AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

Roxadustat, ASP 1517, FG 4592

 Phase 3 drug, Uncategorized  Comments Off on Roxadustat, ASP 1517, FG 4592
Jun 212016
 

STR1

 

ROXADUSTAT

ASP1517; ASP 1517; ASP-1517; FG-4592; FG 4592; FG4592; Roxadustat.

CAS 808118-40-3
Chemical Formula: C19H16N2O5
Exact Mass: 352.10592

Fibrogen, Inc.

THERAPEUTIC CLAIM, Treatment of anemia

Roxadustat nonproprietary drug name

CHEMICAL NAMES

(4-hydroxy-1-methyl-7-phenoxyisoquinoline-3-carbonyl)glycine

1. Glycine, N-[(4-hydroxy-1-methyl-7-phenoxy-3-isoquinolinyl)carbonyl]-

2. N-[(4-hydroxy-1-methyl-7-phenoxyisoquinolin-3-yl)carbonyl]glycine

MF C19H16N2O5
MW  352.3
SPONSOR FibroGen
CODE FG-4592; ASP1517
CAS 808118-40-3
WHO NUMBER 9717

Roxadustat, also known as ASP1517 and FG-4592, is an HIF α prolyl hydroxylase inhibitor in a cell-free assay. It stabilizes HIF-2 and induces EPO production and stimulates erythropoiesis. Roxadustat transiently and moderately increased endogenous erythropoietin and reduced hepcidin

FG-4592 (also known as ASP1517), 2-(4-hydroxy-1-methyl-7-phenoxyisoquinoline-3-carboxamido)acetic acid,
 is a potent small molecule inhibitor of hypoxia-inducible factor prolyl hydroxylase (HIF-PH),
an enzyme up-regulating the expression of endogenous human erythropoietin (Epo).
It is currently being investigated as an oral treatment for anemia associated with chronic kidney disease (CKD).
Unlike other anemia treating agents, erythropoiesis-stimulating agents (ESAs),
FG-4592 inhibits HIF, through a distinctive mechanism, by stabilization of HIF. According to previous studies,
FG-4592 is capable of correcting and maintaining hemoglobin levels in CKD patients not
receiving dialysis and in patients of end-stage renal disease
who receives dialysis but do not need intravenous iron supplement.
Reference
1. Luis Borges. Different modalities of erythropoiesis stimulating agents.
 Port J Nephrol Hypert 2010; 24(2): 137-145
2. “FibroGen and Astellas announce initiation of phase 3 trial of FG-4592/ASP1517 for treatment 
of anemia of chronic kidney disease” Fibrogen Press Release. Dec 11 2012
3. “FibroGen announces initiation of phase 2b studies of FG-4592, 
an oral HIF prolyl hydroxylase inhibitor, for treatment of anemia”
  • Originator FibroGen
  • Developer Astellas Pharma; AstraZeneca; FibroGen
  • Class Amides; Antianaemics; Carboxylic acids; Isoquinolines; Small molecules
  • Mechanism of Action Basic helix loop helix transcription factor modulators; Hypoxia-inducible factor-proline dioxygenase inhibitors
  • Phase III Anaemia
  • Discontinued Sickle cell anaemia

Most Recent Events

  • 09 Jun 2016 Phase-III clinical trials in Anaemia in Japan (PO)
  • 20 May 2016 In collaboration with FibroGen, Astellas Pharma plans a phase III trial for Anaemia (In chronic kidney disease patients undergoing peritoneal dialysis) in Japan (PO) (NCT02780726)
  • 19 May 2016 In collaboration with FibroGen, Astellas Pharma plans a phase III trial for Anaemia (In erythropoiesis stimulating agent-naive, chronic kidney disease patients undergoing haemodialysis) in Japan (PO) (NCT02780141)

 

 

 

Roxadustat (FG-4592) is a novel new-generation oral hypoxia-induciblefactor (HIF) prolyl hydroxylase inhibitor (PHI) for the treatment of ane-mia in patients with chronic kidney disease (CKD). HIF is a cytosolic tran-scription factor that induces the natural physiological response to lowoxygen conditions, by stimulating erythropoiesis and other protectivepathways. Roxadustat has been shown to stabilize HIF and induce ery-thropoiesis. Consequently, it corrects anemia and maintains hemoglo-bin levels without the need for intravenous iron supplementation in CKDpatients not yet receiving dialysis and in end-stage renal disease pa-tients receiving dialysis. There are many concerns about the use of ery-thropoiesis-stimulating agents (ESA) to treat anemia as they causesupra-physiologic circulating erythropoietin (EPO) levels and are asso-ciated with adverse cardiovascular effects and mortality. Available clin-ical data show that modest and transient increases of endogenous EPOinduced by HIF-PHI (10- to 40-fold lower than ESA levels) are sufficientto mediate erythropoiesis in CKD patients. Evidence suggests that rox-adustat is well tolerated and, to date, no increased risk of cardiovascu-lar events has been found. This suggests that roxadustat provides adistinct pharmacological and clinical profile that may provide a saferand more convenient treatment of CKD anemia

 

FG-4592 is a new-generation hypoxia-inducible factor prolyl hydroxylase inhibitor in early clinical trials at FibroGen for the oral treatment of iron deficiency anemia and renal failure anemia. Preclinical studies are ongoing for the treatment of sickle cell anemia.

The investigational therapy is designed to restore balance to the body’s natural process of erythropoiesis through mechanisms including: natural EPO production, suppression of the effects of inflammation, downregulation of the iron sequestration hormone hepcidin, and an upregulation of other iron genes, ensuring efficient mobilization and utilization of the body’s own iron stores. In April 2006, FG-4592 was licensed to Astellas Pharma by originator FibroGen in Asia, Europe and South Africa for the treatment of anemia. FibroGen retains rights in the rest of the world. In 2007, the FDA put the trial on clinical hold due to one case of death by fulminant hepatitis during a phase II clinical trial for patients with anemia associated with chronic kidney disease and not requiring dialysis. However, in 2008, the FDA informed the company that clinical trials could be resumed. Phase II/III clinical trials for this indication resumed in 2012. In 2013, the compound was licensed to AstraZeneca by FibroGen for development and marketing in US, CN and all major markets excluding JP, Europe, the Commonwealth of Independent States, the Middle East and South Africa, for the treatment of anemia associated with chronic kidney disease (CKD) and end-stage renal disease (ESRD).
PATENTS
WO 2004108681
WO 2008042800
WO 2009058403
WO 2009075822
WO 2009075824
WO 2012037212
WO 2013013609
WO 2013070908

 

STR1

PATENT

CN 104892509

MACHINE TRANSLATED

Connaught orlistat (Roxadustat) by the US company Phibro root (FibroGen) R & D, Astellas AstraZeneca and licensed by a hypoxia-inducible factor (HIF) prolyl hydroxylase small molecule inhibitors, codenamed FG-4592.As a first new oral drug, FG-4592 is currently in Phase III clinical testing stage, for the treatment of chronic kidney disease and end-stage renal disease related anemia. Because the drug does not have a standard Chinese translation, so the applicant where it is transliterated as “Connaught Secretary him.”

Connaught orlistat (Roxadustat, I) the chemical name: N_ [(4- hydroxy-1-methyl-7-phenoxy-3-isoquinolinyl) carbonyl] glycine, its structural formula is:

 

Figure CN104892509AD00031

The original research company’s international patent W02004108681 Division provides a promise he was prepared from the intermediate and intermediate Connaught Secretary for his synthetic route:

 

Figure CN104892509AD00032

 Zhejiang Beida company’s international patent W02013013609 preparation and acylation of core intermediate was further optimized synthesis route is:

 

Figure CN104892509AD00041

n PhO. eight XOOH

 

 original research company’s international patent W02014014834 and W02014014835 also provides another synthetic route he Connaught Secretary prepared:

 

Figure CN104892509AD00042

Analysis of the above synthetic route, although he continued to Connaught Division to improve and optimize the synthesis, but its essence rings manner that different form quinoline ring is basically the same mother. Especially methyl isoquinoline replaced either by way of introducing the Suzuki reaction catalyzed by a noble metal element, either through amine reduction achieved. Moreover, the above reaction scheme revelation raw materials are readily available, many times during the reaction need to be protected and then deprotected. Clearly, the preparation process is relatively complicated, high cost, industrial production has brought some difficulties.

Figure CN104892509AD00052

Example One:

tyrosine was added to the reaction flask and dried (18. lg, 0.1 mmol) and methanol 250mL, cooling to ice bath 0_5 ° C, was added dropwise over 1 hour a percentage by weight of 98% concentrated sulfuric acid 10g. Drops Albert, heating to reflux. The reaction was stirred for 16-20 hours, TLC the reaction was complete. Concentrated under atmosphere pressure, the residue was added water 100mL, using 10% by weight sodium hydroxide to adjust the pH to 6. 5-7.0, precipitated solid was filtered, washed with methanol and water chloro cake (I: 1) and dried in vacuo tyrosine methyl ester as a white solid (11) 15.38, yield 78.5% out 1–] \ ^ 111/2: 196 [] \ 1 + 1] +!.

