disease, multiple sclerosis and cognitive function associated with cardiopulmonary bypass surgery and related proceduresAnd (4) descending.

In one embodiment of the invention, the compounds of the invention may be used for the treatment of a disease selected from the group consisting of: sepsis (and related diseases), COPD, rheumatoid arthritis, lupus nephritis or multiple sclerosis.

The methods of treatment provided herein generally comprise administering to a patient an effective amount of one or more compounds provided herein. Suitable patients include those patients suffering from or susceptible to (i.e., prophylactically treated) a condition or disease identified herein. Typical patients for treatment as described herein include mammals, particularly primates, and especially humans. Other suitable patients include domesticated companion animals such as dogs, cats, horses, etc., or livestock such as cattle, pigs, sheep, etc.

In general, the methods of treatment provided herein comprise administering to a patient an effective amount of one or more compounds provided herein. In a preferred embodiment, the compounds of the invention are preferably administered to a patient (e.g., a human) orally or topically. An effective amount may be an amount sufficient to modulate the activity of the C5a receptor and/or to reduce or alleviate symptoms present in a patient. Preferably, the amount administered is sufficient to produce a sufficiently high plasma concentration of the compound (or its active metabolite if the compound is a prodrug) to detectably inhibit chemotaxis of leukocytes (neutrophils) in vitro. The treatment regimen may vary depending upon the compound used and the particular disease being treated; for the treatment of most diseases, it is preferred that the administration is carried out 4 times per day or less frequently. In general, a dosing regimen of 2 times per day is more preferred, and once daily dosing is particularly preferred. It will be understood, however, that the specific dose level and treatment regimen for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration and the like, the route of administration, the rate of excretion, drug combination (i.e., other drugs administered to the patient) and the severity of the particular disease undergoing therapy, and the judgment of the prescribing physician. Generally, it is preferred to use the minimum dose sufficient to provide effective treatment. The effectiveness of a patient's treatment can generally be monitored using medical or veterinary standards appropriate to the disease being treated or prevented.

Dosage levels of about 0.1mg to about 140mg per kilogram of body weight per day are useful for treating or preventing diseases related to pathogenic C5a activity (about 0.5mg to about 7g per person per day). The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Dosage unit forms will typically contain from about 1mg to about 500mg of the active ingredient. For compounds administered orally, transdermally, intravenously or subcutaneously, it is preferred to administer sufficient amount of the compound to achieve a compound serum concentration of 5ng (nanogram)/mL to 10 μ g (microgram)/mL of serum, more preferred to administer sufficient compound to achieve a compound serum concentration of 20ng to 1 μ g/mL of serum, and most preferred that sufficient compound be administered to achieve a compound serum concentration of 50ng/mL to 200ng/mL of serum. For direct injection into the synovium (for the treatment of arthritis), sufficient compound should be administered to achieve a local concentration of about 1 micromolar.

The frequency of administration may also vary depending on the compound used and the particular disease being treated. However, for the treatment of most diseases, a dosage regimen of 4 times per day, 3 times per day or less is preferred, with a dosage regimen of once per day or 2 times per day being particularly preferred. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination (i.e., other drugs administered to the patient), the severity of the particular disease being treated, and other factors, including the judgment of the prescribing physician.

Combination therapy

The compounds disclosed herein may be used in combination with one or more other therapeutic agents for treating, preventing, inhibiting, or ameliorating a disease or condition in which the compounds and compositions of the present invention are useful. The compound or composition of the present invention and such one or more additional therapeutic agents may be administered by the route and amount in which they are commonly used, either simultaneously or sequentially. When a compound or composition of the present invention is used concurrently with one or more other drugs, a pharmaceutical composition comprising such other drugs in addition to the compound or composition of the present invention is preferred. Accordingly, the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients or therapeutic agents in addition to the compounds or compositions of the present invention.

