Saturday 14 March 2015

AMG 925 from Amgen

AMG 925

AMG 925
AMG 925
1401033-86-0
2-Hydroxy-1-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone
2-Hydroxy-l-(2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido [4′,3 ‘ :4,5] pyrrolo [2,3-d] pyrimidin-2-yl)amino)-7,8-dihydro-l ,6- naphthyridin-6(5H)-yl)ethanone
2-Hydroxy-1-(2-((9-((1R,4R)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone (AMG 925)
FLT3/CDK4 inhibitor,potent and selective
AMG 925 is a dual kinase inhibitor of FLT3 and CDK4 with IC50 value of 1 nM and 3 nM, respectively
C26H29N7O2., 471.55

BY
SECTION 1
STEP A
STEP B
Figure imgf000125_0003
STEP C
Figure imgf000126_0001
STEP D
Figure imgf000127_0001
9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-amine
COUPLER 1
Figure imgf000128_0001
tert-butyl 2-chloro-7,8-dihydro-l,6-naphthyridine-6(5H)-carboxylate
STEP E
Figure imgf000128_0002
tert-butyl 2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido [4′,3 ‘ :4,5] pyrrolo [2,3-d] pyrimidin-2-yl)amino)-7,8-dihydro-l ,6- naphthyridine-6(5H)-carboxylate
STEP F
Figure imgf000129_0001COMPD 1
9-((l r,4r)-4-methylcyclohexyl)-N-(5,6,7,8-tetrahydro- l,6-naphthyridin-2-yl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-amine (1)
SECTION B
COUPLER2
Figure imgf000134_0002
2,5-dioxopyrrolidin-l-yl 2-acetoxyacetate
STEP G
Figure imgf000134_0003
2-(2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido [4′,3 ‘ :4,5] pyrrolo [2,3-d] pyrimidin-2-yl)amino)-7,8-dihydro-l ,6- naphthyridin-6(5H)-yl)-2-oxoethyl acetate
STEP H
Figure imgf000135_0001AMG 925
 STEP I
AMG 925 is a potent, selective, and orally available FLT3/CDK4 dual inhibitor. It also inhibits CDK6 potently in kinase assay. In acute myeloid leukemia (AML) cell lines MOLM13 and Mv4-11, AMG 925 inhibits cell growth (IC50 values of 19nM and 18nM, respectively) through inhibiting P-FLT3 and P-STAT5 and inducing apoptosis. FLT3 mutants cause resistance to the current FLT3 inhibitors. AMG 925 is reported to inhibit cell growth in AML cells with FLT3 mutants FLT3-D835Y and FLT3-D835V. In AML tumor –bearing mice, administration of AMG 925 shows inhibition of P-STAT5 and P-RB as well as cell growth both in subcutaneous MOLM13 xenograft tumor model and systemic MOLM13-Luc xenograft tumor model. AMG 925 is also reported to have antitumor activity in a dose-dependent manner in theRB-positive Colo205 colon adenocarcinoma xenograft model

AMGEN
cute myeloid leukemia (AML) represents a significant unmet medical need. It is a hematological malignancy characterized by a block in differentiation and aberrant proliferation of the myeloid lineage of hematopoietic progenitor cells. There are approximately 13,000 new cases and 9,000 deaths per year in the United States. The survival rate is 25-70% in patients younger than 60 years and 5-15% in older patients, with worse outcomes in patients with poor risk cytogenetics. Current standard of care treatment is daunorubicin and cytarabine chemotherapy with induction and consolidation phases. Bone marrow stem cell transplant is also used for treating AML in younger patients.
Cyclin-dependent kinases (CDKs) are a family of serine/ threonine protein kinases playing important cellular functions. The cyclins are the regulatory subunits that activate the catalytic CDKs. CDKl/Cyclin B 1 , CDK2/Cyclin A, CDK2/Cyclin E, CDK4/Cyclin D, CDK6/Cyclin D are critical regulators of cell cycle progression. CDKs also regulate transcription, DNA repair, differentiation, senescence and apoptosis (Morgan, D. O., Annu. Rev. Cell. Dev. Biol., 13:261-291 (1997)).
Small molecule inhibitors of CDKs have been developed to treat cancer
(de Career, G. et al., Curr. Med. Chem., 14:969-85 (2007)). A large amount of genetic evidence supports that CDKs, their substrates or regulators have been shown to be associated with many human cancers (Malumbres, M. et al, Nature Rev. Cancer, 1 :222- 231 (2001)). Endogenous protein inhibitors of CDKs including p 16, p21 and p27 inhibit CDK activity and their overexpression results in cell cycle arrest and inhibition of tumor growth in preclinical models (Kamb, A., Curr. Top. Microbiolo. Immunol., 227: 139- 148 (1998)).
Small molecule inhibitors of CDKs may also be used to treat variety of other diseases that result from aberrant cell proliferation, including cardiovascular disorders, renal diseases, certain infectious diseases and autoimmune diseases. Cell proliferation pathways including genes involved in the cell cycle Gl and S phase checkpoint (p53, pRb, pi 5, pi 6, and Cyclins A, D, E, CDK 2 and CDK4) have been associated with plaque progression, stenosis and restenosis after angioplasty. Over- expression of the CDK inhibitor protein p21 has been shown to inhibit vascular smooth muscle proliferation and intimal hyperplasia following angioplasty (Chang, M. W. et al., J. Clin. Invest, 96:2260 (1995); Yang, Z-Y. et al., Proc. Natl. Acad. Sci. (USA) 93:9905 (1996)). A small molecule CDK2 inhibitor CVT-313 (Ki = 95 nM) was shown to cause significant inhibition of neointima formation in animal models (Brooks, E. E. et al., J. Biol. Chem., 272:29207-2921 1 (1997)). Disregulation of cell cycle has been associated with polycystic kidney diseases, which are characterized by the growth of fluid-filled cysts in renal tubules. Treatment with small molecule inhibitors of CDKs yielded effective arrest of cystic disease in mouse models (Bukanov, N. O., et al., Nature, 4444:949-952 (2006)).
Infection by a variety of infectious agents, including fungi, protozoan parasites such as Plasmodium falciparum, and DNA and RNA viruses may be treated with CDK inhibitors. CDKs have been shown to be required for replication of herpes simplex virus (HSV) (Schang, L. M. et al., J. Virol., 72:5626 (1998)). Synovial tissue hyperplasia plays important roles in the development of rheumatoid arthritis; inhibition of synovial tissue proliferation may suppress inflammation and prevent joint destruction. It has been shown that over-expression of CDK inhibitor protein pl6 inhibited synovial fibroblast growth (Taniguchi, K. et al., Nat. Med., 5:760-767 (1999)) and joint swelling was substantially inhibited in animal arthritis models.
Selective inhibitors of some CDKs may also be used to protect normal untransformed cells by inhibiting specific phases of cell cycle progression (Chen, et al., J. Natl. Cancer Institute, 92: 1999-2008 (2000)). Pre-treatment with a selective CDK inhibitor prior to the use of a cytotoxic agent that inhibits a different phase of the cell cycle may reduce the side effects associated with the cytotoxic chemotherapy and possibly increase the therapeutic widow. It has been shown that induction of cellular protein inhibitors of CDKs (pi 6, p27 and p21) conferred strong resistance to paclitaxel- or cisplatin-mediated cytotoxicity on the inhibitor-responsive cells but not on the inhibitor-unresponsive cells (Schmidt, M, Oncogene, 2001 20:6164-71).
CDK4 and CDK6 are two functionally indistinguishable cyclin D dependent kinases. They are widely expressed with high levels of expression observed in cells of hematopoeitic lineage (CDK4/6 will be used throughout this document to reference both CDK4 and CDK6). CDK4/6 promotes Gl-S transition of the cell cycle by phosphorylating the retinoblastoma protein (Rb). CDK4 and CDK6 single knockout mice are viable and double knockout mice die around birth with defective
hematopoiesis (Satyanarayana, A. et al., Oncogene, 28:2925-39 (2009); Malumbres, M. et al., Cell, 1 18:493-504 (2004)). Strong evidence supports a significant involvement of the cyclin D-CDK4-pl6INK4A-Rb pathway in cancer development (Malumbres, M. et al., Nature Rev. Cancer, 1 :222-31 (2001)). Rb negatively regulates the cell cycle at Gl by sequestering E2F proteins that are required for initiation of S phase, p 1 is a key member of the ΓΝΚ4 family of CDK4/6 cellular inhibitors. The genes for Rb and pl6INK4A are tumor suppressors that are often deleted or silenced in cancer cells.
Additionally CDK4, CDK6 and cyclin D are reported to be amplified in hematologic malignancies and solid tumors. The importance of this pathway in oncogenesis is further supported by the finding that depletion or inactivation of CDK4 inhibits tumor growth in mouse tumor models (Yu, Q. et al., Cancer Cell, 9:23-32 (2006); Puyol, M. Cancer Cell, 18:63-73 (2010)). Rb and p 16^^ are rarely deleted in AML. However, the plS1^^ gene, another member of the ΓΝΚ4 family, has been reported to be down regulated by hypermethylation in up to 60% of AML (Naofumi, M. et al., Leukemia Res., 29:557-64 (2005); Drexler, H. G. Leukemia, 12:845-59 (1998); Herman, J. G. et al., Cancer Res., 57:837-41 (1997)), suggesting a possible critical role for CDK4/6 in AML cells.
FLT3 (Fms-like tyrosine kinase 3, FLK2) is a class III receptor tyrosine kinase. It is activated by the FLT3 ligand (FL) and signals through the PI3K, RAS, and JAK/STAT pathways (Scholl C. et al., Semin. Oncol., 35:336-45 (2008); Meshinchi S. et al., Clin. Cancer Res., 15:4263-9 (2009)). FLT3 plays a role in early hematopoiesis and FLT3 deficient mice have reduced numbers of progenitors of multiple lymphoid lineages (Mackarehtschian K, et al., Immunity, 3: 147-61 (1995). Activating mutations in FLT3 are found in approximately 30% of AML patients, representing the most frequent genetic alteration in the disease. About 75% of the activating mutations are internal tandem duplications (ITD) and 25% are point mutations in the activation loop of the kinase domain.
The most frequently identified activating point mutation is D835Y (Yamamoto et al., Blood, 97(8): 2434-2439 (2001)). However, mutations have also been found at N841I (Jiang, J. et al., Blood, 104(6): 1855-1858 (2004)) and Y842C (Kindler et al., Blood, 105(1): 335-340 (2005)). Additional point mutations have been identified in the juxtamembrane domain and kinase domain, although these have been shown to result in lower transforming potential (Reindel et al., Blood 107(9): 3700- 3707 (2006)).
Murine bone marrow transplanted with a retrovirus expressing the
FLT3-ITD has been shown to result in the production of a lethal myeloproliferative disease in mice (Kelly et al., Blood 99: 310-318 (2002)) characterized by leukocytosis consisting of mature neutrophils. This disease did not show a block in differentiation as seen in human AML suggesting that FLT3 mutations confer a proliferative or survival advantage to the cells. Additional oncogene mutation producing a block in
differentiation such as AML1/ETO is hypothesized to be required to produce disease that is more similar to human AML.
A number of FLT3 inhibitors have been tested in clinical trials.
Although they have shown initial clinical responses in AML, the responses observed were transient and resistance can develop rapidly (Weisberg, E. et al., Oncogene, 29:5120-34 (2010)). The major resistance mechanism appears to be through the acquisition of secondary mutations in FLT3, which may interfere with the binding of FLT3 inhibitors to the FLT3 receptor (Weisberg, E. et al., Oncogene, 29:5120-34 (2010); Chu, S. H. et al., Drug Resist. Update, 12:8-16 (2009)). One such resistance mutation (N676K) was identified in a patient at the time of clinical relapse while on multi-kinase FLT3 inhibitor midostaurin (PKC412) monotherapy (Heidel, F. et al., Blood, 107:293-300 (2006)). Combinations of FLT3 inhibitors with chemotherapy are being tested in clinical trials despite the recognition that chemotherapy is poorly tolerated. Additional possible mechanisms for lack of durable responses include inadequate target coverage (Pratz, K. W., et al., Blood, 139:3938-46 (2009)) and protection of AML cells in the bone marrow where stromal growth factors may provide proliferative signals in addition to FLT3 activation (Tarn, W. F. et al., Best Pract. Res. Clin. Haematol., 21 : 13-20 (2008)). Inhibitors with combined FLT3 and CDK4/6 inhibitory activities are novel and may prove beneficial in treating various cancers including, but not limited to, AML.
Fused tricyclic pyridine, pyrimidine, and triazine compounds useful for treating diseases mediated by CDK4 are disclosed in WO 2009/085185, published on July 9, 2009, which is hereby incorporated by reference in its entirety and for all purposes as if fully set forth herein. Various gem-disubstituted and spirocyclic compounds useful for treating diseases mediated by CDK4 are disclosed in WO 2009/0126584, published on October 15, 2009, which is hereby incorporated by reference in its entirety and for all purposes as if fully set forth herein.
A continued need exists for new compounds that can be used to modulate CDK4, CDK6, and/or FLT3 and can be used to treat various disease conditions associated with these kinases. The compounds of the present invention provide significant improvements in inhibition in one or more of these kinases and have properties making them excellent therapeutic candidates.
………………………
[01 16] In some embo f Formula I, the compound is
Figure imgf000049_0002
SCHEME 3