Example Two:

[0041] a nitrogen atmosphere and ice bath, was added to the reaction flask tyrosine methyl ester (II) (9. 8g, 50mmol), potassium methoxide (3. 5g, 50mmol) and methanol 50mL, until no gas generation after, was heated to reflux, the reaction was stirred for 2 hours. Concentrated under atmosphere pressure to remove the solvent, the residue was added dimethylsulfoxide 25mL, freshly prepared copper powder (0.2g, 3. Lmmol), was slowly warmed to 150-155 ° C, for about half an hour later, a solution of bromobenzene ( 7. 9g, 50mmol), continue to heat up to 170-175 ° C, the reaction was stirred for 3 hours, TLC detection of the end of the reaction. Was cooled to 60 ° C, and methanol was added to keep micro-boiling, filtered while hot, the filter cake washed three times with hot ethanol, and the combined organic phases, was cooled to square ° C, filtered, and dried in vacuo to give a white solid of 2-amino-3- ( 4-phenoxyphenyl) propanoate (111) 8 11.5, yield 84.9% as 1 -] \ ^ 111/2:! 272 [] \ 1 + 1] +.

 Example Three:

 in the reaction flask was added 2-amino-3- (4-phenoxyphenyl) propionic acid methyl ester (III) (10. 8g, 40mmol), 40% by weight acetaldehyde (20g, 0. 2mol ) and the percentage by weight of 35% concentrated hydrochloric acid 50mL, refluxed for 1 hour. Continue 40% by weight was added acetaldehyde (10g, 0.1mol), and the percentage by weight of 35% concentrated hydrochloric acid 25mL, and then the reaction was refluxed for 3-5 hours. Was cooled to 4-7 ° C, ethyl acetate was added, and extracted layers were separated. The aqueous layer was adjusted with sodium hydroxide solution to pH 11-12, extracted three times with ethyl acetate. The combined organic phase was dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a white solid of 1-methyl-3-carboxylate -7- phenoxy-1,2,3,4-tetrahydroisoquinoline (IV) 8 4g, 70.7% yield; Mass spectrum (EI): EI-MS m / z: 298 [M + H] + .

 Example Four:

Under ice bath, the reaction flask was added methyl 3-carboxylate I- -7- phenoxy-1,2, 3,4-tetrahydro-isoquinoline (IV) (5. 9g, 20mmol) and dichloromethane 100mL, 0 ° C and under stirring added potassium carbonate (13. 8g, 0. lmol), p-toluenesulfonyl chloride (11. 4g, 60mmol), the addition was completed, the ice bath was removed and stirred at room temperature 3 hour. Water was added 30mL, after stirring standing layer, the organic phase was washed with dilute hydrochloric acid, water and saturated brine, and concentrated, the resulting product was added a 30% by weight sodium hydroxide solution (8. 0g, 60mmol) and dimethyl sulfoxide 60mL, gradually warming to 120-130 ° C, the reaction was stirred for 2-4 hours to complete the reaction by TLC. Cooled to room temperature, water was added lOOmL, extracted three times with ethyl acetate, the combined organic phase was successively washed with water and saturated brine, dried over anhydrous magnesium sulfate, and concentrated, the resulting oil was treated with ethyl acetate and n-hexane (1: 3) recrystallization, vacuum dried to give an off-white solid 1-methyl-3-carboxylate 7-phenoxyheptanoic isoquinoline (V) 5. 25g, yield 89. 6%; EI-MS m / z: 294 [M + H] VH NMR (DMS0-d6) δ 2. 85 (s, 3H), 3 · 97 (s, 3H), 7 · 16-7. 24 (m, 3H), 7 · 49-7. 60 (m, 4Η), 8 · 35 (d, J = 9 · 0,1Η), 8 · 94 (s, 1Η).

Example five:

[0047] added 1-methyl-3-carboxylic acid methyl ester 7-phenoxyheptanoic isoquinoline (V) (2. 93g, IOmmol) and glacial acetic acid 50mL reaction flask, stirring solution of 30% by weight hydrogen peroxide 5mL, warmed to 60-70 ° C, was slowly added dropwise within 10 hours the percentage by weight of a mixture of 30% hydrogen peroxide 2mL and 12mL of glacial acetic acid, a dropping was completed, the reaction was continued for 20-24 hours. Concentrated under reduced pressure, ethanol was added, distillation is continued to be divisible remaining glacial acetic acid. The residue was dissolved with dichloromethane, washed with 5% by weight of sodium bicarbonate, the organic phase was separated, dried over anhydrous sodium sulfate. Filtered and the resulting solution was added p-toluenesulfonyl chloride (3. 8g, 20mmol), was heated to reflux, the reaction was stirred for 3-4 hours, TLC detection completion of the reaction. The solvent was distilled off under reduced pressure, cooled to room temperature, methanol was added, the precipitated solid, cooled to square ° C, allowed to stand overnight. Filtered, the filter cake washed twice with cold methanol and vacuum dried to give an off-white solid 1- methyl-3-methyl-4-hydroxy-phenoxy-isoquinoline -7- (VI) I. 86g, yield 60.2 %; EI-MS m / z:.. 310 [M + H] +, 1H NMR (DMS0-d6) δ 2.90 (s, 3H), 4.05 (s, 3H), 7 17-7 26 (m, 3H ), 7. 49-7. 61 (m, 4H), 8. 38 (d, J = 9. 0,1H), 11. 7 (s, 1H) 〇

 Example VI:

 in the reaction flask with magnetic stirring and pressure to join I- methyl-3-methyl-4-hydroxy-7-phenoxyheptanoate isoquinoline (VI) (1.55g, 5mmol), glycine (I. 13g, 15mmol) and sodium methoxide (3. 25g, 6mmol) in methanol (30mL).Sealed, slowly heated to 120 ° C, the reaction was stirred for 8-10 hours to complete the reaction by TLC. Cooled to room temperature, solid precipitated. Filtration, and the resulting solid was recrystallized from methanol, acetone and then beating the resulting solid was dried under vacuum to give a white solid Connaught orlistat 1.40g, yield 79.5%;

EI-MS m / z: 353 [M + H] +,

1H NMR (DMS0-d6) S2.72 (s, 3H), 3 · 99 (d, J = 6 · 0, 2H), 7 · 18-7. 28 (m, 3H), 7 · 49-7. 63 (m, 4H), 8 · 31 (d, J = 8 · 8,1H), 9 · 08 (s, lH), 13.41 (brs, lH).

PATENT

WO 2014014835

Example 10. Preparation of Compound A

a) 5-Phenoxyphthalide

Figure imgf000056_0001

[0200] A reactor was charged with DMF (68 Kg), and stirring was initiated. The reactor was then charged with phenol (51 Kg), acetylacetone (8 Kg), 5-bromophthalide (85 Kg), copper bromide (9 Kg), and potassium carbonate (77 Kg). The mixture was heated above 85 °C and maintained until reaction completion and then cooled. Water was added. Solid was filtered and washed with water. Solid was dissolved in dichloromethane, and washed with aqueous HCl and then with water. Solvent was removed under pressure and methanol was added. The mixture was stirred and filtered. Solid was washed with methanol and dried in an oven giving 5- phenoxyphthalide (Yield: 72%, HPLC: 99.6%). b) 2-Chloromethyl-4-phenoxybenzoic acid methyl ester

Figure imgf000056_0002

[0201] A reactor was charged with toluene (24 Kg), and stirring was initiated. The reactor was then charged with 5-phenoxyphthalide (56 Kg), thionyl chloride (41 Kg), trimethyl borate (1

Kg), dichlorotriphenylphosphorane (2.5 Kg), and potassium carbonate (77 Kg). The mixture was heated to reflux until reaction completion and solvent was removed leaving 2-chloromethyl-4- phenoxybenzoyl chloride. Methanol was charged and the mixture was heated above 50 °C until reaction completion. Solvent was removed and replaced with DMF. This solution of the product methyl 2-chloromethyl-4-phenoxybenzoic acid methyl ester in DMF was used directly in the next step (HPLC: 85%). c) 4-Hydroxy-7-phenoxyisoquinoline-3-carboxylic acid methyl ester (la)

Figure imgf000057_0001

[0202] A reactor was charged with a solution of 2-chloromethyl-4-phenoxybenzoic acid methyl ester (~68 Kg) in DMF, and stirring was initiated. The reactor was then charged with p- toluenesulfonylglycine methyl ester (66 Kg), potassium carbonate (60 Kg), and sodium iodide (4 Kg). The mixture was heated to at least 50 °C until reaction completion. The mixture was cooled. Sodium methoxide in methanol was charged and the mixture was stirred until reaction completion. Acetic acid and water were added, and the mixture was stirred, filtered and washed with water. Solid was purified by acetone trituration and dried in an oven giving la (Yield from step b): 58%; HPLC: 99.4%). 1H NMR (200 MHz, DMSO-d6) δ 11.60 (s, 1 H), 8.74 (s, 1H),