Examples of one or more additional therapeutic agents that may be combined with a compound or composition of the invention, which may be administered alone or in the same pharmaceutical composition, include, but are not limited to, corticosteroids, steroids, immunosuppressive agents, immunoglobulin G agonists, dipeptidyl peptidase IV inhibitors, lymphocyte function antigen 3 receptor antagonists, interleukin 2 ligands, interleukin 1 β ligand inhibitors, IL-2 receptor α subunit inhibitors, HGF gene stimulators, IL-6 antagonists, IL-5 antagonists, Alpha 1 antitrypsin stimulators, cannabinoid receptor antagonists, histone deacetylase inhibitors, AKT protein kinase inhibitors, CD20 inhibitors, Abl tyrosine kinase inhibitors, TNF α ligand inhibitors, hemoglobin modulators, TNF antagonists, proteasome inhibitors, CD3 modulators, Hsp 70 family inhibitors, immunoglobulin differentiation agonists, CD30 antagonists, tubulin antagonists, JAK-1-phosphate receptor 1 agonists, Symphotein growth factor ligand inhibitors, caspase inhibitors, BtBtK 1 kinase inhibitors, BtK receptor kinase inhibitors, BtBtK receptor kinase inhibitors, CDK receptor kinaseInhibitors of kinases, interferon type I receptor antagonists, inhibitors of interferon α ligands, inhibitors of macrophage migration inhibitory factor, integrin α body inhibitors-6 antagonists, cysteine protease stimulators, p38MAP kinase inhibitors, TP53 gene inhibitors, shiga-like toxin I inhibitors, fucosyltransferase 6 stimulators, interleukin 22 ligands, IRS1 gene inhibitors, protein kinase C stimulators, protein kinase C α inhibitors, CD74 antagonists, immunoglobulin gamma Fc receptor IIB antagonists, T cell antigen CD7 inhibitors, CD95 antagonists, NMO-mannosamine kinase stimulators, cardiotrophin 1 ligands, leukocyte elastase inhibitors, CD40 ligand receptor antagonists, CD40 ligand modulators, IL-17 antagonists, TLR-2 antagonists, mannan-binding lectin serine protease 2(MASP-2) inhibitors, factor B inhibitors, factor D inhibitors, C3aR modulators, C5aR2 modulators, T cell receptor antagonists, 82receptors, VLR 56, VLR 3927 ligand inhibitors, VLR 3 ligand inhibitors, VLR-ketoprofen receptor agonists, hydrocortisone receptor antagonists, such as dexamethasone acetate, hydrocortisone receptor antagonists, receptor antagonist for example, hydrocortisone receptor tyrosine receptor antagonists, hydrocortisone receptor tyrosine receptor antagonists, hydrocortisone receptor antagonists, such as prednisone, hydrocortisone (e, hydrocortisone receptor antagonist, hydrocortisone receptor antagonist, hydrocortisone receptor antagonist (e, hydrocortisone receptor antagonist, hydrocortisone including, hydrocortisone receptor antagonistBetasol (including clobetasol 17-butyrate); (c) immunosuppressive agents, such as cyclosporine (cyclosporin a,) Tacrolimus (FK-506,) Rapamycin (sirolimus,

Figure BDA0002474403990000233

) And other FK-506 type immunosuppressants, as well as mycophenolic acid, such as mycophenolate mofetil (CellCept 8); (d) antihistamines (H1-histamine antagonists) such as brompheniramine, chlorpheniramine, dexchlorpheniramine, triphenidine, clemastine, diphenhydramine, dibenzopyran, triphenylamine, hydroxyzine, methylpyridazine, promethazine, trimetperazine, azatadine, cyproheptadine, anthrazoline, phenamine pyrrolamide, astemizole, terfenadine, loratadine, cetirizine, fexofenadine, norethoxyloratadine, and the like; (e) non-steroidal anti-asthmatics (e.g., terbutaline, metaproterenol, fenoterol, isoephedrine, albuterol, bitoltiprole and piriboteron), theophylline, cromolin sodium, atropine, ipratropium bromide, leukotriene antagonists (e.g., zafirlukast, montelukast, pruilast, pulilast, SKB-106,203), leukotriene biosynthesis inhibitors (zileuton, BAY-1005); (f) non-steroidal anti-inflammatory drugs (NSAIDs), such as propionic acid derivatives (e.g., amoprofen, benoxaprofen, bucloxic acid, carprofen, fenbufen, fenoprofen, fluprofen, flurbiprofen, ibuprofen, indoprofen, ketoprofen, nilofen, aposen, valofen, suprofen, tiaprofenic acid, and tixaprofen), acetic acid derivatives (e.g., indomethacin, acemetacin, alclofenac, clindamic acid, diclofenac, fenclofenac, fentanyl, furfenac, ibufenac, isoxepac, oxpinac (oxpinac), sulindac, tiopinate, zidometacin, and sodium zomepirac), fenamic acid derivatives (e.g., flufenamic acid, meclofenamic acid, mefenamic acid)Niflumic and tolfenamic acids), biphenylcarboxylic acid derivatives (e.g., diflunisal and flufenisal), oxicams (e.g., isoxicam, piroxicam, sudoxicam and tenoxicam), salicylates (e.g., acetylsalicylic acid and sulfasalazine) and pyrazolones (e.g., apalone, benperidone, haloperidol, moxibutone, oxybenzone and phenylbutazone); (g) cyclooxygenase 2(COX-2) inhibitors, e.g. celecoxibAnd rofecoxib(h) Phosphodiesterase type IV inhibitors (PDE IV); (i) gold compounds, such as auranofin and chlorthioglucose, (j) etanercept (Enbre10), (k) cyclophosphamide, (l) antibody therapeutics, such as Moluomab (OKT3), daclizumabBasiliximabAnd infliximab