R1′-CI
Figure imgf000121_0001
Figure imgf000121_0002
Figure imgf000121_0003
Figure imgf000121_0004
3G …………………………………………………………..3H

Example 1. 9-((lr,4r)-4-Methylcyclohexyl)-N-(5,6,7,8-tetrahydro-1 ,6-naphthyridin-2-yl)-9H-py 3-d] pyrimidin-2-amine
Figure imgf000125_0001 KEY INTERMEDIATE 1
……………………………………..SECTION 1 BELOW
STEP A
Figure imgf000125_0002
4-Chloropyrimidine-2-amine (commercially available from Sigma-Aldrich, St. Louis, MO) (1000 g, 7.72 mol, 1.0 eq), trans- 4-methylcyclohexylamine hydrochloride (commercially available from TCI America, M1780) (1500 g, 10.03 mol, 1.3 eq) and TEA (3.23 L, 23.2 mol, 3.0 eq) were mixed together in n-butanol (8 L). The reaction mixture was heated at reflux for 36 hours and monitored using LCMS. Upon completion, the reaction mixture was cooled to room temperature, diluted with water (8 L) and extracted with EtOAc (2 x 10 L). The organic layers were combined, dried over Na2S04, and concentrated under reduced pressure to give the title compound (1770 g) which was us
STEP B
Figure imgf000125_0003
Synthesis of 5-iodo-A^-((lr,4r)-4-methylcyclohexyl)pyridine-2,4- diamine. N4-((lr,4r)-4-Methylcyclohexyl)pyridine-2,4-diamine (1770 g, 8.58 mol, 1.0 eq) was dissolved in anhydrous DMF (8 L). To this solution under N2 atmosphere at 10 °C was added NIS (1.93 kg, 8.58 mol, 1.0 eq) in portions over 10 minutes. Upon completion of the addition, the reaction mixture was stirred at room temperature for 2 hours. The reaction was monitored using LCMS. Upon completion, the reaction mixture was cooled using an ice bath, quenched with saturated aqueous sodium carbonate (5 L) and extracted with EtOAc (2 x 15 L). The combined organic extracts were washed with saturated aqueous sodium carbonate (2 x 5 L), water (3 x 2 L), dried over Na2S04, and concentrated under reduced pressure. The residue was purified using column chromatography eluting with 25% to 40% EtOAc in hexanes to provide the title compound (1.47 kg, 57% over two steps). ^-NMR (300 MHz, DMSO-d6) δ ppm 0.85 (3H, d, J= 7.2 Hz), 0.98 (1H, dd, J= 12.9, 2.7 Hz), 1.41 – 1.27 (3H, m), 1.66 (2H, d, J = 12.3 Hz), 1.78 (2H, d, J= 12.3 Hz), 3.85 (1H, m), 5.48 (1H, d, J= 8.1 Hz), 6.16 (2H, br s), 7.86
STEPC
Figure imgf000126_0001
Synthesis of 5-(3-fluoropyridin-4-yl)-N -((lr,4r)-4- methylcyclohexyl)pyrimidine-2,4-diamine. To a solution of 2,2,6,6- tetramethylpiperidine (commercially available from Sigma-Aldrich, St. Louis, MO) (997 mL, 5.87 mol, 3 eq) in anhydrous THF (6 L) under N2 atmosphere at 0 °C, was added n-BuLi (2.5 M in hexanes, 2.35 L, 5.87 mol, 3 eq) via an addition funnel over 30 minutes. Upon completion of the addition, the reaction mixture was stirred at 0 °C for 1 hour. The reaction mixture was cooled to -74 °C (acetone/ dry ice bath) and a solution of 3-fluoropyridine (commercially available from Sigma-Aldrich, St. Louis, MO) (561 g, 5.773 mol, 2.95 eq) in anhydrous THF (500 mL) was added over 15 minutes keeping the temperature below -63 °C. Upon completion of the addition, the reaction mixture was stirred at -74 °C for an additional 2 hours. A solution of ZnBr2 (1422 g, 6.32 mol, 3.22 eq) in anhydrous THF (3 L) was then added dropwise over 35 minutes keeping the temperature below -60 °C. Upon completion of the addition, the cold bath was removed and the reaction mixture was allowed to warm to room temperature. Then 5- iodo-N4-((lr,4r)-4-methylcyclohexyl)pyridine-2,4-diamine (650 g, 1.95 mol, 1.0 eq) was added in one portion followed by Pd(PPh3)4 (113 g, 97.8 mmol, 0.05 eq). The reaction mixture was heated at reflux overnight and monitored using LCMS. Upon completion, the reaction mixture was cooled to room temperature, quenched with saturated aqueous NaHC03 (6 L) and extracted with EtOAc (10 L x 2). The organic extracts were washed with saturated NaHC03 (2.5 L x 2) and brine (2.5 L), and were then concentrated under vacuum. The residue was dissolved in 2N HC1 (2.5 L) and washed with DCM (1.25 L x 3). The aqueous phase was adjusted to pH 10-12 by addition of aqueous 4N NaOH and extracted with DCM (1.5 L x 3). The organic extracts were washed with water (1.25 L x 2), dried and concentrated to give the title compound (540 g, 92%). ^-NMR (300 MHz, DMSO-d6) δ ppm 0.85 (3H, d, J= 7.2 Hz), 0.98 (1H, dd, J= 12.9, 2.7 Hz), 1.30 – 1.18 (3H, m), 1.64 (2H, d, J= 12.3 Hz), 1.74 (2H, d, J= 1 1.7 Hz), 3.96 (1H, m), 5.00 (1H, d, J= 8.4 Hz), 6.24 (2H, br s), 7.35 (1H, dd, J= 6.6, 4.4 Hz), 7.58 (1H, s), 8.37 (1H, d, J= 4.8 Hz), 8.50 (1H, d, J= 6.6 Hz) ppm.

STEPD
Figure imgf000127_0001
Synthesis of 9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-amine. To a solution of 5-(3- fluoropyridin-4-yl)-N4-((lr,4r)-4-methylcyclohexyl)pyrimidine-2,4-diamine (854 g, 2.84 mol, 1.0 eq) in anhydrous 1 -methyl-2-pyrrolidinone (8 L) under N2atmosphere at room temperature, was added LiHMDS (1.0 M in toluene, 8.5 L, 8.5 mol, 3.0 eq) over 30 minutes. Upon completion of the addition, the reaction mixture was heated at 90 °C overnight and monitored using LCMS. Upon completion, the reaction mixture was cooled to room temperature, quenched with ice cold water (10 L) and extracted with EtOAc (12 L). The organic phase was washed with saturated aqueous NaHC03 (4 L x 2), and water (2 L x 3). The aqueous layers were combined and back extracted with EtOAc (15 L x 2). The organic layers were combined, dried over Na2SO/t, and concentrated under reduced pressure. The solid thus obtained was suspended in DCM (2.5 L) and agitated using a rotary evaporator for 30minutes. The solid was collected by filtration, washed with DCM and dried to afford the title compound (400 g). The mother liquor was purified by column chromatography (eluting with DCM/MeOH = 50: 1) to afford, after triturating with DCM (750 mL), additional title compound (277 g, total: 677 g, yield: 84%). ¾ NMR (300 MHz, CD3OD) δ ppm 1.02 (d, J= 6.3 Hz, 3H), 1.33-1.20 (m, 2H), 1.67-1.60 (m, 2H), 1.95-1.84 (m, 4H), 1.58-1.45 (m, 2H), 4.87-4.77 (m, 1H), 7.94 (d, J= 5.1 Hz, 1H), 8.31 (d, J= 5.1 Hz, 1H), 8.87 (s, 1H), 8.96 (s, 1H) ppm; MS m/z: 28
……………………………………
COUPLER 1
Figure imgf000128_0001
Synthesis of tert-butyl 2-chloro-7,8-dihydro-l,6-naphthyridine-6(5H)-carboxylate. To a slurry of 2-chloro-5,6,7,8-tetrahydro-l,6-naphthyridine hydrochloride (106.1 g, 517 mmol, commercially available from D-L Chiral Chemicals, ST-0143) and N,N-diisopropylethylamine (80 g, 108 mL, 621 mmol, 1.2 eq) in DCM (1 L) was added a solution of di-tert-butyl dicarbonate (119 g, 543 mmol, 1.05 eq) in
DCM (100 mL) via an addition funnel within 1 hr. The reaction mixture became a clean solution and the solution thus obtained was stirred at room temperature for an additional hour and monitored using LCMS. Upon completion, the reaction mixture was concentrated. The residue was dissolved in EtOAc (1 L) and washed with water (3 x 300 mL), washed with brine (300 mL) and dried over MgSOzt. The solvent was evaporated under vacuum to give the title compound as an off- white solid (139 g, yield: 100%). lH NMR (400MHz ,CDC13) δ ppm 1.49 (9H, s), 2.97 (2H, t, J= 5.9 Hz), 3.73 (2H, t, J= 6.0 Hz), 4.57 (2H, s), 7.17 (1H, d, J= 8.0 Hz), 7.38 (1H, d, J= 8.0 Hz) ppm;
……………………………
STEP E
Figure imgf000128_0002
Synthesis of tert-butyl 2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido [4′,3 ‘ :4,5] pyrrolo [2,3-d] pyrimidin-2-yl)amino)-7,8-dihydro-l ,6- naphthyridine-6(5H)-carboxylate. To a solution of 9-((lr,4r)-4-methylcyclohexyl)- 9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-amine (2.81 g, lO mmol) in 1,4-dioxane (45 mL) were added tert-butyl 2-chloro-7,8-dihydro-l,6-naphthyridine-6(5H)- carboxylate (2.57 g, 9.55 mmol), 4,5-bis(diphenylphosphino)-9,9-dimethylxanene (231 mg, 0.40 mmol), and sodium t-butoxide (1.44 g, 15 mmol). Argon was bubbled through the mixture for 10 minutes. Tris(dibenzylideneacetone)dipalladium (0)(183 mg, 0.20 mmol) was added, and argon was again bubbled through the mixture for 5 minutes. The reaction mixture thus obtained was stirred at 100 °C for 3 hours whereupon HPLC-MS analysis indicated that the reaction was complete. The reaction mixture was cooled to 40 °C and diluted with DCM (90 mL) and treated with Si- triamine (functionalized silica gel, from Silicycle, FR31017TR130B) (2.8 g) overnight at room temperature. Celite® brand filter aid 545 (6 g) was added, and the mixture was filtered with a sintered glass funnel and the solid phase was rinsed with DCM (100 mL). The filtrate was concentrated to 25 mL on a rotary evaporator and diluted with a mixture of EtOAc and hexane (20 mL, 4: 1). The resulting slurry was stirred at room temperature for 5 hours. The solid was collected by filtration, washed with a mixture of EtOAc and hexane (20 mL, 1 : 1) and air dried for a few hours to provide the title compound as an off-white solid (4.90 g, 100% yield). lH NMR (500 MHz, CD2C12) δ ppm 1.06 (3H, d, J= 6.4 Hz), 1.34 – 1.22 (2H, m), 1.48 (9H, s), 1.67 (1H, br. s), 2.02 – 1.93 (4H, m), 2.63 (2H, dq, J= 3.1, 12.8 Hz), 2.88 (2H, t, J= 5.7 Hz), 3.74 (2H, t, J= 6.0 Hz), 4.57 (2H, s), 7.51 (1H, d, J= 8.6 Hz), 7.85 (1H, d, J= 5.1 Hz), 8.10 (1H, br. s), 8.42 (1H, d, J= 8.3 Hz), 8.46 (1H, d, J= 4.9 Hz), 8.97 (1H, s), 9.10 (1H, s) ppm;
…………………………
STEP F