8.32 (d, J = 9.0 Hz, 1 H), 7.60 (dd, J = 2.3 & 9.0 Hz, 1H), 7.49 (m, 3 H), 7.24 (m, 3 H), 3.96 (s, 3 H); MS-(+)-ion M+l = 296.09 d) Methyl l-((dimethylamino)methyl)-4-hydroxy-7-phenoxyisoquinoline-3-carboxylate

(lb)

Figure imgf000057_0002

[0203] A flask was charged with la (29.5 g) and acetic acid (44.3 g ± 5%), and then stirred. Bis-dimethylaminomethane (12.8 g ± 2%) was slowly added. The mixture was heated to 55 ± 5 °C and maintained until reaction completion. The reaction product was evaluated by MS, HPLC and 1H NMR. 1H NMR (200 MHz, DMSO-d6) δ 11.7 (s, 1 H), 8.38 (d, J = 9.0 Hz, 1 H), 7.61 (dd, J = 9.0, 2.7 Hz, 1 H), 7.49 (m, 3 H), 7.21 (m, 3 H), 5.34 (s, 2 H), 3.97 (s, 3 H), 1.98 (s, 3 H); MS-(+)-ion M+l = 368.12. e) Methyl l-((acetoxy)methyl)-4-hydroxy-7-phenoxyisoquinoline-3-carboxylate (lc)

Figure imgf000058_0001

[0204] The solution of lb from a) above was cooled below 25 °C, at which time acetic anhydride (28.6 g ± 3.5 %) was added to maintain temperature below 50 °C. The resulting mixture was heated to 100 ± 5 °C until reaction completion.

[0205] The solution of lc and Id from above was cooled to less than 65 ± 5 °C. Water (250 mL) was slowly added. The mixture was then cooled to below 20 ± 5 °C and filtered. The wet cake was washed with water (3 x 50 mL) and added to a new flask. Dichloromethane (90 mL) and water (30 mL) were added, and the resulting mixture was stirred. The dichloromethane layer was separated and evaluated by HPLC.

[0206] The organic layer was added to a flask and cooled 5 ± 5 °C. Morpholine was added and the mixture was stirred until reaction completion. Solvent was replaced with acetone/methanol mixture. After cooling, compound lc precipitated and was filtered, washed and dried in an oven (Yield: 81%, HPLC: >99.7%). 1H NMR (200 MHz, DMSO-d6) δ 11.6 (S, 1 H), 8.31 (d, J = 9.0 Hz, 1 H), 7.87 (d, J = 2.3 Hz, 1 H), 7.49 (m, 3 H), 7.24 (m, 3 H), 3.95 (s, 3 H), 3.68 (s, 2H), 2.08 (s, 6 H); MS-(+)-ion M+l = 357.17. f) Methyl 4-hydroxy-l-methyl-7-phenoxyisoquinoline-3-carboxylate (le)

Figure imgf000058_0002

[0207] A reactor was charged with lc (16.0 g), Pd/C (2.08 g), anhydrous Na2C03 (2.56 g) and ethyl acetate (120 mL). The flask was vacuum-purged with nitrogen (3X) and vacuum-purged with hydrogen (3X). The flask was then pressurized with hydrogen and stirred at about 60 °C until completion of reaction. The flask was cooled to 20-25 °C, the pressure released to ambient, the head space purged with nitrogen three times and mixture was filtered. The filtrate was concentrated. Methanol was added. The mixture was stirred and then cooled. Product precipitated and was filtered and dried in an oven (Yield: 90%, HPLC: 99.7%). g) [(4-Hydroxy-l-methyl-7-phenoxy-isoquinoline-3-carbonyl)-amino]-acetic acid

(Compound A)

Figure imgf000059_0001

[0208] A pressure flask was charged with le (30.92 g), glycine (22.52 g), methanol (155 mL), sodium methoxide solution (64.81 g) and sealed (as an alternative, sodium glycinate was used in place of glycine and sodium methoxide). The reaction was heated to about 110 °C until reaction was complete. The mixture was cooled, filtered, washed with methanol, dried under vacuum, dissolved in water and washed with ethyl acetate. The ethyl acetate was removed and to the resulting aqueous layer an acetic acid (18.0 g) solution was added. The suspension was stirred at room temperature, filtered, and the solid washed with water (3 x 30 mL), cold acetone (5-10 °C, 2 x 20 mL), and dried under vacuum to obtain Compound A (Yield: 86.1%, HPLC: 99.8%). Example 11. Biological Testing

[0209] The solid forms provided herein can be used for inhibiting HIF hydroxylase activity, thereby increasing the stability and/or activity of hypoxia inducible factor (HIF), and can be used to treat and prevent HIF-associated conditions and disorders (see, e.g., U.S. Patent No. 7,323,475, U.S. Patent Application Publication No. 2007/0004627, U.S. Patent Application Publication No. 2006/0276477, and U.S. Patent Application Publication No. 2007/0259960, incorporated by reference herein).

SYNTHESIS……..

http://zliming2004.lofter.com/post/1cc9dc55_79ad5d8

FG-4592 - zliming2004 - zliming2004的博客

Condensation of 5-bromophthalide (I) with phenol (II) in the presence of K2CO3, CuBr and acetylacetone in DMF gives 5-phenoxyphthalide (III), which upon lactone ring opening using SOCl2, Ph3PCl2, B(OMe)3 and K2CO3 in refluxing toluene yields 2-chloromethyl-4-phenoxybenzoyl chloride (IV). Esterification of acid chloride (IV) with MeOH at 50 °C furnishes the methyl ester (V), which is then condensed with methyl N-tosylglycinate (VI) in the presence of K2CO3 and NaI in DMF at 50 °C to afford N-substituted aminoester (VII). Cyclization of the intermediate diester (VII) using NaOMe in MeOH leads to methyl 4-hydroxy-7-phenoxyisoquinoline-3-carboxylate (VIII), which is submitted to Mannich reaction with bis-dimethylaminomethane (IX) in the presence of AcOH at 57 °C to provide the dimethylaminomethyl compound (X). Treatment of amine (X) with Ac2O at 103 °C, followed by selective hydrolysis of the phenolic acetate with morpholine leads to methyl 1-acetoxymethyl-4-hydroxy-7-phenoxyisoquinoline-3-carboxylate (XI). Hydrogenolysis of the benzylic acetate (XII) in the presence of Pd/C and Na2CO3 in EtOAc yields methyl 4-hydroxy-1-methyl-7-phenoxyisoquinoline-3-carboylate (XII), which finally couples with glycine (XIII) in the presence of NaOMe in MeOH at 110 °C to afford the target roxadustat (1-3).

FG-4592 - zliming2004 - zliming2004的博客

Cyclization of 4-phenoxyphthalic acid (I) with glycine (II) at 215 °C gives the phthalimide (III), which upon esterification with MeOH and H2SO4 at reflux yields methyl ester (IV). Subsequent rearrangement of phthalimidoacetate (IV) by means of Na in BuOH at 97 °C, followed by flash chromatography provides the isoquinoline-2-carboxylate (V). Bromination of intermediate (V) using POBr3 and NaHCO3 in acetonitrile leads to butyl 8-bromo-3-hydroxy-6-phenoxy-isoquinoline-2-carboxylate (VI), which upon hydrolysis with NaOH in refluxing H2O/EtOH furnishes carboxylic acid (VII). Substitution of bromine in intermediate (VII) using MeI and BuLi in THF at -78 °C, followed by alkylation with PhCH2Br in the presence of K2CO3 in refluxing acetone affords the 2-methyl isoquinoline (VIII). Ester hydrolysis in intermediate (VIII) using KOH in MeOH gives the corresponding carboxylic acid (IX), which is then activated with i-BuOCOCl and Et3N in CH2Cl2, followed by coupling with benzyl glycinate hydrochloride (X) to yield benzylated roxadustat (XI). Finally, debenzylation of intermediate (XI) with H2 over Pd/C in EtOAc/MeOH provides the title compound (1).

FG-4592 - zliming2004 - zliming2004的博客

Condensation of 4-nitro-ortho-phthalonitrile (I) with phenol (II) in the presence of K2CO3 in DMSO gives 4-phenoxy-ortho-phthalonitrile (III) (1), which upon hydrolysis with NaOH (1) or KOH (2) in refluxing MeOH yields 4-phenoxyphthalic acid (IV) (1,2). Dehydration of dicarboxylic acid (IV) using Ac2O and AcOH at reflux furnishes the phthalic anhydride (V), which is then condensed with methyl 2-isocyanoacetate (VI) using DBU in THF to provide oxazole derivative (VII). Rearrangement of intermediate (VII) with HCl in MeOH at 60 °C leads to isoquinoline derivative (VIII), which is partially chlorinated by means of POCl3 at 70 °C to afford 1-chloro-isoquinoline derivative (IX). Substitution of chlorine in intermediate (IX) using Me3B, Pd(PPh3)4 and K2CO3 in refluxing dioxane gives methyl 4-hydroxy-1-methyl-7-phenoxy-3-carboxylate (X), which is then hydrolyzed with aqueous NaOH in refluxing EtOH to yield the carboxylic acid (XI). Coupling of carboxylic acid (XI) with methyl glycinate hydrochloride (XII) by means of PyBOP, (i-Pr)2NH and Et3N in CH2Cl2 yields roxadustat methyl ester (XII), which is finally hydrolyzed with aqueous NaOH in THF to afford the target roxadustat (1).