Figure BDA0002474403990000245

(m) antibody therapeutics targeting CD20, such as abicyclotuzumab, rituximab, or ocrelizumab; (n) chemotherapeutic agents such as anthracyclines (e.g., daunorubicin (daunomycin; erythromycin), doxorubicin, epirubicin, idarubicin, and valirubicin), mitoxantrone, and pioxanone; platinum-based drugs (e.g., "cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, nidaplatin, triplatin, and lipoplatin); tamoxifen and its metabolites, such as 4-hydroxy tamoxifen (amoxifen) and N-desmethyl-4-hydroxy tamoxifen (entorfin); taxanes such as paclitaxel (taxol) and docetaxel; alkylating agents (nitrogen mustards, such as methoxyethylamine (HN2), cyclophosphamide, ifosfamide, melphalan (L-myolysin) and chlorambucil); ethyleneimine and methyl melamine (e.g. hexamethylmelamine, thioteraba)Alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine (BCNU), lomustine (CCNLJ), semustine (methyl-CCN-U) and streptozotocin (streptozotocin) and triazenes, such as dacarbazine (DTIC; dimethyltriazenimidazole-carboxamide)); antimetabolites (e.g. folic acid analogues such as methotrexate, pyrimidine analogues such as fluorouracil (5-fluorouracil; 5-FU), floxuridine (fluorodeoxyuridine; FUdR) and cytarabine (cytosine arabinoside), as well as purine analogues and related drug inhibitors such as mercaptopurine (6-mercaptopurine; 6-MP), thioguanine (6-thioguanine; 6-TG) and pentostatin (2-deoxychrysanthomycin), chemokine receptors, in particular CXCR1, CXCR2, CXCR3, CXCR4, CXCR7, CCR1, CCR2, CCR3, CCR4, CCR5, CCR7, CCR9, CX3CR1 and other antagonists of CXCR 6.

In some embodiments, the additional therapeutic agent is selected from CCX354, CCX721, CCX9588, CCX140, CCX872, CCX598, CCX6239, CCX587, CCX624, CCX282, CCX025, CCX507, CCX430, CCX765, CCX758, CCX771, CCX662, or CCX650, and combinations thereof.

The disease or condition being treated will identify the other therapeutic agent or agents most suitable for administration in combination with the compounds of the present invention-such identification can be made by one skilled in the art.

The weight ratio of the compound of the invention to the second active ingredient may vary and depends on the effective dose of each ingredient. Generally, an effective dose of each ingredient will be used. Thus, for example, when a compound of the invention is combined with an NSAID, the weight ratio of the compound of the invention to NSAID is typically from about 1000: 1 to about 1: 1000, preferably from about 200: 1 to about 1: 200. combinations of the compounds of the invention and other active ingredients will generally also be within the above ranges, but in each case an effective dose of each active ingredient should be used.