Figure imgf000129_0001
1
Synthesis of 9-((l r,4r)-4-methylcyclohexyl)-N-(5,6,7,8-tetrahydro- l,6-naphthyridin-2-yl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-amine (1).
To a suspension of tert-butyl 2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amm^
6(5H)-carboxylate: 9-((lr,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3- d]pyrimidin-2-amine (4.65 g, 9.05 mmol) in MeOH (30 mL) were added concentrated HC1 (6.74 mL) and water (14 mL). The mixture thus obtained was stirred at room temperature overnight. 50% NaOH in water (4.8 mL) was added at 0 °C to the reaction mixture to adjust the pH value to 9. The precipitated yellow solid was collected by filtration, rinsed with water (25 mL) and air dried for 3 days to give the title compound (3.75 g, 100%). lH NMR (400 MHz, CDC13) δ ppm 1.07 (3H, d, J= 6.5 Hz), 1.29 – 1.25 (3H, m), 2.00 – 1.95 (3H, m), 2.02 (2H, s), 2.69 – 2.53 (2H, m), 2.89 (2H, t, J= 6.0 Hz), 3.26 (2H, t, J= 6.0 Hz), 4.04 (2H, s), 4.71 (1H, m, J= 12.8, 12.8 Hz), 7.41 (1H, d, J= 8.4 Hz), 7.84 (1H, d, J= 6.1 Hz), 7.84 (1H, d, J= 6.1 Hz), 8.03 (1H, s), 8.34 (1H, d, J= 8.4 Hz), 8.50 (1H, d, J= 5.3 Hz), 8.96 (1H, s), 9.08 (1H, s) ppm; LCMS m/z: 414 (M+l).
SECTION 2 BELOW
SYNTHESIS OF LABEL 5 FROM 1