CLIPS

SAN FRANCISCO, Nov 12, 2013 (BUSINESS WIRE) — FibroGen, Inc. (FibroGen), today announced that data from a China-based Phase 2 study of roxadustat (FG-4592), a first-in-class oral compound in late stage development for the treatment of anemia associated with chronic kidney disease (CKD) and end-stage renal disease (ESRD), were presented in an oral session at the 2013 American Society of Nephrology (ASN) Kidney Week in Atlanta, Georgia.
Roxadustat is an orally administered, small molecule inhibitor of hypoxia-inducible factor (HIF) prolyl hydroxylase. HIF is a protein that responds to oxygen changes in the cellular environment and meets the body’s demands for oxygen by inducing erythropoiesis, the process by which red blood cells are produced and iron is incorporated into hemoglobin (Hb).
The randomized, double-blind, placebo-controlled study was designed to evaluate the efficacy, safety, and tolerability of roxadustat in the correction of anemia in patients (N=91) with chronic kidney disease who had not received dialysis treatment, were not receiving erythropoiesis-stimulating agents (ESAs), and had Hb levels less than 10 g/dL. The correction study randomized patients 2:1 between roxadustat and placebo for 8 weeks of dosing, and included a low-dose cohort (n=30) and high-dose cohort (n=31). Intravenous (IV) iron was not allowed. The study also evaluated iron utilization, changes in serum lipids, and other biomarkers during treatment with roxadustat.
Data from this study suggest that roxadustat effectively corrected hemoglobin levels in anemic CKD patients in a dose-dependent manner as compared to placebo, and did so in the absence of IV iron supplementation regardless of degree of iron repletion at baseline. At the end of the 8-week treatment period, subjects showed mean maximum Hb increases from baseline of 2.6 g/dL in the high dose cohort and 1.8 g/dL in the low dose cohort, as compared to 0.7 g/dL in the placebo group (p < 0.0001) from mean baseline Hb of 8.8 g/dL, 8.8 g/dL, and 8.9 g/dL in the high dose, low dose, and placebo groups, respectively. 87% of patients in the high-dose cohort, 80% of patients in the low-dose cohort, and 23% of patients in the placebo group experienced a hemoglobin increase of 1 g/dL or greater from baseline (p < 0.0001). Similarly, 71% of patients in the high-dose cohort, 50% of patients in the low-dose cohort, and 3% of patients in the placebo group achieved target hemoglobin of 11 g/dL or greater (p < 0.0001). Serum iron levels remained stable in subjects randomized to roxadustat while the subjects underwent brisk erythropoiesis.
Study data also suggest that roxadustat may lower cholesterol. Dyslipidemia is highly prevalent in chronic kidney disease patients and a major cardiovascular risk factor in this population. Patients treated with roxadustat experienced a statistically significant reduction in total cholesterol (p <0.0001) and low-density lipoprotein (LDL) cholesterol (p <0.0001) at the end of the treatment period. The relative proportion of high density lipoprotein (HDL) cholesterol to LDL cholesterol increased significantly (p <0.02). Overall LDL cholesterol levels declined by a mean of 26% and median of 23% from a mean baseline value of 103 mg/dL.
Roxadustat was well tolerated by patients in the study with incidence of adverse events similar across all groups. In contrast to the exacerbation of hypertension observed in studies in which patients received currently available ESA therapies, subjects who received roxadustat in the present study showed small decreases in blood pressure that were similar to blood pressure changes in the placebo group. No cardiovascular serious adverse events were reported in patients treated with roxadustat.
The efficacy and safety of roxadustat are currently being investigated in a global pivotal Phase 3 development program.
“There is a global need for effective, safe, and accessible anemia therapies,” said Thomas B. Neff, Chief Executive Officer of FibroGen. “Side effects associated with current treatments include exposure to supra-physiological levels of erythropoietin and depletion of iron stores. Preliminary clinical findings show that oral administration of roxadustat (FG-4592) is able to correct anemia and maintain hemoglobin levels in patients with chronic kidney disease, to do so with peak erythropoietin levels within physiological range, and to achieve these effects without the administration of intravenous iron. These results suggest roxadustat, as an oral agent, has the potential to overcome the treatment barriers and inconveniences of current ESA therapies, including administration by injection and IV iron supplementation, in treating anemia in CKD patients.”
About Chronic Kidney Disease (CKD) and Anemia
Diabetes, high blood pressure, and other conditions can cause significant damage to the kidneys. If left untreated, those can result in chronic kidney disease and progress to kidney failure. Such deterioration can lead to patients needing a kidney transplant or being placed on dialysis to remove excess fluid and toxins that build up in the body. The progression of CKD also increases the prevalence of anemia, a condition associated with having fewer of the red blood cells that carry oxygen through the body, and/or lower levels of hemoglobin, the protein that enables red blood cells to carry oxygen. As hemoglobin falls, the lower oxygen-carrying capacity of an anemic patients’ blood results in various symptoms including fatigue, loss of energy, breathlessness, and angina. Anemia in CKD patients has been associated with increased hospitalization rates, increased mortality, and reduced quality of life.
Chronic kidney disease is a worldwide critical healthcare problem that affects millions of people and drives significant healthcare cost. In the US, prevalence of CKD has increased dramatically in the past 20 years, from 10 percent of the adult population (or approximately 20 million U.S. adults) as stated in the National Health and Nutrition Evaluation Survey (NHANES) 1988-1994, to 15 percent (or approximately 30 million U.S. adults) in NHANES 2003-2006. In 2009, total Medicare costs for CKD patients were $34 billion. China has an estimated 145 million CKD patients, or approximately five times the number of CKD patients in the U.S. (Lancet April 2012).
About Roxadustat / FG-4592
Roxadustat (FG-4592) is an orally administered small molecule inhibitor of hypoxia-inducible factor (HIF) prolyl hydroxylase activity, in development for the treatment of anemia in patients with chronic kidney disease (CKD). HIF is a protein transcription factor that induces the natural physiological response to conditions of low oxygen, “turning on” erythropoiesis (the process by which red blood cells are produced) and other protective pathways. Roxadustat has been shown to correct anemia and maintain hemoglobin levels without the need for supplementation with intravenous iron in CKD patients not yet receiving dialysis and in end-stage renal disease patients receiving dialysis. An Independent Data Monitoring Committee has found no signals or trends to date to suggest that treatment with roxadustat is associated with increased risk of cardiovascular events, thrombosis, or increases in blood pressure requiring initiation or intensification of antihypertensive medications.
About FibroGen
FibroGen is a privately-held biotechnology company focused on the discovery, development, and commercialization of therapeutic agents for treatment of fibrosis, anemia, cancer, and other serious unmet medical needs. FibroGen’s FG-3019 monoclonal antibody is in clinical development for treatment of idiopathic pulmonary fibrosis and other proliferative diseases, including pancreatic cancer and liver fibrosis. Roxadustat (FG-4592), FibroGen’s small molecule inhibitor of hypoxia-inducible factor (HIF) prolyl hydroxylase, is currently in clinical development for the treatment of anemia. FibroGen is also currently pursuing the use of proprietary recombinant human type III collagens in synthetic corneas for treatment of corneal blindness. For more information please visit: www.fibrogen.com .

References

1: Besarab A, Provenzano R, Hertel J, Zabaneh R, Klaus SJ, Lee T, Leong R, Hemmerich S, Yu KH, Neff TB. Randomized placebo-controlled dose-ranging and pharmacodynamics study of roxadustat (FG-4592) to treat anemia in nondialysis-dependent chronic kidney disease (NDD-CKD) patients. Nephrol Dial Transplant. 2015 Oct;30(10):1665-73. doi: 10.1093/ndt/gfv302. Epub 2015 Aug 3. PubMed PMID: 26238121; PubMed Central PMCID: PMC4569392.

2: Forristal CE, Levesque JP. Targeting the hypoxia-sensing pathway in clinical hematology. Stem Cells Transl Med. 2014 Feb;3(2):135-40. doi: 10.5966/sctm.2013-0134. Epub 2013 Dec 26. PubMed PMID: 24371328; PubMed Central PMCID: PMC3925058.

3: Bouchie A. First-in-class anemia drug takes aim at Amgen’s dominion. Nat Biotechnol. 2013 Nov;31(11):948-9. doi: 10.1038/nbt1113-948b. PubMed PMID: 24213751.

4: Flight MH. Deal watch: AstraZeneca bets on FibroGen’s anaemia drug. Nat Rev Drug Discov. 2013 Oct;12(10):730. doi: 10.1038/nrd4135. PubMed PMID: 24080688.

5: Beuck S, Schänzer W, Thevis M. Hypoxia-inducible factor stabilizers and other small-molecule erythropoiesis-stimulating agents in current and preventive doping analysis. Drug Test Anal. 2012 Nov;4(11):830-45. doi: 10.1002/dta.390. Epub 2012 Feb 24. Review. PubMed PMID: 22362605.

6: Cases A. The latest advances in kidney diseases and related disorders. Drug News Perspect. 2007 Dec;20(10):647-54. PubMed PMID: 18301799.