Non-medical applications

In another aspect of the invention, the compounds of the invention may be used in a variety of non-pharmaceutical in vitro and in vivo applications. For example, the compounds of the invention may be labeled and used as probes (cell preparations or tissue slice samples) for detecting and localizing the C5a receptor. The compounds of the invention may also be used as positive controls in assays for C5a receptor activity, i.e., as standards to determine the ability of a candidate agent to bind to the C5a receptor, or as radiotracers for Positron Emission Tomography (PET) imaging or for Single Photon Emission Computed Tomography (SPECT). Such methods can be used to characterize the C5a receptor in a living subject. For example, the C5a receptor modulator may be labeled (e.g., radiolabeled with a radionuclide such as tritium) using any of a variety of well-known techniques, and incubated with the sample for an appropriate incubation time (the time course of binding is first determined). After incubation, unbound compounds are removed (e.g., by washing) and bound compounds are detected using any method appropriate to the label used (e.g., methods of autoradiography or scintillation counting of radiolabeled compounds; luminophores and fluorophores can be detected using spectroscopic methods). As a control, a matched sample containing a labeled compound and more (e.g., 10-fold greater) unlabeled compound can be treated in the same manner. A greater amount of detectable label remains in the test sample compared to the control, indicating the presence of C5a receptor in the sample. Detection assays, including autoradiography (receptor localization) of the C5a receptor in cultured cell or tissue samples, can be performed as described by Kuhar in Current protocols of pharmacology (1998) John Wiley & Sons, New York, sections 8.1.1 through 8.1.9.

The compounds provided herein can also be used in a variety of well-known cell separation methods. For example, a modulator may be attached to the inner surface of a tissue culture plate or other support, serving as an affinity ligand, for in vitro immobilization and thus isolation of the C5a receptor (e.g., to isolate receptor-expressing cells). In a preferred application, a modulator linked to a fluorescent marker, such as fluorescein, is contacted with the cells and then analyzed (or isolated) by Fluorescence Activated Cell Sorting (FACS).

V. examples

The following examples are provided to illustrate, but not to limit, the claimed invention.

The reagents and solvents used below may be selected fromCommercially available, for example, from Older Rich chemical company (Milwaukee, Wis., USA).1H-NMR spectra were recorded on a Varian Mercury 400MHz NMR spectrometer. Significant peaks were provided relative to TMS and are listed in the following order: multiplicity (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet) and number of protons. Mass spectrometry results are reported as the ratio of mass to charge, followed by the relative abundance of each ion (in parentheses). In the examples, the individual M/e values of the M + H (or, as noted above, M-H) ions containing the most common atomic isotopes are reported. In all cases, the isotope patterns fit the expected formula. Electrospray ionization (ESI) mass spectrometry was performed on a Hewlett-Packard MSD electrospray mass spectrometer using HP1100HPLC for sample delivery. Typically, the analyte is dissolved in 0.1mg/mL of methanol and 1 microliter of this is injected with the delivery solvent into a mass spectrometer that scans from 100 to 1500 daltons. All compounds can be analyzed in the positive ESI mode using acetonitrile/water and 1% formic acid as the transport solvent. Using 2mM NH in acetonitrile/water4OAc as a delivery system, the compounds provided below can also be analyzed in the negative ESI mode.

The following abbreviations are used in the examples and throughout the specification: mL, mL; millimole; ethanol, ethanol; EtOAc, ethyl acetate; EtON, sodium ethoxide; THF, tetrahydrofuran; TLC, thin layer chromatography; MeOH, methanol.

Compounds within the scope of the present invention may be synthesized as described below using a variety of reactions known to those skilled in the art. Those skilled in the art will also recognize that alternative methods may be employed to synthesize the subject compounds of the present invention, and that the methods described herein are not exhaustive, but do provide a widely applicable and practical route to the subject compounds. .

Certain molecules claimed in this patent may exist in different enantiomeric and diastereomeric forms, and all such variations of these compounds are claimed.

The detailed description herein of the experimental procedures used to synthesize key compounds is described in terms of the physical data that the molecules recognize as well as the structural depictions associated with them.

Those skilled in the art will also recognize that acids and bases are often used in standard processing procedures in organic chemistry. Salts of the parent compound are sometimes generated during the experimental procedures described in this patent if they possess the requisite intrinsic acidity or basicity.

Some starting materials and intermediates were prepared according to the methods provided in WO2010/075257 and US20160090357, which are incorporated herein by reference.

Intermediate:

2-fluoro-6- (methyl-d)3) Synthesis of-benzoyl chloride

Step a-addition of 2-amino-2-methyl-1-propanol (10.1g, 113.3mmol) to CH at 0 ℃ under nitrogen2Cl2(75mL) to a solution of 2, 6-difluorobenzoyl chloride (10.0g, 56.6mmol) in CH was added dropwise2Cl2(50mL) of the solution. The reaction mixture was then warmed to room temperature and stirred for 16 hours. Reaction mixture with H2Diluting with O, and adding CH to water phase2Cl2Extracting with Na2SO4Dried and concentrated. The residue was purified by trituration with hexanes to give 2, 6-difluoro-N- (1-hydroxy-2-methylpropan-2-yl) -benzamide as a white solid.