Example 5. 2-Hydroxy-l-(2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido [4′,3 ‘ :4,5] pyrrolo [2,3-d] pyrimidin-2-yl)amino)-7,8-dihydro-l ,6- naphthyridin-6(5H)-yl)ethanone
Figure imgf000134_0001 LABEL 5
…………………………
COUPLER 2
Figure imgf000134_0002
Synthesis of 2,5-dioxopyrrolidin-l-yl 2-acetoxyacetate.
A 3-neck round-bottom flask equipped with a mechanical stirrer, thermocouple and addition funnel with nitrogen inlet was charged with N-hydroxysuccinimide (commercially available from Sigma- Aldrich, St. Louis, MO) (21 1 g, 1.83 mol) and DCM (2.25 L) at room temperature, resulting in a suspension. Pyridine (178 mL, 2.2 mol) was added in one portion with no change in the internal temperature. A solution of acetoxyacetyl chloride (commercially available from Sigma-Aldrich, St. Louis, MO) (197 mL, 1.83 mol) in DCM (225 mL) was added dropwise over 60 minutes and the temperature rose to 35 °C. Stirring was continued at room temperature for 2.5 hours. The reaction mixture was washed with water (IxlL), IN HCl (2xlL) and brine (IxlL). The organic layer was concentrated under vacuum and azeotroped with toluene (IxlL) to obtain the product as a white solid (367 g, 93%). lH NMR (400MHz, CDC13) δ 4.96 (2H, s), 2.86 (4H s), 2.19 (3H, s) ppm; LCMS m/z: 238 (M+Na).
…………………………………
STEP G
Figure imgf000134_0003
Synthesis of 2-(2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido [4′,3 ‘ :4,5] pyrrolo [2,3-d] pyrimidin-2-yl)amino)-7,8-dihydro-l ,6- naphthyridin-6(5H)-yl)-2-oxoethyl acetate.
To a suspension of 9-((lr,4r)-4- methylcyclohexyl)-N-(5,6,7,8-tetrahydro-l,6-naphthyridin-2-yl)-9H- pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-amine (1) (827 mg, 2.0 mmol) in chloroform (10 mL) were added diisopropylethylamine (258 mg, 348 uL, 2.0 mmol) and 2,5- dioxopyrrolidin- l-yl 2-acetoxyacetate (560 mg, 2.6 mmol). The reaction mixture thus obtained was stirred at room temperature for 30 minutes whereupon the mixture became a yellow solution. HPLC-MS analysis indicated that the reaction was complete. The reaction mixture was concentrated. MeOH (5 mL) and water (6 mL) were added to form a slurry which was stirred at room temperature for 1 hour. The solid was collected by filtration to give the title compound as a light yellow solid (1.04 g, 98% yield). lH NMR (400 MHz, CDC13, rotamers) δ ppm 1.08 (3H, d, J= 6.5 Hz), 1.37 – 1.20 (2H, m), 2.03 – 1.97 (4H, m), 2.22 (3H, s), 2.69 – 2.52 (2H, m, J= 2.9, 12.8, 12.8, 12.8 Hz), 3.08 – 2.93 (2H, m), 3.75 (1H, t, J= 5.9 Hz), 3.97 (1H, t, J= 5.6 Hz), 4.59 (1H, s), ), 4.80 – 4.65 (2H, m), ), 4.90 – 4.82 (2H, m), 7.57 – 7.45 (1H, m), 7.86 (1H, d, J= 5.7 Hz), 8.21 – 8.10 (1H, m), 8.49 – 8.40 (1H, m), 8.52 (1H, d, J= 5.3 Hz), 8.
…………………………………..
STEPH
Figure imgf000135_0001
LABEL 5
Synthesis of 2-hydroxy-l-(2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido [4′,3 ‘ :4,5] pyrrolo [2,3-d] pyrimidin-2-yl)amino)-7,8-dihydro-l ,6- naphthyridin-6(5H)-yl)ethanone (5).
To a solution of 2-(2-((9-((lr,4r)-4- methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8- dihydro-l,6-naphthyridin-6(5H)-yl)-2-oxoethyl acetate (514 mg, 1.0 mmol) in DCM (7.5 mL) and MeOH (2.5 mL) was added 0.5 M sodium methoxide solution in MeOH (0.30 mL, 0.15 mmol), and the reaction mixture was stirred at room temperature for 1 hour and monitored using LCMS. Upon completion, the reaction mixture was concentrated. The residue was treated with EtOH (5 mL) and water (10 mL) to provide a solid which was collected by filtration, washed with water, and dried in a vacuum oven at 55 °C overnight to give the title compound (5) as a white solid (468 mg, 99% yield).
lH NMR (500 MHz, acetic acid-d4, 373 K) δ ppm 1.09 (3H, d, J= 6.5 Hz), 1.31-1.43 (2H, m), 1.70-1.80 (1H, m), 1.99-2.03 (2H, m), 2.06-2.13 (2H, m), 2.68 (2H, dq, J= 3.3, 12.7 Hz), 3.10 (2H, t, J= 5.4 Hz), 3.88 (2H, br. s.), 4.46 (2H, br. s.), 4.77 (2H, br. s), 4.90 (1H, tt, J= 3.9, 12.4 Hz), 7.76 (1H, d, J= 8.5 Hz), 8.33 (1H, d, J= 8.5 Hz), 8.40 (1H, d, J= 6.0 Hz), 8.63 (1H, d, J= 6.0 Hz), 9.35 (1H, s), 9.43 (1H, s) ppm; L
………………………………………………
STEP I
Figure imgf000136_0001
5                                                                                          LABEL HCI Dihydrate
[0222] Synthesis of 2-hydroxy-l-(2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido [4′,3 ‘ :4,5] pyrrolo [2,3-d] pyrimidin-2-yl)amino)-7,8-dihydro-l ,6- naphthyridin-6(5H)-yl)ethanone monohydrochloride dihydrate. To a suspension of 2-hydroxy-l-(2-((9-((lr,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3- d]pyrimidin-2-yl)amino)-7,8-dihydro-l,6-naphthyridin-6(5H)-yl)ethanone (472 mg, 1.0 mmol) in water (2 mL) was added 2 N HCI (2 mL). The mixture became a clear solution. The pH value of the solution was adjusted to 4 by addition of 2 N NaOH at 0 °C and the precipitated light yellow solid was collected by filtration. The collected solid was washed with cold water three times. The solid was dried under vacuum to give the title compound as a light yellow solid (469 mg, 92% yield).
¾ NMR (500 MHz, DMSO-d6) δ 1.02 (3H, d, J= 5.0 Hz), 1.20- 1.30 (2H, m), 1.64 (1H, m), 1.88-1.90 (4H, m), 2.59-2.66 (2H, m), 2.85-2.95 (2H, m), 3.71(1H, m), 3.83 (1H, m), 4.19-4.22 (2H, m), 4.60-4.67 (2H, m), 4.85 (1H, m), 7.75 (1H, d, J= 8.5 Hz), 8.19 (1H, d, J= 8.5 Hz), 8.55 (1H, d, J= 5.0 Hz), 8.63 (1H, d, J= 5.0 Hz), 9.47 (1H, s), 9.58 (1H, s), 10.59 (1H, br.s) ppm; LCMS m/z: 472 (M+l). Anal.
Figure imgf000136_0002
Calc: C = 57.40, H = 6.30, N = 18.02; Found: C = 57.06, H = 6.31, N = 17.92. [0223] Alternative Synthesis of Hydrochloride Salt of 2-Hydroxy-l-(2-((9-
((lr,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2- yl)amino)-7,8-dihydro-l,6-naphthyridin-6(5H)-yl)ethanone. To a suspension of 2- hydroxy- 1 -(2-((9-(( 1 r,4r)-4-methylcyclohexyl)-9H-pyrido [4′,3 ‘ :4,5]pyrrolo [2,3 – d]pyrimidin-2-yl)amino)-7,8-dihydro-l,6-naphthyridin-6(5H)-yl)ethanone (2.385 g, 5.0 mmol) in water (10 mL) was added 2N HC1 (10 mL) at 20°C. The mixture became a clear light yellow solution. The pH value of the solution was adjusted to 4 by addition of 2N NaOH through addition funnel at 0° C, and the precipitated yellow solid was collected by filtration. The resulting solid was washed with cold water three times. The solid was dried under vacuum at 50° C for two days to provide 2.49 g of the hydrochloride salt of 2-hydroxy-l-(2-((9-((lr,4r)-4-methylcyclohexyl)-9H- pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-l,6-naphthyridin- 6(5H)-yl)ethanone as a solid. This salt was also obtained as a hydrate.
………………
Example 5
2-Hydroxy-1-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone
Figure US20140163052A1-20140612-C00127
Figure US20140163052A1-20140612-C00128
Synthesis of 2,5-dioxopyrrolidin-1-yl 2-acetoxyacetate
A 3-neck round-bottom flask equipped with a mechanical stirrer, thermocouple and addition funnel with nitrogen inlet was charged with N-hydroxysuccinimide (commercially available from Sigma-Aldrich, St. Louis, Mo.) (211 g, 1.83 mol) and DCM (2.25 L) at room temperature, resulting in a suspension. Pyridine (178 mL, 2.2 mol) was added in one portion with no change in the internal temperature. A solution of acetoxyacetyl chloride (commercially available from Sigma-Aldrich, St. Louis, Mo.) (197 mL, 1.83 mol) in DCM (225 mL) was added dropwise over 60 minutes and the temperature rose to 35° C. Stirring was continued at room temperature for 2.5 hours. The reaction mixture was washed with water (1×1 L), 1N HCl (2×1 L) and brine (1×1 L). The organic layer was concentrated under vacuum and azeotroped with toluene (1×1 L) to obtain the product as a white solid (367 g, 93%). 1H NMR (400 MHz, CDCl3) δ 4.96 (2H, s), 2.86 (4H, s), 2.19 (3H, s) ppm; LCMS m/z: 238 (M+Na).
Figure US20140163052A1-20140612-C00129
Synthesis of 2-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)-2-oxoethyl acetate
To a suspension of 9-((1r,4r)-4-methylcyclohexyl)-N-(5,6,7,8-tetrahydro-1,6-naphthyridin-2-yl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-amine (1) (827 mg, 2.0 mmol) in chloroform (10 mL) were added diisopropylethylamine (258 mg, 348 uL, 2.0 mmol) and 2,5-dioxopyrrolidin-1-yl 2-acetoxyacetate (560 mg, 2.6 mmol). The reaction mixture thus obtained was stirred at room temperature for 30 minutes whereupon the mixture became a yellow solution. HPLC-MS analysis indicated that the reaction was complete. The reaction mixture was concentrated. MeOH (5 mL) and water (6 mL) were added to form a slurry which was stirred at room temperature for 1 hour. The solid was collected by filtration to give the title compound as a light yellow solid (1.04 g, 98% yield). 1H NMR (400 MHz, CDCl3, rotamers) δ ppm 1.08 (3H, d, J=6.5 Hz), 1.37-1.20 (2H, m), 2.03-1.97 (4H, m), 2.22 (3H, s), 2.69-2.52 (2H, m, J=2.9, 12.8, 12.8, 12.8 Hz), 3.08-2.93 (2H, m), 3.75 (1H, t, J=5.9 Hz), 3.97 (1H, t, J=5.6 Hz), 4.59 (1H, s),), 4.80-4.65 (2H, m),), 4.90-4.82 (2H, m), 7.57-7.45 (1H, m), 7.86 (1H, d, J=5.7 Hz), 8.21-8.10 (1H, m), 8.49-8.40 (1H, m), 8.52 (1H, d, J=5.3 Hz), 8.98 (1H, s), 9.11 (1H, s) ppm; LCMS m/z: 514 (M+1).
Figure US20140163052A1-20140612-C00130
Synthesis of 2-hydroxy-1-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone (5)
To a solution of 2-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)-2-oxoethyl acetate (514 mg, 1.0 mmol) in DCM (7.5 mL) and MeOH (2.5 mL) was added 0.5 M sodium methoxide solution in MeOH (0.30 mL, 0.15 mmol), and the reaction mixture was stirred at room temperature for 1 hour and monitored using LCMS. Upon completion, the reaction mixture was concentrated. The residue was treated with EtOH (5 mL) and water (10 mL) to provide a solid which was collected by filtration, washed with water, and dried in a vacuum oven at 55° C. overnight to give the title compound (5) as a white solid (468 mg, 99% yield). 1H NMR (500 MHz, acetic acid-d4, 373 K) δ ppm 1.09 (3H, d, J=6.5 Hz), 1.31-1.43 (2H, m), 1.70-1.80 (1H, m), 1.99-2.03 (2H, m), 2.06-2.13 (2H, m), 2.68 (2H, dq, J=3.3, 12.7 Hz), 3.10 (2H, t, J=5.4 Hz), 3.88 (2H, br. s.), 4.46 (2H, br. s.), 4.77 (2H, br. s), 4.90 (1H, tt, J=3.9, 12.4 Hz), 7.76 (1H, d, J=8.5 Hz), 8.33 (1H, d, J=8.5 Hz), 8.40 (1H, d, J=6.0 Hz), 8.63 (1H, d, J=6.0 Hz), 9.35 (1H, s), 9.43 (1H, s) ppm; LCMS m/z: 472 (M+1).
Figure US20140163052A1-20140612-C00131
Synthesis of 2-hydroxy-1-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone monohydrochloride dihydrate
To a suspension of 2-hydroxy-1-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone (472 mg, 1.0 mmol) in water (2 mL) was added 2 N HCl (2 mL). The mixture became a clear solution. The pH value of the solution was adjusted to 4 by addition of 2 N NaOH at 0° C. and the precipitated light yellow solid was collected by filtration. The collected solid was washed with cold water three times. The solid was dried under vacuum to give the title compound as a light yellow solid (469 mg, 92% yield). 1H NMR (500 MHz, DMSO-d6) δ 1.02 (3H, d, J=5.0 Hz), 1.20-1.30 (2H, m), 1.64 (1H, m), 1.88-1.90 (4H, m), 2.59-2.66 (2H, m), 2.85-2.95 (2H, m), 3.71 (1H, m), 3.83 (1H, m), 4.19-4.22 (2H, m), 4.60-4.67 (2H, m), 4.85 (1H, m), 7.75 (1H, d, J=8.5 Hz), 8.19 (1H, d, J=8.5 Hz), 8.55 (1H, d, J=5.0 Hz), 8.63 (1H, d, J=5.0 Hz), 9.47 (1H, s), 9.58 (1H, s), 10.59 (1H, br.s) ppm; LCMS m/z: 472 (M+1). Anal. (C26H29N7O2—HCl.2H2O) Calc: C=57.40, H=6.30, N=18.02. Found: C=57.06, H=6.31, N=17.92.
Alternative Synthesis of Hydrochloride Salt of 2-Hydroxy-1-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl) amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl) ethanone
To a suspension of 2-hydroxy-1-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone (2.385 g, 5.0 mmol) in water (10 mL) was added 2N HCl (10 mL) at 20° C. The mixture became a clear light yellow solution. The pH value of the solution was adjusted to 4 by addition of 2N NaOH through addition funnel at 0° C., and the precipitated yellow solid was collected by filtration. The resulting solid was washed with cold water three times. The solid was dried under vacuum at 50° C. for two days to provide 2.49 g of the hydrochloride salt of 2-hydroxy-1-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone as a solid. This salt was also obtained as a hydrate.
………………………
J. Med. Chem.201457 (8), pp 3430–3449
DOI: 10.1021/jm500118j
2-Hydroxy-1-(2-((9-((1r,4r)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone (28)
 compound 28 as a white solid (468 mg, 99% yield).
1H NMR (500 MHz, acetic acid-d4, 373 K) δ ppm 9.43 (1 H, s), 9.35 (1 H, s), 8.63 (1H, d, J = 6.0 Hz), 8.40 (1 H, d, J = 6.0 Hz), 8.33 (1 H, d, J = 8.5 Hz), 7.76 (1 H, d, J = 8.5 Hz), 4.90 (1 H, m), 4.77 (2 H, br s), 4.46 (2 H, br s), 3.88 (2 H, br s), 3.10 (2 H, t, J = 5.4 Hz), 2.68 (2 H, dq, J = 12.7, 3.3 Hz), 2.06–2.13 (2 H, m), 1.99–2.03 (2 H, m), 1.70–1.80 (1 H, m), 1.31–1.43 (2 H, m), 1.09 (3H, d, J = 6.5 Hz).
HRMS (ESI) m/z: calculated for [M + H]+ 472.2455, found 472.2461.