//////////ASP1517,  ASP 1517,  ASP-1517,  FG-4592,  FG 4592,  FG4592,  Roxadustat, PHASE 3, ASTELLAS, FibroGen, 808118-40-3
O=C(O)CNC(C1=C(O)C2=C(C(C)=N1)C=C(OC3=CC=CC=C3)C=C2)=O
Share

ASP 3026

 Uncategorized  Comments Off on ASP 3026
May 022016
 

Figure

ASP3026

ASP3026;

CAS 1097917-15-1; ASP-3026; ASP 3026; UNII-HP4L6MXF10;

N2-[2-Methoxy-4-[4-(4-methyl-1-piperazinyl)-1-piperidinyl]phenyl]-N4-[2-[(1-methylethyl)sulfonyl]phenyl]-1,3,5-triazine-2,4-diamine;

2-N-[2-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl]-4-N-(2-propan-2-ylsulfonylphenyl)-1,3,5-triazine-2,4-diamine

(N-{2-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}-N′-[2-(propane-2-sulfonyl)phenyl]-1,3,5-triazine-2,4-diamine)  was developed as a novel selective inhibitor of the fusion protein EML4-ALK.

1H NMR (CDCl3, 400 MHz) (ppm) = 1.31 (d, 6H, J = 6.8 Hz), 1.58–1.80 (m, 4H), 1.90–2.04 (m, 2H), 2.16–2.84 (m, 12H), 3.18–3.32 (m, 1H), 3.66–3.76 (m, 2H), 3.88 (s, 3H), 6.48–6.60 (m, 2H), 7.18–7.26 (m, 1H), 7.50–7.72 (m, 2H), 7.86–7.92 (dd, 1H, J = 1.2 Hz, J = 7.6 Hz), 8.06–8.16 (m, 1H), 8.28–8.48 (m, 1H), 8.48–8.62 (m, 1H), 9.28 (s, 1H).

Molecular Formula: C29H40N8O3S
Molecular Weight: 580.7447 g/mol

ASP3026 is a novel and selective inhibitor for the ALK kinase. ASP3026 potently inhibited ALK kinase activity and was more selective than crizotinib in a Tyr-kinase panel. In an anchorage independent in vitro cell growth assay, ASP3026 inhibited the growth of NCI-H2228, a human NSCLC tumor cell line endogenously expressing EML4-ALK variant 3 and that of 3T3 cells expressing EML4-ALK variant 1, 2 and 3. The plasma and tumor concentrations of ASP3026 in mice xenografted with NCI-H2228 tumor were determined using high-performance liquid chromatography-tandem mass spectrometry. Significant tumor penetration was observed. The antitumor activities were evaluated using mice bearing subcutaneous NCI-H2228 tumor xenografts.

ASP-3026 was studied in P1 clinical trials at Astellas Pharma for the oral treatment of advanced solid tumors and advanced B-cell lymphoma. In 2014 the product was discontinued by Astellas due to strategic reasons

JP 2012153674

WO 2012102393

WO 2011145548

WO 2009008371

PATENT

WO2012102393

The compound of the formula (1) has an excellent EML4-ALK fusion protein and inhibitory activity of the kinase of the mutant EGFR protein, we are already reported to be useful as an active ingredient of a pharmaceutical composition for cancer treatment (Patent Document 1). Further, it is the compound of formula (1) there are five polymorphs shown as A01 ~ A05 type, among others A04 type crystal is in finding reported that the most stable type crystals (Japanese Patent Document 2).
[Formula 1]  a compound of formula (1) described in Patent Document 1 production method of (Patent Document 1 of Example 23), referring to Production Examples and Examples described in this document, the reaction formula (I) It is shown in. That is, 2,4-dichloro-1,3,5-triazine (hereinafter, may be referred to as “compound of formula (15)”.), 2- (isopropylsulfonyl) aniline (hereinafter, “the formula (8) sometimes referred to compound “.) using, by reacting according to the method described in production example 7 of this document, to give compounds of formula (14) to (production example 22 of Patent Document 1), then , the resulting compound of formula (14), a known method (e.g., International Publication No. 2005/016894 pamphlet reference) was prepared by 2-methoxy-4- [4- (4-methylpiperazin-1- yl) piperidin-1-yl] aniline (hereinafter, may be referred to as “formula (13) compounds of.”) is used to react according to the method described in example 1 of the document, and the target it is a method for producing a compound of formula (1) to.

 

[Formula 2]

Patent Document 1: International Publication No. 2009/008371 pamphlet
Patent Document 2: WO 2011/145548 pamphlet

Example 1
The first step 4,4-dimethoxy-1- (3-methoxy-4-nitrophenyl) piperidine (R 1 and R 2 Synthesis of methyl Any compound of formula (10))
 4,4 – N and dimethoxy piperidine monohydrochloride (35.9 g), N-dimethylformamide and (75 mL) were mixed, and the mixed solution of 1,8-diazabicyclo [5.4.0] undec-7-ene (57.5 mL) was added It was. It was separately prepared here 5-fluoro-2-nitroanisole (30.0 g) and N, N-dimethylformamide (30 mL) was stirred for 5 hours at room temperature. Water (120 mL) was added at room temperature to the reaction mixture, after stirring for 4 hours, the precipitated crystals were collected by filtration. The resulting crystals N, N-dimethylformamide and a mixed solution of water (1: 1) (60mL) , water (60 mL), was further washed sequentially with water (60 mL), and dried under reduced pressure at 40 ° C. to give 4,4-dimethoxy-1- (3-methoxy-4-nitrophenyl) piperidine (49.9 g, 96.1% yield) as crystals.
D2: 1.72-1.80 (4H, m) , 3.14 (6H, s), 3.44-3.50 (4H, m), 3.91 (3H, m), 6.52 (1H, d, J = 2.4Hz), 6.60 (1H, dd, J = 2.4,9.2Hz), 7.88 (1H, D, J = 9.2Hz)
ESI Tasu: 297

The second step 4- (R (4,4-dimethoxy-1-yl) -2-methoxyaniline 1 and R 2 is methyl none has the formula (Compound 6)) Synthesis of

 4,4-dimethoxy – 1- (3-methoxy-4-nitrophenyl) piperidine and (45.0 g) in tetrahydrofuran and a (225 mL) were mixed, 5% palladium carbon (about 50% wet product, 4.5 g) to this mixed solution was added at room temperature, under a hydrogen atmosphere (2.4821×10 5 Pa), and the mixture was stirred for 5 hours and a half at room temperature. Then filtered off and palladium-carbon, washed with tetrahydrofuran (90mL), was concentrated under reduced pressure filtrate until total volume of about 90mL obtain a slurry. After the slurry was stirred for 1 hour at 40 ° C., n- heptane (135 mL) was added and after stirring for 1 hour at 40 ° C., cooled to 0 ° C., was added n- heptane (405 mL), precipitated crystals It was collected by filtration.The obtained crystals were washed with a mixed solution of tetrahydrofuran (9 mL) and n- heptane (54 mL), and dried in vacuo at 40 ℃, 4- (4,4- dimethoxy-1-yl) -2-methoxy to give aniline (37.9g, 93.7% yield) as crystals.
D2: 1.72-1.80 (4H, m) , 2.90-2.97 (4H, m), 3.11 (6H, s), 3.73 (3H, m), 4.21 (1H, br), 6.30 (1H, d, J = 2.4 , 8.4Hz), 6.46_6.56 (2H, M)
ESI Tasu: 267

The third step 4,6-dichloro-N- [2-(propane-2-sulfonyl) phenyl] -1,3,5-triazin-2-amine (Lv is Cl any, compounds of formula (7) synthesis of)

 cyanuric chloride (25.0 g), sodium bicarbonate (13.7 g), were mixed 2- (isopropylsulfonyl) aniline (29.7 g) and acetone (200 mL), and stirred at room temperature for 25 hours. After adding water (200 mL) at room temperature the reaction mixture was stirred for 19 hours, the precipitated crystals were collected by filtration. The resulting crystals acetone and a mixed solution of water (1: 1) was washed with (100 mL), and dried in vacuo at 40 ° C., 4,6-dichloro-N- [2-(propane-2-sulfonyl) to give phenyl] -1,3,5-triazin-2-amine (45.1g, 95.8% yield) as crystals.
D1: 1.32 (6H, d, J = 6.8Hz), 3.22 (1H, sept, J = 6.8Hz), 7.37 (1H, m), 7.74 (1H, m), 7.93 (1H, m), 8.44 (1H , M), 10.02 (1H, Br)
ESI-: 345, 347

 