Step b: to a stirred solution of 2, 6-difluoro-N- (1-hydroxy-2-methylpropan-2-yl) benzamide (6.0g, 26.2mmol) in CH at 0 deg.C2Cl2(30mL) to the solution was added SOCl2(4.9g, 41.9 mmol). The reaction mixture was allowed to warm to room temperature and stirred for 6 hours. The reaction mixture was concentrated and the residue was triturated with diethyl ether. Dissolving the filter cake in H2O, and basified with 6n naoh aqueous solution. CH for the mixture2Cl2And (4) extracting. The organic phases were combined and passed over Na2SO4Drying and concentration gave 2- (2, 6-difluorophenyl) -4, 5-dihydro-4, 4-dimethyloxazole as a white solid.

Step c: to an ice bathTo a cooled solution of 2- (2, 6-difluorophenyl) -4, 5-dihydro-4, 4-dimethyloxazole (1.5g, 7.1mmol) in anhydrous THF (30mL) was added dropwise 1MCD3MgI solution in THF (20.6mL, 20.6 mmol). The reaction mixture was stirred at 0 ℃ for 1h, then the ice bath was removed and the reaction mixture was stirred at room temperature for 24 h. After the reaction was complete, the mixture was washed with saturated NH4Cl quenched and extracted with EtOAc. H for organic layer2O, washed with brine and Na2SO4And (5) drying. The solution was filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 50% EtOAc in hexanes) to give 2- (2-fluoro-6- (methyl-d) as a white solid3) -phenyl) -4, 4-dimethyl-4, 5-dihydrooxazole.

Step d: stirring 2- (2-fluoro-6- (methyl-d)3) -phenyl) -4, 4-dimethyl-4, 5-dihydrooxazole (700mg, 3.31mmol) in acetonitrile (5mL) and iodomethane (1.41g, 9.93mmol) were added. The reaction mixture was heated to reflux for 6 hours. The reaction mixture was stirred and allowed to cool to room temperature overnight. The reaction mixture was concentrated and the residue was triturated with ether and filtered. The solid was dissolved in equal parts of 20% NaOH and methanol (5mL) and heated at reflux for 6 h. The reaction mixture was cooled to room temperature, the organic solvent was removed in vacuo, and the aqueous phase was extracted with EtOAc to remove by-products. The aqueous layer was then acidified with 2N HCl (pH 1) and extracted with EtOAc. The organic layer was washed with brine, over Na2SO4Drying, filtering and concentrating to obtain 2-fluoro-6- (methyl-d)3) -benzoic acid.1H NMR(400MHz,CDCl3)7.25-7.37(m,1H),7.03(dd,J=0.8,7.4Hz,1H),6.96-7.02(m,1H)MS:(ES)C8H4FD3O2[M+H]+The calculated m/z was 158.3, found 158.3.

Step e: at 0 deg.C to 2-fluoro-6- (methyl-d)3) A50 mL round bottom flask of benzoic acid (0.45g, 2.84mmol) and anhydrous dichloromethane (15mL) was charged with oxalyl chloride (0.72g, 5.69mmol added dropwise) for 5 min. The reaction mixture was stirred at room temperature for 16 hours. After completion of the reaction, the solvent was removed in vacuo to give 2-fluoro-6-methylbenzoyl chloride, which was used directly in the next step without further purification.

2-fluoro-6-methylbenzoyl chloride-4-d

Step a: 4-bromo-2-fluoro-6-methylbenzoic acid (0.5g, 2.14mmol) and K were reacted at room temperature2CO3(0.35g, 2.57mmol) in D2To the mixture in O (8mL) was added 10% Pd-C (150mg, deuterium atmosphere, in CD3Pre-wash in OD). The resulting mixture was stirred under deuterium atmosphere (balloon pressure) overnight. The reaction mixture was filtered through celite, first with Et2The aqueous filtrate was extracted to remove by-products and then acidified to pH 1 with 2N HCl. The aqueous layer was extracted with EtOAc, washed with brine, over Na2SO4Dried, filtered and concentrated. The residue was dried under vacuum for 2 hours to give 2-fluoro-6- (methylbenzoic acid-4-d) acid.1H NMR(400MHz,CDCl3)7.04(br,s,1H),6.97(d,J=9.8Hz,1H),2.4(s,3H)。MS:(ES)C8H6DFO2[M+H]+The calculated m/z was 156.1, found 156.1