…………………………

PAPER

OPRD
 Chemical Process R&D, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California91320
 Norchim S.A.S., 33 Quai d’Amont, Saint Leu d’Esserent, France 60340
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/op500367p
Abstract Image
The development of a synthetic route to manufacture the drug candidate AMG 925 on kilogram scale is reported herein. The hydrochloride salt of AMG 925 was prepared in 23% overall yield over eight steps from commercially available raw materials, and more than 8 kg of the target molecule were delivered. The synthetic route features a Buchwald–Hartwig amination using BrettPhos as ligand and conducted to afford 12 kg of product in a single batch. In addition, this work highlights the challenges associated with the use of poorly soluble process intermediates in the manufacture of active pharmaceutical ingredients. Creative solutions had to be devised to conduct seemingly routine activities such as salt removal, pH adjustment, and heavy metal scavenging due to the low solubility of the process intermediates. Finally, a slurry-to-slurry amidation protocol was optimized to allow for successful scale-up.
Manufacture of 2-Hydroxy-1-(2-((9-((1R,4R)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone (AMG 925)
AMG 925 was isolated in 91.5% yield (8.31 kg), 95.5% overall mass balance, 99.9 wt %, and 99.7 LCAP.
Mp 213–215 °C;
1H NMR (400 MHz, acetic acid-d4, mixture of two rotamers at 20 °C) 9.47–9.59 (m, 2H), 8.76 (d, 1H, J = 6 Hz), 8.55 (d, 1H, J = 6 Hz), 8.48 (d, 1H,J = 9 Hz), 7.79–7.92 (m, 1H), 4.95 (t, 1H,J = 12 Hz), 4.87 and 4.68 (2 singlets, 2H), 4.47–4.59 (m, 2H), 4.04 and 3.80 (2 triplets, 2H, J= 6 Hz), 3.03–3.17 (m, 2H), 2.65–2.82 (m, 2H), 1.96–2.15 (m, 4H), 1.77 (br s, 1H), 1.39 (q, 2H, J = 12 Hz), 1.09 (d, 3H, J = 7 Hz);
13C NMR (100 MHz, acetic acid-d4, mixture of two rotamers at 20 °C) 171.9, 171.8, 158.4, 157.8, 154.7, 149.0, 148.9, 141.6, 135.2, 132.9, 126.3, 124.1, 123.6, 117.7, 113.7, 113.6, 107.5, 107.4, 60.1, 59.9, 56.3, 43.7, 42.6, 40.5, 38.7, 34.0, 31.5, 29.8, 28.8, 28.1, 21.5.
Manufacture of 2-Hydroxy-1-(2-((9-((1R,4R)-4-methylcyclohexyl)-9H-pyrido[4′,3′:4,5]pyrrolo[2,3-d]pyrimidin-2-yl)amino)-7,8-dihydro-1,6-naphthyridin-6(5H)-yl)ethanone Hydrochloride (AMG 925 HCl)
AMG 925 HCl was isolated in 92.5% yield (7.96 kg), 99.1% overall mass balance, 83.8 wt % AMG 925, 99.75 LCAP AMG 925, 6.2 wt % Cl, 9.6 wt % water, 3800 ppm AcOH, d10 4.0 μm,d50 15.2 μm, d90 38.8 μ, Vm 18.7 μm, BET surface area 1.5 m2/g.
1H NMR (400 MHz, acetic acid-d4, mixture of two rotamers at 20 °C) 9.63 (s, 1H), 9.56 (s, 1H), 8.71–8.76 (m, 1H), 8.60–8.66 (m, 1H), 8.20–8.29 (m, 1H), 7.90–7.98 (m, 1H), 4.90–5.01 (m, 1H), 4.86 and 4.70 (2 singlets, 2H), 4.53 and 4.51 (2 singlets, 2H), 4.05 and 3.82 (2 triplets, 2H, J = 6 Hz), 3.11–3.26 (m, 2H), 2.68 (q, 2H, J = 12 Hz), 1.95–2.13 (m, 4H), 1.74 (br s, 1H), 1.36 (q, 2H, J = 12 Hz), 1.06 (d, 3H, J = 8 Hz);
13C NMR (100 MHz, acetic acid-d4, mixture of two rotamers at 20 °C) 174.9, 174.8, 161.3, 161.2, 160.5, 157.5, 151.6, 151.5, 149.3, 148.9, 145.5, 138.1, 136.0, 129.3, 129.2, 127.1, 126.6, 120.9, 116.7, 116.6, 110.8, 110.7, 63.0, 62.9, 59.3, 46.4, 45.3, 43.2, 41.3, 36.9, 34.3, 32.6, 31.3, 30.6, 24.4; exact mass [C26H29N7O2 + H]+: calculated = 472.2461, measured = 472.2451.

References:

1. K. Keegan et al, Preclinical evaluation of AMG 925, a FLT3/CDK4 dual kinase inhibitor for treating acute myeloid leukemia. Mol Cancer Ther. 2014 Apr;13(4):880-9.
2. ZH Li, et al, Discovery of AMG 925, a FLT3 and CDK4 Dual Kinase Inhibitor with Preferential Affinity for the Activated State of FLT3, J. Med Chem, March 18, 2014

CS-3150, (XL550) The next Japanese sartan in clinical trials




CS-3150,  (XL550)

CS 3150, angiotensin II receptor antagonist,  for the treatment or prevention of such hypertension and heart disease similar to olmesartan , losartan, candesartan , valsartan,  irbesartan,  telmisartan, eprosartan,
 Cas name 1H-​Pyrrole-​3-​carboxamide, 1-​(2-​hydroxyethyl)​-​4-​methyl-​N-​[4-​(methylsulfonyl)​phenyl]​-​5-​[2-​(trifluoromethyl)​phenyl]​-​, (5S)​-
CAS 1632006-28-0 for S conf
MF C22 H21 F3 N2 O4 S
MW 466.47
(S)-1-(2-hydroxyethyl)-4-methyl-N-[4-(methylsulfonyl)phenyl]-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxamide
CAS 1632006-28-0 for S configuration
1- (2-hydroxyethyl) -4-methyl -N- [4- (methylsulfonyl) phenyl] -5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxamide
(S) -1- (2- hydroxyethyl) -4-methyl -N- [4- (methylsulfonyl) phenyl] -5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxamide
(+/-)-1-(2-hydroxyethyl)-4-methyl-N-[4-(methylsulfonyl)phenyl]-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxamide, CAS 880780-76-7
(+)-1-(2-hydroxyethyl)-4-methyl-N-[4-(methylsulfonyl)phenyl]-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxamide..1072195-82-4
(-)-1-(2-hydroxyethyl)-4-methyl-N-[4-(methylsulfonyl)phenyl]-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxamide..1072195-83-5
WO2008 / 126831 (US Publication US2010-0093826)http://www.google.co.in/patents/EP2133330A1?cl=en
WO2006 / 012642 (US Publication US2008-0234270)


JAPAN PHASE 2……….Phase 2 Study to Evaluate Efficacy and Safety of CS-3150 in Patients with Essential Hypertension
Phase II Diabetic nephropathies; Hypertension
  • 01 Jan 2015 Daiichi Sankyo initiates a phase IIb trial for Diabetic nephropathies in Japan (NCT02345057)
  • 01 Jan 2015 Daiichi Sankyo initiates a phase IIb trial for Hypertension in Japan (NCT02345044)
  • 01 May 2013 Phase-II clinical trials in Diabetic nephropathies in Japan (PO)
  •  Currently, angiotensin II receptor antagonists and calcium antagonists are widely used as a medicament for the treatment or prevention of such hypertension or heart disease.
     Mineralocorticoid receptor (MR) (aldosterone receptor) has been known to play an important role in the control of body electrolyte balance and blood pressure, spironolactone having a steroid structure, MR antagonists such as eplerenone, are known to be useful in the treatment of hypertension-heart failure.
     Renin – angiotensin II receptor antagonists are inhibitors of angiotensin system is particularly effective in renin-dependent hypertension, and show a protective effect against cardiovascular and renal failure. Also, the calcium antagonists, and by the function of the calcium channel antagonizes (inhibits), since it has a natriuretic action in addition to the vasodilating action, is effective for hypertension fluid retention properties (renin-independent) .
     Therefore, the MR antagonist, when combined angiotensin II receptor antagonists or calcium antagonists, it is possible to suppress the genesis of multiple hypertension simultaneously, therapeutic or prophylactic effect of the stable and sufficient hypertension irrespective of the etiology is expected to exhibit.
     Also, diuretics are widely used as a medicament for the treatment or prevention of such hypertension or heart disease. Diuretic agent is effective in the treatment of hypertension from its diuretic effect. Therefore, if used in combination MR antagonists and diuretics, the diuretic effect of diuretics, it is possible to suppress the genesis of multiple blood pressure at the same time, shows a therapeutic or prophylactic effect of the stable and sufficient hypertension irrespective of the etiology it is expected.
     1- (2-hydroxyethyl) -4-methyl -N- [4- (methylsulfonyl) phenyl] -5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxamide (hereinafter, compound ( I)) is, it is disclosed in Patent Documents 1 and 2, hypertension, for the treatment of such diabetic nephropathy are known to be useful.
CS-3150 (XL550) is a small-molecule antagonist of the mineralocorticoid receptor (MR), a nuclear hormone receptor implicated in a variety of cardiovascular and metabolic diseases. MR antagonists can be used to treat hypertension and congestive heart failure due to their vascular protective effects. Recent studies have also shown beneficial effects of adding MR antagonists to the treatment regimen for Type II diabetic patients with nephropathy. CS-3150 is a non-steroidal, selective MR antagonist that has the potential for the treatment of hypertension, congestive heart failure, or end organ protection due to vascular damage.
Useful as a mineralocorticoid receptor (MR) antagonist, for treating hypertension, cardiac failure and diabetic nephropathy. It is likely to be CS-3150, a non-steroidal MR antagonist, being developed by Daiichi Sankyo (formerly Sankyo), under license from Exelixis, for treating hypertension and diabetic nephropathy (phase 2 clinical, as of March 2015). In January 2015, a phase II trial for type 2 diabetes mellitus and microalbuminuria was planned to be initiated later that month (NCT02345057).
Exelixis discovered CS-3150 and out-licensed the compound to Daiichi-Sankyo. Two phase 2a clinical trials, one in hypertensive patients and the other in type 2 diabetes with albuminuria, are currently being conducted in Japan by Daiichi-Sankyo.