Fourth step 6-chloro -N- [4- (4,4- dimethoxy-1-yl) -2-methoxy-phenyl] -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3 , (a Lv is Cl, R 5- triazine-2,4-diamine 1and R 2 none is methyl, the formula (compound 5)) synthesis of
4,6-dichloro-N- [2-( propane-2-sulfonyl) phenyl] -1,3,5-triazin-2-amine (40.0 g) was mixed with tetrahydrofuran (400 mL), to this mixed solution 4- (4,4-dimethoxy-piperidin-1 yl) -2-methoxyaniline (32.2 g) and N, N- diisopropylethylamine (16.38g) was stirred for 4 hours at room temperature.Thereafter, isopropyl acetate (40 mL), then extracted by adding a mixed solution of potassium carbonate (2.0 g) and water (40 mL). The obtained organic layer was concentrated under reduced pressure until the total volume of about 200 mL, as a seed crystal, 6-chloro -N- [4- (4,4- dimethoxy-1-yl) -2-methoxyphenyl] -N ‘- inoculated with [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-crystalline diamine (4 mg), to give a slurry and stirred for about 15 minutes. The slurry n- heptane (200 mL) was added and filtered off cooled to 18 hours with stirring to precipitate crystals to 0 ° C.. The resulting crystals were washed with a mixed solution of tetrahydrofuran (40 mL) and n- heptane (40 mL), and dried in vacuo at 40 ° C., 6- Chloro -N- [4- (4,4- dimethoxy-piperidine – 1-yl) -2-methoxyphenyl] -N ‘- [2- (the propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (61.4 g, 92.4% yield) It was obtained as a crystal.
D1: 1.30 (6H, d, J = 6.8Hz), 1.88-1.92 (4H, m), 3.18-3.26 (1H, m), 3.23 (3H, s), 3.87 (1H, br), 6.53 (2H, br), 7.21-7.23 (1H, m ), 7.62 (1H, br), 7.88 (1H, d, J = 7.9Hz), 8.05 (1H, br), 8.48 (1H, br), 9.41 (1H, br )
ESI-: 575,577

 

The fourth alternative process (e.g. without using a seed crystal) 6-Chloro-N- [4- (4,4-dimethoxy-1-yl) -2-methoxyphenyl] -N ‘- [2- (propane 2-sulfonyl) phenyl] (a Lv is Cl, R-1,3,5-triazine-2,4-diamine 1 and R 2 none is methyl, the formula (5) synthesis of compound of)
4 , and mixed 6-dichloro -N- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazin-2-amine (23.0 g) in tetrahydrofuran (230 mL), to this mixed solution 4- (4,4-dimethoxy-1-yl) -2-methoxyaniline (18.5 g) and N, N- diisopropylethylamine (12.7 mL) was stirred for 2 hours at room temperature. Thereafter, isopropyl acetate (57.5 mL), then extracted by adding potassium carbonate (5.75 g) and a mixed solution of water (115 mL). The resulting organic layer was concentrated under reduced pressure. The resulting residue is added and stirred in tetrahydrofuran (50mL) to obtain a slurry. After stirring for 1 hour at the slurry was added tetrahydrofuran (75 mL) and n- heptane (75mL) 40 ℃, cooled to 0 ° C., and stirred for a further 18 hours.Thereafter, n- heptane (50 mL) was added, and the precipitated crystals were collected by filtration. The resulting crystals tetrahydrofuran and n- heptane mixed solution (5: 7) After washing with (24 mL), and dried in vacuo at 40 ° C., 6- chloro-N- [4- (4,4-dimethoxy piperidin-1-yl) -2-methoxyphenyl] -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (30.6g, 80.0% yield ) was obtained as a crystal.

 

D1: 1.30 (6H, d, J = 6.8Hz), 1.88-1.92 (4H, m), 3.18-3.26 (1H, m), 3.23 (3H, s), 3.87 (1H, br), 6.53 (2H, br), 7.21-7.23 (1H, m ), 7.62 (1H, br), 7.88 (1H, d, J = 7.9Hz), 8.05 (1H, br), 8.48 (1H, br), 9.41 (1H, br )
ESI-: 575,577

 

The fifth step and the sixth step (continuous process) 1- [3-methoxy-4 – ({4- [2- (propane-2-sulfonyl) anilino] -1,3,5-triazin-2-yl} amino ) phenyl] piperidin-4-one synthesis of compound) (formula (3)
6-chloro-N- [4- (4,4-dimethoxy-1-yl) -2-methoxyphenyl] -N ‘- [ 2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (60.0 g), tetrahydrofuran (540 mL) and 10% palladium carbon (about 50% wet product, 10.7 g) and mixed, N to the mixture, added to N- diisopropylethylamine (16.11g) and 2-propanol (60 mL), under a hydrogen atmosphere (2.4131X10 5 of 5 Pa), and stirred for 7 hours at 40 ° C.. Filtration of the palladium-carbon, and washed with tetrahydrofuran (120 mL), the resulting filtrate activated carbon (12.0 g) was added to, and stirred at room temperature overnight. Then filtered off and the activated carbon, and washed with tetrahydrofuran (120mL), N- [4- ( 4,4- dimethoxy-1-yl) -2-methoxyphenyl] -N ‘- [2- (propane – to obtain a solution containing 2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine. To this solution was added a mixed solution of 35% hydrochloric acid (21.7 g) and water (120 mL), and stirred for 21 hours at room temperature. To the reaction mixture, it was added a mixed solution of potassium carbonate (35.9 g) and water (120 mL), and extracted. Activated carbon (12.0 g) was added to the obtained organic layer was stirred for 16 h, filtered, washed with activated carbon in tetrahydrofuran (120 mL). The filtrate obtained total amount was concentrated under reduced pressure to approximately 120 mL. After addition of acetone (180 mL) to the resulting mixture, as a seed crystal, 1- [3-methoxy-4 – ({4- [2- (propane-2-sulfonyl) anilino] -1,3,5 after stirring for 1 hour and inoculated triazin-2-yl} amino) phenyl] piperidin-4-one crystals (60 mg), water (480 mL) was stirred for 20 hours was added, and the precipitated crystals were collected by filtration . The obtained crystals were washed with a mixed solution of acetone (36 mL) and water (96 mL), and dried in vacuo at 40 ℃, 1- [3- methoxy-4 – ({4- [2- (propane -2 – was obtained sulfonyl) anilino] -1,3,5-triazine-2-yl} amino) phenyl] piperidine-4-one (45.8g, 88.7% yield (yield in a continuous two steps)) as crystals .
D2,343K: 1.17 (6H, d, J = 6.8Hz), 2.46-2.50 (4H, m), 3.40 (1H, sept, J = 6.8Hz), 3.61 (4H, dd, J = 6.1,6.2Hz) , 3.79 (3H, s), 6.57 (1H, dd, J = 2.6,8.7Hz), 6.70 (1H, d, J = 2.6Hz), 7.25-7.29 (1H, m), 7.38 (1H, d, J 8.7 Hz =), 7.61 (1H, br), 7.77-7.80 (1H, yd), 8.28 (1H, s), 8.50 (1H, br), 8.66 (1H, br), 9.25 (1H, br)
ESI +: 497

 

Fifth Step N- [4- (4,4- dimethoxy-1-yl) -2-methoxyphenyl] -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine 2,4-diamine (R 1 and R 2 is methyl any formula (4) of compound) synthesis of
6-chloro-N- [4- (4,4-dimethoxy-1-yl) – 2-methoxyphenyl] -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (5.0 g), tetrahydrofuran (45 mL), 2-propanol (5mL ), 10% palladium-carbon (about 50% wet product, 1.0 g) were mixed, added N, N- diisopropylethylamine (1.81 mL) to this mixed solution, under a hydrogen atmosphere (2.4821X10 5 of 5 Pa), 40 ° C. in and the mixture was stirred for 5 hours and a half. Filtration of the palladium-carbon was washed with tetrahydrofuran (10 mL), and extraction was performed with 10% brine (20 mL). The resulting organic layer was concentrated under reduced pressure. Acetone to the concentrated residue (10 mL), was added diisopropyl ether (40 mL), it was collected by filtration stirred precipitated crystals 30 minutes. The obtained crystals were washed with diisopropyl ether (20 mL), and dried in vacuo at 40 ℃, N- [4- (4,4- dimethoxy-1-yl) -2-methoxyphenyl]-N’- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (4.31 g, 91.6% yield) as crystals.
D2,343K: 1.17 (6H, d, J = 6.8Hz), 1.80 (4H, dd, J = 5.5,5.7Hz), 3.15 (6H, s), 3.21 (4H, dd, J = 5.5,5.7Hz) , 3.77 (3H, s), 6.50 (1H, dd, J = 2.5,8.7Hz), 6.62 (1H, d, J = 2.5Hz), 7.25-7.28 (1H, m), 7.34 (1H, d, J 8.7 Hz =), 7.58 (1H, br), 7.77-7.79 (1H, yd), 8.28 (1H, s), 8.49 (1H, br), 8.63 (1H, br), 9.25 (1H, br)
ESI +: 543

 