Step b: to a 50mL round bottom flask charged with 2-fluoro-6- (methylbenzoic acid-4-d) acid (0.27g, 1.73mmol) and anhydrous dichloromethane (6mL) was added oxalyl chloride (0.54g, 4.32mmol) dropwise at 0 deg.C for 5 min. The reaction mixture was stirred at room temperature for 16 hours. After completion of the reaction, the solvent was removed under reduced pressure and the residue was dried under vacuum for 20 minutes to give 2-fluoro-6-methylbenzoyl chloride-4-d, which was used directly in example 3 without further purification.

Example 1: synthesis of (2R, 3S) -2- (4- (cyclopentylamino) phenyl) -1- (2-fluoro-6- (methyl-d)3) Benzoyl) -N- (4-methyl-3- (trifluoromethyl) phenyl) piperidine-3-carboxamide

Step a: to tert-butyl (4- ((2R, 3S) -3- ((4-methyl-3- (trifluoromethyl) phenyl) carbamoyl) piperidin-2-yl) phenyl) carbamate at-10 deg.C(0.6g, 1.25mmol) and N, N-diisopropylethylamine (0.32g, 2.51mmol) in CH2Cl2(10mL) to the stirred solution was added 2-fluoro-6- (methyl-d)3) Benzoyl chloride (220mg, 1.25mmol) and the mixture was stirred vigorously at room temperature for 2 h. After completion, the reaction mixture was washed with H2Dilution with O and CH2Cl2And (4) extracting. The organic layers were combined and washed with Na2SO4Dried, filtered and concentrated. The residue was purified by silica gel column chromatography (10 to 60% EtOAc in hexanes) to give tert-butyl- (4- (((2R, 3S) -1- (2-fluoro-6- (methyl-d)3) Benzoyl) -3- ((4-methyl-3- (trifluoromethyl) phenyl) carbamoyl) piperidin-2-yl) phenyl) carbamate. MS (ES) C33H32D3F4N3O4[M+H]+The calculated m/z is 617.3, found 617.3.

Step b: to tert-butyl- (4- ((2R, 3S) -1- (2-fluoro-6- (methyl-d)3) Benzoyl) -3- ((4-methyl-3- (trifluoromethyl) phenyl) carbamoyl) piperidin-2-yl) phenyl) carbamate (0.40g, 0.64mmol) in CH2Cl2(8mL) to a stirred solution was added a 4N HCl in dioxane (2.5mL, 2.58 mmol). The reaction mixture was stirred at room temperature for 3 hours. After completion, the reaction mixture was treated with CH2Cl2And (6) diluting. The organic layer was washed with saturated NaHCO3Washing with Na2SO4Dried, filtered and concentrated. The residue was purified by flash chromatography on silica gel (0-100% EtOAc in hexane) to give (2R, 3S) -2- (4-aminophenyl) -1- (2-fluoro-6-methyl-d3) -benzoyl) -N- (4-methyl-3- (trifluoromethyl) -phenyl) -piperidine-3-carboxamide. MS (ES) is C28H24D3F4N3O2[M+H]+The calculated m/z was 517.2, found 517.2.

Step c: (2R, 3S) -2- (4-aminophenyl) -1- (2-fluoro-6-methylbenzoyl) -N- (4-methyl-3- (trifluoromethyl) -phenyl) piperidine-3-carboxamide (150mg, 0.29mmol), cyclopentanone (73mg, 0.87mmol), NaBH (OAc) at room temperature3(183mg, 0.72mmol) and HOAc (17mg, 0.29mmol) in CH2Cl2(4mL) inStirred for 6 hours. The mixture was then washed with saturated NaHCO3Alkalizing with CH2Cl2And (4) extracting. The combined organic layers were washed with brine, over Na2SO4Dried, filtered and concentrated. The residue was purified by flash chromatography on silica gel (0 to 60% EtOAc in hexanes) to give (2R, 3S) -2- (4- (cyclopentylamino) phenyl) -1- (2-fluoro-6- (methyl-d)3) -benzoyl) -N- (4-methyl-3- (trifluoromethyl) phenyl) piperidine-3-carboxamide. MS (ES) C33H32D3F4N3O2[M+H]+The calculated m/z is 585.3, found 585.2.

46页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:肠胃外制剂及其用途

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!