Mineralocorticoid receptor (MR) (aldosterone receptor) has been known to play an important role in the control of body electrolyte balance and blood pressure, spironolactone having a steroid structure, MR antagonists such as eplerenone, are known to be useful in the treatment of hypertension-heart failure.
CS-3150 (XL550) is a small-molecule antagonist of the mineralocorticoid receptor (MR), a nuclear hormone receptor implicated in a variety of cardiovascular and metabolic diseases. MR antagonists can be used to treat hypertension and congestive heart failure due to their vascular protective effects. Recent studies have also shown beneficial effects of adding MR antagonists to the treatment regimen for Type II diabetic patients with nephropathy. CS-3150 is a non-steroidal, selective MR antagonist that has the potential for the treatment of hypertension, congestive heart failure, or end organ protection due to vascular damage.
Exelixis discovered CS-3150 and out-licensed the compound to Daiichi-Sankyo. Two phase 2a clinical trials, one in hypertensive patients and the other in type 2 diabetes with albuminuria, are currently being conducted in Japan by Daiichi-Sankyo.
Daiichi Sankyo (formerly Sankyo), under license from Exelixis, is developing CS-3150 (XL-550), a non-steroidal mineralocorticoid receptor (MR) antagonist, for the potential oral treatment of hypertension and diabetic nephropathy, microalbuminuria ,  By October 2012, phase II development had begun ; in May 2014, the drug was listed as being in phase IIb development . In January 2015, a phase II trial for type 2 diabetes mellitus and microalbuminuria was planned to be initiated later that month. At that time, the trial was expected to complete in March 2017 .
Exelixis, following its acquisition of X-Ceptor Therapeutics in October 2004 , was investigating the agent for the potential treatment of metabolic disorders and cardiovascular diseases, such as hypertension and congestive heart failure . In September 2004, Exelixis expected to file an IND in 2006. However, it appears that the company had fully outlicensed the agent to Sankyo since March 2006 .
DescriptionSmall molecule antagonist of the mineralocorticoid receptor (MR)
Molecular TargetMineralocorticoid receptor
Mechanism of ActionMineralocorticoid receptor antagonist
Therapeutic ModalitySmall molecule
In January 2015, a multi-center, placebo-controlled, randomized, 5-parallel group, double-blind, phase II trial (JapicCTI-152774;  NCT02345057; CS3150-B-J204) was planned to be initiated later that month in Japan, in patients with type 2 diabetes mellitus and microalbuminuria, to assess the efficacy and safety of different doses of CS-3150 compared to placebo. At that time, the trial was expected to complete in March 2017; later that month, the trial was initiated in the Japan
By October 2012, phase II development had begun in patients with essential hypertension
By January 2011, phase I trials had commenced in Japan
Several patents WO-2014168103,
WO-2015012205 and WO-2015030010
XL-550, claimed in WO-2006012642,
………………………………………………………………….
(Example 3)(+/-)-1-(2-hydroxyethyl)-4-methyl-N-[4-(methylsulfonyl)phenyl]-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxamide
  • After methyl 4-methyl-5-[2-(trifluoromethyl) phenyl]-1H-pyrrole-3-carboxylate was obtained by the method described in Example 16 of WO 2006/012642 , the following reaction was performed using this compound as a raw material.
  • Methyl 4-methyl-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxylate (1.4 g, 4.9 mmol) was dissolved in methanol (12 mL), and a 5 M aqueous sodium hydroxide solution (10 mL) was added thereto, and the resulting mixture was heated under reflux for 3 hours. After the mixture was cooled to room temperature, formic acid (5 mL) was added thereto to stop the reaction. After the mixture was concentrated under reduced pressure, water (10 mL) was added thereto to suspend the resulting residue. The precipitated solid was collected by filtration and washed 3 times with water. The obtained solid was dried under reduced pressure, whereby 4-methyl-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxylic acid (1.1 g, 83%) was obtained as a solid. The thus obtained solid was suspended in dichloromethane (10 mL), oxalyl chloride (0.86 mL, 10 mmol) was added thereto, and the resulting mixture was stirred at room temperature for 2 hours. After the mixture was concentrated under reduced pressure, the residue was dissolved in tetrahydrofuran (10 mL), and 4-(methylsulfonyl)aniline hydrochloride (1.0 g, 4.9 mmol) and N,N-diisopropylethylamine (2.8 mL, 16 mmol) were sequentially added to the solution, and the resulting mixture was heated under reflux for 18 hours. After the mixture was cooled to room temperature, the solvent was distilled off under reduced pressure, and acetonitrile (10 mL) and 3 M hydrochloric acid (100 mL) were added to the residue. A precipitated solid was triturated, collected by filtration and washed with water, and then, dried under reduced pressure, whereby 4-methyl-N-[4-(methylsulfonyl) phenyl]-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxamide (1.4 g, 89%) was obtained as a solid.
    1H-NMR (400 MHz, DMSO-d6) δ11.34 (1H, brs,), 9.89 (1H, s), 7.97 (2H, d, J = 6.6 Hz), 7.87-7.81 (3H, m), 7.73 (1H, t, J = 7.4 Hz), 7.65-7.61 (2H, m), 7.44 (1H, d, J = 7.8 Hz), 3.15 (3H, s), 2.01 (3H, s).
  • Sodium hydride (0.12 g, 3 mmol, 60% dispersion in mineral oil) was dissolved in N,N-dimethylformamide (1.5 mL), and 4-methyl -N-[4-(methylsulfonyl)phenyl]-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxamide (0.47 g, 1.1 mmol) was added thereto, and then, the resulting mixture was stirred at room temperature for 30 minutes. Then, 1,3,2-dioxathiolane-2,2-dioxide (0.14 g, 1.2 mmol) was added thereto, and the resulting mixture was stirred at room temperature. After 1 hour, sodium hydride (40 mg, 1.0 mmol, oily, 60%) was added thereto again, and the resulting mixture was stirred for 30 minutes. Then, 1,3,2-dioxathiolane-2,2-dioxide (12 mg, 0.11 mmol) was added thereto, and the resulting mixture was stirred at room temperature for 1 hour. After the mixture was concentrated under reduced pressure, methanol (5 mL) was added to the residue and insoluble substances were removed by filtration, and the filtrate was concentrated again. To the residue, tetrahydrofuran (2 mL) and 6 M hydrochloric acid (2 mL) were added, and the resulting mixture was stirred at 60°C for 16 hours. The reaction was cooled to room temperature, and then dissolved in ethyl acetate, and washed with water and saturated saline. The organic layer was dried over anhydrous sodium sulfate and filtered. Then, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (ethyl acetate), whereby the objective compound (0.25 g, 48%) was obtained.
    1H-NMR (400 MHz, CDCl3) δ: 7.89-7.79 (m, 6H), 7.66-7.58 (m, 2H), 7.49 (s, 1H), 7.36 (d, 1H, J = 7.4Hz), 3.81-3.63 (m, 4H), 3.05 (s, 3H), 2.08 (s, 3H).
    HR-MS (ESI) calcd for C22H22F3N2O4S [M+H]+, required m/z: 467.1252, found: 467.1246.
    Anal. calcd for C22H21F3N2O4S: C, 56.65; H, 4.54; N, 6.01; F, 12.22; S, 6.87. found: C, 56.39; H, 4.58; N, 5.99; F, 12.72; S, 6.92.
(Example 4)
Optical Resolution of Compound of Example 3
  • Resolution was performed 4 times in the same manner as in Example 2, whereby 74 mg of Isomer C was obtained as a solid from a fraction containing Isomer C (tR = 10 min), and 71 mg of Isomer D was obtained as a solid from a fraction containing Isomer D (tR = 11 min).
  • Isomer C: (+)-1-(2-hydroxyethyl)-4-methyl-N-[4-(methylsulfonyl)phenyl]-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxamide
    [α]D 21: +7.1° (c = 1.0, EtOH) .
    1H-NMR (400 MHz, CDCl3) δ: 7.91 (s, 1H), 7.87-7.79 (m, 5H), 7.67-7.58 (m, 2H), 7.51 (s, 1H), 7.35 (d, 1H, J = 7.0 Hz), 3.78-3.65 (m, 4H), 3.05 (s, 3H), 2.07 (s, 3H).
    HR-MS (ESI) calcd for C22H22F3N2O4S [M+H]+, required m/z: 467.1252, found: 467.1260.
    Retention time: 4.0 min.
  • Isomer D: (-)-1-(2-hydroxyethyl)-4-methyl-N-[4-(methylsulfonyl)phenyl]-5-[2-(trifluoromethyl)phenyl]-1H-pyrrole-3-carboxamide
    [α]D 21: -7.2° (c = 1.1, EtOH) .
    1H-NMR (400 MHz, CDCl3) δ: 7.88-7.79 (m, 6H), 7.67-7.58 (m, 2H), 7.50 (s, 1H), 7.36 (d, 1H, J = 7.5 Hz), 3.79-3.65 (m, 4H), 3.05 (s, 3H), 2.08 (s, 3H).
    HR-MS (ESI) calcd for C22H22F3N2O4S [M+H]+, required m/z: 467.1252, found: 467.1257.
    Retention time: 4.5 min.
……………………………………………….
WO 2014168103