Sixth Step 1- [3-methoxy-4 – ({4- [2- (propane-2-sulfonyl) anilino] -1,3,5-triazin-2-yl} amino) phenyl] piperidin-4-one (equation (3) a compound of) synthesis of
N- [4- (4,4- dimethoxy-1-yl) -2-methoxyphenyl] -N ‘- [2- (propane-2-sulfonyl) phenyl] – 1,3,5-triazine-2,4-diamine (4.0 g), and tetrahydrofuran (36 mL) and 2-propanol (4 mL) solution of 35% hydrochloric acid containing (1.44 g) a mixture of water (4 mL) was added on, and the mixture was stirred for 17 hours at room temperature. To the reaction mixture, it was added a mixed solution of potassium carbonate (2.4 g) and water (4 mL), and extracted.The resulting organic layer was concentrated under reduced pressure. After stirring for 30 minutes by addition of acetone (12 mL) and water (4 mL) to the concentrated residue, add water (28 mL) was stirred for 1 hour, the precipitated crystals were collected by filtration. The obtained crystals were washed with a mixed solution of acetone (8 mL) and tetrahydrofuran (3 mL), and dried in vacuo at 40 ℃, 1- [3- methoxy-4 – ({4- [2- (propane -2 – give sulfonyl) anilino] -1,3,5-triazin-2-yl} amino) phenyl] piperidin-4-one (3.42g, 99.2% yield) as crystals.
D2,343K: 1.17 (6H, d, J = 6.8Hz), 2.46-2.50 (4H, m), 3.40 (1H, sept, J = 6.8Hz), 3.61 (4H, dd, J = 6.1,6.2Hz) , 3.79 (3H, s), 6.57 (1H, dd, J = 2.6,8.7Hz), 6.70 (1H, d, J = 2.6Hz), 7.25-7.29 (1H, m), 7.38 (1H, d, J 8.7 Hz =), 7.61 (1H, br), 7.77-7.80 (1H, yd), 8.28 (1H, s), 8.50 (1H, br), 8.66 (1H, br), 9.25 (1H, br)
ESI +: 497

 

Seventh Step N- {2- methoxy-4- [4- (4-methylpiperazin-1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] – 1,3,5-triazine-2,4-diamine (formula (1) compounds) synthesis
of 1- [3-methoxy-4 – ({4- [2- (propane-2-sulfonyl) anilino] -1 , 3,5-triazin-2-yl} amino) phenyl] piperidin-4-one (20.0 g), methyl piperazine (8.07 g), were mixed in toluene (200 mL) and acetic acid (9.0 mL), 1 hour at room temperature It stirred. To this mixture solution was added sodium triacetoxyborohydride (17.06 g), and stirred at room temperature for 20 hours. To the reaction mixture, water (60 mL) and methanol (20 mL) was added, extraction to give an organic layer and an aqueous layer 1. This organic layer, water (20 mL) and re-extracted to give a water layer 2. After mixing the aqueous layer 1 and aqueous layer 2 was extracted by adding isopropyl acetate (200 mL). Methanol (220 mL) to the resulting aqueous layer, a mixed solution of sodium hydroxide (9.68 g) and water (48 mL) was added, as a seed crystal, N-{2-methoxy-4- [4- (4-methylpiperazin- 1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-crystal of diamine (2.0mg) inoculated, after stirring at room temperature for 1.5 hours, add water (220 mL), further stirred for 2 hours at room temperature, the precipitated crystals were collected by filtration. The resulting crystals were washed with a mixed solution of methanol (40mL) and water (40mL), and then dried under reduced pressure at 50 ℃, N- {2- methoxy-4- [4- (4-methyl-piperazine -1 – yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (20.15g, 86.1% yield) It was obtained as A06-form crystals.
D1: 1.31 (6H, d, J = 6.8Hz), 1.59-1.78 (2H, m), 1.90-2.01 (2H, m), 2.24-2.80 (11H, m), 2.30 (3H, s), 3.19- 3.32 (1H, m), 3.65-3.75 (2H, m), 3.88 (3H, s), 6.50-6.59 (2H, m), 7.18-7.30 (1H, m), 7.53-7.70 (2H, m), 7.88 (1H, dd, J = 1.5,8.3Hz), 8.10 (1H, br), 8.37 (1H, br), 8.53 (1H, br), 9.29 (1H, s)
ESI +: 581

Alternatively 1 (Example not using seed crystals) N-{2-methoxy-4- [4- (4-methylpiperazin-1-yl) piperidin-1-yl] phenyl} -N seventh step ‘- [ 2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (compound of formula (1))

 1- [3-methoxy-4 – ({4- [2 – (propane-2-sulfonyl) anilino] -1,3,5-triazin-2-yl} amino) phenyl] piperidin-4-one (5.0 g), methyl piperazine (2.02 g), toluene (50 mL) and acetic acid (1.5 mL) were mixed and stirred at room temperature for 1 hour. To this mixture solution was added sodium triacetoxyborohydride (4.72 g), and stirred at room temperature for 18 hours. To the reaction mixture, water (15 mL) and methanol (5 mL) was added, extraction to give an organic layer and an aqueous layer 1. This organic layer, water (5 mL) and re-extracted to give a water layer 2. After mixing the aqueous layer 1 and aqueous layer 2 was extracted by adding isopropyl acetate (50 mL). The resulting aqueous layer methanol (55 mL), a mixed solution was added sodium hydroxide (2.0 g) and water (10 mL), was stirred for 62 hours at room temperature, add water (55 mL), at room temperature for a further 2 hours stirring, the formed crystals were separated by filtration. The obtained crystals were washed with a mixed solution of methanol (5 mL) and water (5 mL), and dried in vacuo at 40 ℃, N- {2- methoxy-4- [4- (4-methylpiperazin–1 – yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (4.56g, 78.0% yield) It was obtained as A06-form crystals.
D1: 1.31 (6H, d, J = 6.8Hz), 1.59-1.78 (2H, m), 1.90-2.01 (2H, m), 2.24-2.80 (11H, m), 2.30 (3H, s), 3.19- 3.32 (1H, m), 3.65-3.75 (2H, m), 3.88 (3H, s), 6.50-6.59 (2H, m), 7.18-7.30 (1H, m), 7.53-7.70 (2H, m), 7.88 (1H, dd, J = 1.5,8.3Hz), 8.10 (1H, br), 8.37 (1H, br), 8.53 (1H, br), 9.29 (1H, s)
ESI +: 581
alternative seventh step 2 (example using reducing catalyst) N-{2-methoxy-4- [4- (4-methylpiperazin-1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane -2 – sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine synthesis of compounds of formula (1)
1- [3-methoxy-4 – ({4- [2- (propan-2 sulfonyl) anilino] -1,3,5-triazin-2-yl} amino) phenyl] piperidin-4-one (5.0 g), tetrahydrofuran (30 mL), methylpiperazine (1.81 g) and 10% palladium carbon (about 50 % wet product, were mixed 0.8 g), under a hydrogen atmosphere (2.4821X10 5 of 5Pa), and stirred for 7 hours at 40 ° C.. Filtration of the palladium-carbon, and washed with tetrahydrofuran (10 mL), the resulting filtrate was concentrated under reduced pressure. To the concentrated residue 2-butanone (9 mL) was added, followed by stirring at 60 ° C. 30 minutes, cooled slowly, at 30 ° C. n-heptane (9 mL) was added, and stirred for 19 hours at room temperature, the precipitated crystals were collected by filtration did.The resulting crystals of 2-butanone and (1 mL) was washed with a mixture of n- heptane (1 mL), and dried in vacuo at 40 ℃, N- {2- methoxy-4- [4- (4-methyl piperazin-1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (3.09 g, yield: 88.0%) was obtained.
D1: 1.31 (6H, d, J = 6.8Hz), 1.59-1.78 (2H, m), 1.90-2.01 (2H, m), 2.24-2.80 (11H, m), 2.30 (3H, s), 3.19- 3.32 (1H, m), 3.65-3.75 (2H, m), 3.88 (3H, s), 6.50-6.59 (2H, m), 7.18-7.30 (1H, m), 7.53-7.70 (2H, m), 7.88 (1H, dd, J = 1.5,8.3Hz), 8.10 (1H, br), 8.37 (1H, br), 8.53 (1H, br), 9.29 (1H, s)
ESI +: 581

 

 N- {2- methoxy-4- [4- (4-methylpiperazin-1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3 , 5-triazine-2,4-diamine by recrystallization purification steps (formula (1 compound of))
(the a method) N-{2-methoxy-4- [4- (4-methylpiperazin-1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (8.80 g), 2-butanone (211 mL) after mixing and confirmation of dissolution and stirring at 65 ° C. 30 minutes for clarifying filtration. After filtrate was total volume concentrated normal pressure to approximately 70 mL, and cooled to 70 ° C., as a seed crystal N- {2- methoxy-4- [4- (4-methylpiperazin-1-yl) piperidine-1 yl] phenyl} -N ‘- [2- inoculated with (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-crystalline diamine (0.9 mg), and stirred for about 10 minutes to obtain a slurry. After stirring for 3 hours at 70 ° C., cooled to 5 ℃ at a rate of 20 ° C. / h and stirred for 17 hours, the precipitated crystals were collected by filtration. The resulting crystals were washed with 2-butanone were cooled with ice water (35.2 mL), and dried in vacuo at 50 ℃, N- {2- methoxy-4- [4- (4-methylpiperazin-1- yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (7.88 g, 89.5% yield, purity 99.4%) was obtained as a A04 type crystal (A04 type ratio 98.9%).