 Step B: pyrrole derivative compounds (A ‘)
[Of 16]
(Example 1) 2-bromo-1- [2- (trifluoromethyl) phenyl] propan-1-one
[Of 19]
 1- [2- (trifluoromethyl) phenyl] propan-1-one 75 g (370 mmol) in t- butyl methyl ether (750 mL), and I was added bromine 1.18 g (7.4 mmol). After confirming that the stirred bromine color about 30 minutes at 15 ~ 30 ℃ disappears, cooled to 0 ~ 5 ℃, was stirred with bromine 59.13 g (370 mmol) while keeping the 0 ~ 10 ℃. After stirring for about 2.5 hours, was added while maintaining 10 w / v% aqueous potassium carbonate solution (300 mL) to 0 ~ 25 ℃, was further added sodium sulfite (7.5 g), was heated to 20 ~ 30 ℃. The solution was separated, washed in the resulting organic layer was added water (225 mL), to give t- butyl methyl ether solution of the title compound and the organic layer was concentrated under reduced pressure (225 mL).
 1 H NMR (400 MHz, CDCl 3 ) delta: 1.91 (3H, D, J = 4.0 Hz), 4.97 (1H, Q, J = 6.7 Hz), 7.60 ~ 7.74 (4H, M).
(Example 2) 2-cyano-3-methyl-4-oxo-4- [2- (trifluoromethyl) phenyl] butanoate
[Of 20]
 2-bromo-1- [2- (trifluoromethyl) phenyl] propan-1 / t- butyl methyl ether solution (220 mL) in dimethylacetamide (367 mL), ethyl cyanoacetate obtained in Example 1 53.39 g (472 mmol), potassium carbonate 60.26 g (436 mmol) were sequentially added, and the mixture was stirred and heated to 45 ~ 55 ℃. After stirring for about 2 hours, 20 is cooled to ~ 30 ℃, water (734 mL) and then extracted by addition of toluene (367 mL), washed by adding water (513 mL) was carried out in the organic layer (2 times implementation). The resulting organic layer was concentrated under reduced pressure to obtain a toluene solution of the title compound (220 mL).
 1 H NMR (400 MHz, CDCl 3 ) delta: 1.33 ~ 1.38 (6H, M), 3.80 ~ 3.93 (2H, M), 4.28 ~ 4.33 (2H, M), 7.58 ~ 7.79 (4H, M).
(Example 3) 2-chloro-4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid ethyl
[Of 21]
 The 20 ~ 30 ℃ 2-cyano-3-methyl-4-oxo-4 was obtained [2- (trifluoromethyl) phenyl] butanoate in toluene (217 mL) by the method of Example 2 ethyl acetate (362 mL) Te, after the addition of thionyl chloride 42.59 g (358 mmol), cooled to -10 ~ 5 ℃, was blown hydrochloric acid gas 52.21 g (1432 mmol), further concentrated sulfuric acid 17.83 g (179 mmol) was added, and the mixture was stirred with hot 15 ~ 30 ℃. After stirring for about 20 hours, added ethyl acetate (1086 mL), warmed to 30 ~ 40 ℃, after the addition of water (362 mL), and the layers were separated. after it separated organic layer water (362 mL) was added for liquid separation, and further 5w / v% was added for liquid separation aqueous sodium hydrogen carbonate solution (362 mL).
 Subsequently the organic layer was concentrated under reduced pressure, the mixture was concentrated under reduced pressure further added toluene (579 mL), was added toluene (72 mL), and cooled to 0 ~ 5 ℃. After stirring for about 2 hours, the precipitated crystals were filtered, and washed the crystals with toluene which was cooled to 0 ~ 5 ℃ (217 mL). The resulting wet goods crystals were dried under reduced pressure at 40 ℃, the title compound was obtained (97.55 g, 82.1% yield).
 1 H NMR (400 MHz, CDCl 3 ) delta: 1.38 (3H, t, J = 7.1 Hz), 2.11 (3H, s), 4.32 (2H, Q, J = 7.1 Hz), 7.39 (1H, D, J = 7.3 Hz), 7.50 ~ 7.62 (2H, m), 7.77 (1H, d, J = 8.0 Hz), 8.31 (1H, br).
(Example 4) 4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid ethyl
[Of 22]
 Example obtained by the production method of the three 2-chloro-4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylate 97.32 g (293 mmol) in ethanol (662 mL), tetrahydrofuran (117 mL), water (49 mL), sodium formate 25.91 g (381 mmol) and 5% palladium – carbon catalyst (water content 52.1%, 10.16 g) was added at room temperature, heated to 55 ~ 65 ℃ the mixture was stirred. After stirring for about 1 hour, cooled to 40 ℃ less, tetrahydrofuran (97 mL) and filter aid (KC- flock, Nippon Paper Industries) 4.87 g was added, the catalyst was filtered and the residue using ethanol (389 mL) was washed. The combined ethanol solution was used for washing the filtrate after concentration under reduced pressure, and with the addition of water (778 mL) was stirred for 0.5 hours at 20 ~ 30 ℃. The precipitated crystals were filtered, and washed the crystals with ethanol / water = 7/8 solution was mixed with (292 mL). The resulting wet goods crystals were dried under reduced pressure at 40 ℃, the title compound was obtained (86.23 g, 98.9% yield).
 1 H NMR (400 MHz, CDCl 3 ) delta: 1.35 (3H, t, J = 7.1 Hz), 2.18 (3H, s), 4.29 (2H, M), 7.40 ~ 7.61 (4H, M), 7.77 (1H, d, J = 7.9 Hz), 8.39 (1H, br).
(Example 5) (RS) -1- (2- hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid ethyl
[Of 23]
 N to the fourth embodiment of the manufacturing method by the resulting 4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylate 65.15 g (219 mmol), N- dimethylacetamide ( 261 mL), ethylene carbonate 28.95 g (328.7 mmol), 4- dimethylaminopyridine 2.68 g (21.9 mmol) were sequentially added at room temperature, and heated to 105 ~ 120 ℃, and the mixture was stirred. After stirring for about 10 hours, toluene was cooled to 20 ~ 30 ℃ (1303 mL), and the organic layer was extracted by adding water (326 mL). Subsequently, was washed by adding water (326 mL) to the organic layer (three times). The resulting organic layer was concentrated under reduced pressure, ethanol (652 mL) was added, and was further concentrated under reduced pressure, ethanol (130 mL) was added to obtain an ethanol solution of the title compound (326 mL).
 1 H NMR (400 MHz, CDCl 3 ) delta: 1.35 (3H, t, J = 7.1 Hz), 1.84 (1H, Broad singlet), 2.00 (3H, s), 3.63 ~ 3.77 (4H, M), 4.27 (2H , m), 7.35 ~ 7.79 (5H, m).
(Example 6) (RS) -1- (2- hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid
[Of 24]
 Obtained by the method of Example 5 (RS) -1- (2- hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid ethyl / ethanol (321 mL) solution in water (128.6 mL), was added at room temperature sodium hydroxide 21.4 g (519 mmol), and stirred with heating to 65 ~ 78 ℃. After stirring for about 6 hours, cooled to 20 ~ 30 ℃, after the addition of water (193 mL), and was adjusted to pH 5.5 ~ 6.5, while maintaining the 20 ~ 30 ℃ using 6 N hydrochloric acid. was added as seed crystals to the pH adjustment by a liquid (RS) -1- (2- hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid 6.4 mg , even I was added to water (193mL). Then cooled to 0 ~ 5 ℃, again, adjusted to pH 3 ~ 4 with concentrated hydrochloric acid and stirred for about 1 hour. Then, filtered crystals are precipitated, and washed the crystals with 20% ethanol water is cooled to 0 ~ 5 ℃ (93 mL). The resulting wet product crystals were dried under reduced pressure at 40 ℃, to give the title compound (64.32 g, 95.0% yield). 1 H NMR (400 MHz, DMSO-D 6 ) delta: 1.87 (3H, s), 3.38 ~ 3.68 (4H, M), 7.43 ~ 7.89 (5H, M).
(Example 7)
(S) -1- (2- hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid quinine salt 
(7-1) (S) -1- (2- hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid quinine salt
obtained by the method of Example 6 the (RS) -1- (2- hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid 50.00 g (160 mmol), N, N- dimethylacetamide (25 mL), ethyl acetate (85 mL) was added and dissolved at room temperature (solution 1).
 Quinine 31.05 g (96 mmol) in N, N- dimethylacetamide (25 mL), ethyl acetate (350 mL), was heated in water (15 mL) 65 ~ 70 ℃ was added, was added dropwise a solution 1. After about 1 hour stirring the mixture at 65 ~ 70 ℃, and slowly cooled to 0 ~ 5 ℃ (cooling rate standard: about 0.3 ℃ / min), and stirred at that temperature for about 0.5 hours. The crystals were filtered, 5 ℃ using ethyl acetate (100 mL) which was cooled to below are washed crystals, the resulting wet product crystals was obtained and dried under reduced pressure to give the title compound 43.66 g at 40 ℃ (Yield 42.9%). Furthermore, the diastereomeric excess of the obtained salt was 98.3% de. 1 H NMR (400 MHz, DMSO-D 6 ) delta: 1.30 ~ 2.20 (10H, M), 2.41 ~ 2.49 (2H, M), 2.85 ~ 3.49 (6H, M), 3.65 ~ 3.66 (1H, M), 3.88 (3H, s), 4.82 (1H, broad singlet), 4.92 ~ 5.00 (2H, m), 5.23 ~ 5.25 (1H, m), 5.60 (1H, br), 5.80 ~ 6.00 (1H, m), 7.36 ~ 7.92 (9H, M), 8.67 (1H, D, J = 4.6 Hz) (7-2) (S)-1-(2-hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3 diastereomeric excess of the carboxylic acid quinine salt HPLC measurements (% de)  that the title compound of about 10 mg was collected, and the 10 mL was diluted with 50v / v% aqueous acetonitrile me was used as a sample solution.
 Column: DAICEL CHIRALPAK IC-3 (4.6 mmI.D. × 250 mm, 3 μm)
mobile phase A: 0.02mol / L phosphorus vinegar buffer solution (pH 3)
mobile phase B: acetonitrile
solution sending of mobile phase: mobile phase A and I indicates the mixing ratio of mobile phase B in Table 1 below.
[Table 1]
  Detection: UV 237 nm
flow rate: about 0.8 mL / min
column temperature: 30 ℃ constant temperature in the vicinity of
measuring time: about 20 min
Injection volume: 5 μL
diastereomeric excess (% de), the title compound (retention time about 12 min), was calculated by the following equation using a peak area ratio of R-isomer (retention time of about 13 min).
% De = {[(the title compound (S body) peak area ratio) – (R body peak area ratio)] ÷ [(the title compound (S body) peak area ratio) + (R body peak area ratio)]} × 100
(Example 8)
(S) -1- (2- hydroxyethyl) -4-methyl -N- [4- (methylsulfonyl) phenyl] -5- [2- (trifluoromethyl) phenyl] -1H- pyrrole 3-carboxamide (Compound (A)) 
(8-1) (S)-1-(2-hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole -3 – carboxylic acid
obtained by the method of Example 7 (S) -1- (2- hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxylic acid (8α, 9R) -6′- methoxycinnamate Conan-9-ol 40.00 g (63 mmol) in ethyl acetate (400 mL), was added 2N aqueous hydrochloric acid (100 mL) was stirred at room temperature and separated . The resulting organic layer was concentrated under reduced pressure (120 mL), and added ethyl acetate (200 mL), and further concentrated under reduced pressure to obtain a solution containing the title compound (120 mL).
(8-2) N – {[4- (methylsulfonyl) phenyl] amino} oxamic acid 2 – ((S) -3- methyl-4 – {[4- (methylsulfonyl) phenyl] carbamoyl} -2- [ 2- (trifluoromethyl) phenyl] -1H- pyrrol-1-yl) ethyl
ethyl acetate (240 mL), was mixed tetrahydrofuran (80 mL) and oxalyl chloride 20.72 g (163 mmol), and cooled to 10 ~ 15 ℃ was. Then the resulting solution was added while keeping the 10 ~ 15 ℃ Example (8-1) and stirred for about 1 hour by heating to 15 ~ 20 ℃. After stirring, acetonitrile (120 mL) and pyridine 2.46 g (31 mmol) was added and the reaction mixture was concentrated under reduced pressure (120 mL), acetonitrile (200 mL) was added and further concentrated under reduced pressure (120 mL).
 After completion concentration under reduced pressure, acetonitrile (200 mL) was added and cooled to 10 ~ 15 ℃ (reaction 1).
 Acetonitrile (240mL), pyridine 12.39 g (157 mmol), 4- were successively added (methylsulfonyl) aniline 26.85 g (157 mmol), the reaction solution 1 was added while maintaining the 10 ~ 15 ℃, the 20 ~ 25 ℃ and the mixture was stirred and heated to about 1 hour.
 The resulting reaction solution in acetonitrile (40 mL), 2 N hydrochloric acid water (120 mL), was added sodium chloride (10.0 g) was stirred, and the layers were separated. Again, 2N aqueous hydrochloric acid to the organic layer (120 mL), was added sodium chloride (10.0 g) was stirred, and the layers were separated. After filtering the resulting organic layer was concentrated under reduced pressure (400 mL). Water (360 mL) was added to the concentrated liquid, after about 1 hour stirring, the crystals were filtered, washed with 50v / v% aqueous acetonitrile (120 mL), wet product of the title compound (undried product, 62.02 g) and obtained. 1 H NMR (500 MHz, DMSO-D 6 ) delta: 1.94 (s, 3H), 3.19 (s, 3H), 3.20 (s, 3H), 3.81 (t, 1H), 4.12 (t, 1H), 4.45 ( t, 2H, J = 5.81 Hz), 7.62 (t, 1H, J = 4.39 Hz), 7.74 (t, 2H, J = 3.68 Hz), 7.86 (dd, 3H), 7.92 (dd, 3H, J = 6.94 , 2.13 Hz), 7.97 (DD, 2H, J = 6.80, 1.98 Hz), 8.02 (DD, 2H), 10.03 (s, 1H), 11.19 (s, 1H) 
(8-3) (S)-1- (2-hydroxyethyl) -4-methyl -N- [4- (methylsulfonyl) phenyl] -5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxamide (Compound (A))  ( the resulting wet product crystals 8-2), t- butyl methyl ether (200 mL), acetonitrile (40 mL), 48w / w potassium hydroxide aqueous solution (16 g) and water (200 mL) was added, I was stirred for about 2 hours at 25 ~ 35 ℃. After stirring, and the mixture is separated, the resulting organic layer was concentrated under reduced pressure (120 mL), ethanol (240 mL) was added and further concentrated under reduced pressure (120 mL). After completion concentration under reduced pressure, ethanol (36 mL), and heated in water (12 mL) was added 35 ~ 45 ℃, while maintaining the 35 ~ 45 ℃ was added dropwise water (280 mL), and was crystallized crystals. After cooling the crystal exudates to room temperature, I was filtered crystal. Then washed with crystals 30v / v% aqueous ethanol solution (80 mL), where it was dried under reduced pressure at 40 ℃, the title compound was obtained in crystalline (26.26 g, 89.7% yield). Moreover, the enantiomers of the resulting crystals was 0.3%. 
1 H NMR (400 MHz, CDCl 3 ) delta: 1.74 (1H, Broad singlet), 2.08 (3H, s), 3.04 (3H, s), 3.63 ~ 3.80 (4H, M), 7.36 (1H, D, J = 7.2 Hz), 7.48 (1H, s), 7.58 ~ 7.67 (2H, M), 7.77 ~ 7.90 (6H, M). 
(8-4) (S)-1-(2-hydroxyethyl) -4-methyl -N- [4- (methylsulfonyl) phenyl] -5- [2- (trifluoromethyl) phenyl] -1H- pyrrole -3- HPLC method for measuring the amount enantiomer carboxamide (%)  and collected the title compound of about 10 mg is, what was the 10 mL was diluted with 50v / v% aqueous acetonitrile to obtain a sample solution.
see
(Example 12) (S) -1- (2- hydroxyethyl) -4-methyl -N- [4- (methylsulfonyl) phenyl] -5- [2- (trifluoromethyl) phenyl] -1H- pyrrole 3-carboxamide (Compound (A)) Preparation of 2 
(12-1) (S)-1-(2-hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H – pyrrole-3-carboxylic acid
obtained by the method of Example 7 (S) -1- (2- hydroxyethyl) -4-methyl-5- [2- (trifluoromethyl) phenyl] -1H- pyrrole 3-carboxylic acid (8α, 9R) -6′- methoxycinnamate Conan-9-ol 10.00 g (16 mmol) in t- butyl methyl ether (90 mL), water (10 mL) 36w / w% aqueous hydrochloric acid ( 5 mL) was added and stirring at room temperature and separated. The resulting organic layer was concentrated under reduced pressure (30 mL), was added ethyl acetate (50 mL), and further concentrated under reduced pressure to obtain a solution containing the title compound (30 mL). 
(12-2) N – {[4- (methylsulfonyl) phenyl] amino} oxamic acid 2 – ((S) -3- methyl-4 – {[4- (methylsulfonyl) phenyl] carbamoyl} -2- [ 2- (trifluoromethyl) phenyl] -1H- pyrrol-1-yl) ethyl
ethyl acetate (50 mL), was mixed with tetrahydrofuran (20 mL) and oxalyl chloride 5.18 g (41 mmol), and cooled to 0 ~ 5 ℃ was.Then the resulting solution was added in Examples while maintaining the 0 ~ 5 ℃ (12-1), and the mixture was stirred for 6 hours at 0 ~ 10 ℃. After stirring, acetonitrile (30 mL) and pyridine 0.62 g (8 mmol) was added and the reaction mixture was concentrated under reduced pressure (30 mL), acetonitrile (50 mL) was added, and further concentrated under reduced pressure (30 mL).
 After concentration under reduced pressure end, is added acetonitrile (10 mL) and oxalyl chloride 0.10 g (1 mmol), and cooled to 0 ~ 5 ℃ (reaction 1).
 Acetonitrile (30mL), pyridine 3.15 g (40 mmol), 4- were successively added (methylsulfonyl) aniline 6.71 g (39 mmol), the reaction solution 1 was added while maintaining the 10 ~ 15 ℃, the 20 ~ 25 ℃ and the mixture was stirred and heated to about 1 hour.
 Insolubles from the resulting reaction solution was filtered, washed with acetonitrile (10 mL), and stirred for about 2 hours the addition of water (15 mL), followed by dropwise addition of water (75 mL) over about 1 hour . After about 1 hour stirring the suspension was filtered crystals were washed with 50v / v% aqueous acetonitrile (20 mL), wet product of the title compound (undried product, 15.78 g) to give a. 1 H NMR (500 MHz, DMSO-D 6 ) delta: 1.94 (s, 3H), 3.19 (s, 3H), 3.20 (s, 3H), 3.81 (t, 1H), 4.12 (t, 1H), 4.45 ( t, 2H, J = 5.81 Hz), 7.62 (t, 1H, J = 4.39 Hz), 7.74 (t, 2H, J = 3.68 Hz), 7.86 (dd, 3H), 7.92 (dd, 3H, J = 6.94 , 2.13 Hz), 7.97 (DD, 2H, J = 6.80, 1.98 Hz), 8.02 (DD, 2H), 10.03 (s, 1H), 11.19 (s, 1H) 
(12-3) (S)-1- (2-hydroxyethyl) -4-methyl -N- [4- (methylsulfonyl) phenyl] -5- [2- (trifluoromethyl) phenyl] -1H- pyrrole-3-carboxamide (Compound (A))  ( the resulting wet product crystals 12-2), t- butyl methyl ether (50 mL), acetonitrile (10 mL), 48w / w potassium hydroxide aqueous solution (4 g) and water (50 mL) was added, 15 I was about 2 hours of stirring at ~ 25 ℃. After stirring, and the mixture is separated, the resulting organic layer was concentrated under reduced pressure (30 mL), was added ethanol (60 mL), was further concentrated under reduced pressure (30 mL). After completion concentration under reduced pressure, ethanol (14 mL), after addition of water (20 mL), was added a seed crystal, and was crystallized crystals. After dropwise over about 1 hour water (50 mL), and about 1 hour stirring, and crystals were filtered off. Then washed with crystals 30v / v% aqueous ethanol solution (10 mL), where it was dried under reduced pressure at 40 ℃, the title compound was obtained in crystal (6.36 g, 87.0% yield). Moreover, the enantiomers of the resulting crystals was 0.05%. Enantiomers amount, I was measured by the method of (Example 8-4). 1 H NMR (400 MHz, CDCl 3 ) delta: 1.74 (1H, Broad singlet), 2.08 (3H, s), 3.04 (3H, s), 3.63 ~ 3.80 (4H, M), 7.36 (1H, D, J = 7.2 Hz), 7.48 (1H, s), 7.58 ~ 7.67 (2H, m), 7.77 ~ 7.90 (6H, m).
………………………………………………

Patent literature

Patent Document 1: International Publication WO2006 / 012642 (US Publication US2008-0234270)
Patent Document 2: International Publication WO2008 / 056907 (US Publication US2010-0093826)
Patent Document 3: Pat. No. 2,082,519 JP (US Patent No. 5,616,599 JP)
Patent Document 4: Pat. No. 1,401,088 JP (US Pat. No. 4,572,909)
Patent Document 5: US Pat. No. 3,025,292
Angiotensin II receptor 桔抗 agent
Angiotensin II receptor 桔抗 agent used as the component (A), olmesartan medoxomil, olmesartan cilexetil, losartan, candesartan cilexetil, valsartan, biphenyl tetrazole compounds such as irbesartan, biphenyl carboxylic acid compounds such as telmisartan, eprosartan, agile Sultan, and the like, preferably, a biphenyl tetrazole compound, more preferably, olmesartan medoxomil, is losartan, candesartan cilexetil, valsartan or irbesartan, particularly preferred are olmesartan medoxomil, losartan or candesartan cilexetil, Most preferably, it is olmesartan medoxomil.
 Olmesartan medoxomil, JP-A-5-78328, US Patent No. 5,616,599
is described in Japanese or the like, its chemical name is (5-methyl-2-oxo-1,3-dioxolen-4-yl ) methyl 4- (1-hydroxy-1-methylethyl) -2-propyl-1 – in [2 ‘(1H- tetrazol-5-yl) biphenyl-4-ylmethyl] imidazole-5-carboxylate, Yes, olmesartan medoxomil of the present application includes its pharmacologically acceptable salt.
Olmesartan.pngOLMESARTAN
 Losartan (DUP-753) is, JP 63-23868, is described in US Patent No. 5,138,069 JP like, and its chemical name is 2-butyl-4-chloro-1- [2 ‘ – The (1H- tetrazol-5-yl) biphenyl-4-ylmethyl] -1H- is imidazol-5-methanol, application of losartan includes its pharmacologically acceptable salt (losartan potassium salt, etc.).
Skeletal formula
 LOSARTAN
 Candesartan cilexetil, JP-A-4-364171, EP-459136 JP, is described in US Patent No. 5,354,766 JP like, and its chemical name is 1- (cyclohexyloxycarbonyloxy) ethyl-2 ethoxy-1- [2 ‘one (1H- tetrazol-5-yl) -4-Bife~eniru ylmethyl] -1H- benzimidazole-7-carboxylate is a salt application of candesartan cilexetil, which is a pharmacologically acceptable encompasses.
 Valsartan (CGP-48933), the JP-A-4-159718, are described in EP-433983 JP-like, and its chemical name, (S) -N- valeryl -N- [2 ‘- (1H- tetrazol – It is a 5-yl) biphenyl-4-ylmethyl) valine, valsartan of the present application includes its pharmacologically acceptable ester or a pharmacologically acceptable salt thereof.
 Irbesartan (SR-47436), the Japanese Patent Publication No. Hei 4-506222, is described in JP WO91-14679 publication, etc., its chemical name, 2-N–butyl-4-spiro cyclopentane-1- [2′ The (tetrazol-5-yl) biphenyl-4-ylmethyl] -2-imidazoline-5-one, irbesartan of the present application includes its pharmacologically acceptable salts.
 Eprosartan (SKB-108566) is described in US Patent No. 5,185,351 JP etc., the chemical name, 3- [1- (4-carboxyphenyl-methyl) -2-n- butyl – imidazol-5-yl] The 2-thienyl – methyl-2-propenoic acid, present in eprosartan, the carboxylic acid derivatives, pharmacologically acceptable ester or a pharmacologically acceptable salt of a carboxylic acid derivative (eprosartan mesylate, encompasses etc.).
 Telmisartan (BIBR-277) is described in US Patent No. 5,591,762 JP like, and its chemical name is 4 ‘- [[4 Mechiru 6- (1-methyl-2-benzimidazolyl) -2 – is a propyl-1-benzimidazolyl] methyl] -2-biphenylcarboxylic acid, telmisartan of the present application includes its carboxylic acid derivative, a pharmacologically acceptable ester or a pharmacologically acceptable salt thereof of carboxylic acid derivatives .
 Agile Sultan, is described in Patent Publication No. 05-271228 flat JP, US Patent No. 5,243,054 JP like, and its chemical name is 2-ethoxy-1 {[2 ‘- (5-oxo-4,5-dihydro 1,2,4-oxadiazole-3-yl) biphenyl-4-yl] methyl} -1H- benzo [d] imidazole-7-carboxylic acid (2-Ethoxy-1 {[2 ‘- (5- oxo-4,5-dihydro-1,2,4-oxadiazol-3-yl) biphenyl-4-yl] is a methyl} -1H-benzo [d] imidazole-7-carboxylic acid).