 

(B method): N- {2- methoxy-4- [4- (4-methylpiperazin-1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl ] -1,3,5-triazine-2,4-diamine (8.80g), was mixed activated carbon (0.88 g) and 2-butanone (211 mL), after stirring for 1 hour at 75 ° C., was subjected to activated carbon filtration .The filtrate activated carbon (0.88g) in addition to, and the mixture was stirred for 1 hour at 75 ℃, was activated carbon filtration. The filtrate activated carbon (0.88g) in addition to, and the mixture was stirred for 1 hour at 75 ℃, was activated carbon filtration. After filtrate was total volume concentrated normal pressure to approximately 70 mL, and cooled to 70 ° C., as a seed crystal N- {2- methoxy-4- [4- (4-methylpiperazin-1-yl) piperidine-1 yl] phenyl} -N ‘- [2- inoculated with (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-crystalline diamine (0.9 mg), and stirred for about 10 minutes to obtain a slurry. After stirring for 3 hours at 70 ° C., cooled to 5 ℃ at a rate of 20 ° C. / h and stirred for 16 hours, the precipitated crystals were collected by filtration. The resulting crystals were washed with 2-butanone were cooled with ice water (35.2 mL), and dried in vacuo at 50 ℃, N- {2- methoxy-4- [4- (4-methylpiperazin-1- yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (6.60 g, 75.0% yield, purity 99.3%) was obtained as A04 type crystal (A04 type ratio 100%).

 

Example 2
The first step 4,4-dimethoxy-1- (3-methoxy-4-nitrophenyl) piperidine (R 1 and R 2 is methyl Any formula (Compound 10)) Synthesis of
4,4 – dimethoxy piperidine monohydrochloride (69.9kg) and N, N-dimethylformamide (125.7kg) was mixed, to this mixed solution 1,8-diazabicyclo [5.4.0] undec-7-ene and (117.3kg) N It was added N- dimethylformamide (17.0kg). N of separately prepared here 5-fluoro-2-nitroanisole (60.0kg), the N- dimethylformamide (57.0kg) was added at room temperature, N, N- dimethylformamide (29.0 kg) solution was added 5 hours It stirred. At room temperature with a seed crystal of 4,4-dimethoxy-1- (3-methoxy-4-nitrophenyl) piperidine (about 6 g) was added to the reaction mixture was stirred at room temperature for 14 hours. Water (240 kg) was added at room temperature to the reaction mixture, after stirring for 22 hours, the precipitated crystals were collected by filtration. The obtained crystals N, washed with a mixed solution of N- dimethylformamide (56.9kg) and water (60kg), washed twice with water (120 kg), and dried in vacuo at 50 ° C., 4, 4 – to give dimethoxy-1- (3-methoxy-4-nitrophenyl) piperidine (99.7kg, 96.0% yield) as crystals.

 

D2: 1.72-1.80 (4H, m) , 3.14 (6H, s), 3.44-3.50 (4H, m), 3.91 (3H, m), 6.52 (1H, d, J = 2.4Hz), 6.60 (1H, dd, J = 2.4,9.2Hz), 7.88 (1H, D, J = 9.2Hz)
ESI Tasu: 297

 

The second step 4- (R (4,4-dimethoxy-1-yl) -2-methoxyaniline 1 and R 2 is methyl none has the formula (Compound 6)) Synthesis of
4,4-dimethoxy – 1- (3-methoxy-4-nitrophenyl) piperidine (99.0kg), 5% palladium carbon (about 50% wet product, 10.5 kg), were mixed at room temperature in tetrahydrofuran (440 kg), under a hydrogen atmosphere (200 ~ 300 kPa ), and stirred at room temperature for 3 hours. Then filtered off and palladium-carbon, tetrahydrofuran and washed with (180.5Kg), the filtrate was concentrated under reduced pressure until the total volume of about 220L, as a seed crystal 4- (4,4-dimethoxy-1-yl) – crystals of 2-methoxyaniline was inoculated (approximately 10g). To the resulting slurry n- heptane (205.4kg) was added at 40 ° C., after stirring for 1 h, was stirred and cooled to 0 ° C. 16 hours. To this slurry was added n- heptane (613.5kg), After stirring for 2 hours, the crystals were collected by filtration. The obtained crystals were washed with a mixed solution of tetrahydrofuran (17.8 kg) and n- heptane (81.5kg), and dried in vacuo at 50 ℃, 4- (4,4- dimethoxy-1-yl) -2 – give methoxyaniline (84.1kg, 94.5% yield) as crystals.

 

D2: 1.72-1.80 (4H, m) , 2.90-2.97 (4H, m), 3.11 (6H, s), 3.73 (3H, m), 4.21 (1H, br), 6.30 (1H, d, J = 2.4 , 8.4Hz), 6.46_6.56 (2H, M)
ESI Tasu: 267
The third step 4,6-dichloro-N- [2-(propane-2-sulfonyl) phenyl] -1,3,5-triazin-2-amine (Lv is Cl any, compounds of formula (7) synthesis of)
 cyanuric acid chloride (40.0kg) and acetone (249.2kg) was mixed at a 17 ℃. Sodium hydrogen carbonate in the mixed solution (21.9 kg), 2-a (isopropylsulfonyl) aniline (47.5Kg) was added, and stirred at room temperature for 23 hours. After adding to the reaction mixture water (320 kg) at room temperature, and stirred for 3.5 hours, the precipitated crystals were collected by filtration. After washing the obtained crystals with a mixed solution of acetone (63.0kg) and water (80 kg), and dried in vacuo at 50 ° C., 4,6-dichloro -N- [2- (propane-2-sulfonyl) phenyl ] -1,3,5-triazin-2-amine (71.6kg, 95.1% yield) was obtained as crystals.
D1: 1.32 (6H, d, J = 6.8Hz), 3.22 (1H, sept, J = 6.8Hz), 7.37 (1H, m), 7.74 (1H, m), 7.93 (1H, m), 8.44 (1H , M), 10.02 (1H, Br)
ESI-: 345, 347

 

Fourth step 6-chloro -N- [4- (4,4- dimethoxy-1-yl) -2-methoxy-phenyl] -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3 , (a Lv is Cl, R 5- triazine-2,4-diamine 1and R 2 none is methyl, the formula (compound 5)) synthesis of
4,6-dichloro-N- [2-( propane-2-sulfonyl) phenyl] -1,3,5-triazin-2-amine (70.9 kg) in tetrahydrofuran (611.1kg) was mixed at room temperature, to this mixed solution 4- (4,4-dimethoxy-piperidine 1-yl) -2-methoxyaniline (57.1kg), N, N- diisopropylethylamine (29.1 kg) was stirred for 4 hours at room temperature. Thereafter, isopropyl acetate (61.0kg), then extracted by adding potassium carbonate (3.6 kg) and a mixed solution of water (71 kg).The resulting organic layer total amount was concentrated under reduced pressure at an external temperature of about 40 ° C. to approximately 360 L, as a seed crystal, 6-chloro -N- [4- (4,4- dimethoxy-1-yl) -2 – methoxyphenyl] -N ‘- [2- was inoculated with (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-crystalline diamine (approximately 7 g) to give a slurry. To this slurry of 2-propanol (111.0kg), n- heptane (243.1kg) was added and after cooling for 2 hours at room temperature, was collected by filtration stirred precipitated crystals were cooled to 0 ℃ 18 hours. The resulting crystals tetrahydrofuran (74.9kg), 2- propanol (44.6kg), was washed with a mixed solution of n- heptane (97.6kg), and then dried under reduced pressure at 50 ℃, 6- chloro -N- [ 4- (4,4-dimethoxy-1-yl) -2-methoxyphenyl] -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine It was obtained (108.9kg, 92.4% yield) as crystals.

 

D1: 1.30 (6H, d, J = 6.8Hz), 1.88-1.92 (4H, m), 3.18-3.26 (1H, m), 3.23 (3H, s), 3.87 (1H, br), 6.53 (2H, br), 7.21-7.23 (1H, m ), 7.62 (1H, br), 7.88 (1H, d, J = 7.9Hz), 8.05 (1H, br), 8.48 (1H, br), 9.41 (1H, br )
ESI -: 575,577
fifth step and the sixth step (continuous process) 1- [3-methoxy-4 – ({4- [2- (propane-2-sulfonyl) anilino] -1,3,5-triazine – 2-yl} amino) phenyl] piperidin-4-one synthesis of compound) (formula (3)
6-chloro-N- [4- (4,4-dimethoxy-1-yl) -2-methoxyphenyl] -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (108.2kg), tetrahydrofuran (866.0kg), 10% palladium carbon (about 50% wet goods, 23.3 kg) were mixed, N to this mixed solution was added to N- diisopropylethylamine (28.9 kg) and 2-propanol (85.5kg), under a hydrogen atmosphere (100 ~ 300kPa), 4 hours at 40 ° C. did. Filtration of the palladium-carbon was washed with tetrahydrofuran (193.3kg), N- [4- ( 4,4- dimethoxy-1-yl) -2-methoxyphenyl] -N ‘- [2- (propane -2 – to obtain a solution containing a sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine. To this solution was added 35% hydrochloric acid (39.1 kg) of mixed solution of water (217kg), and stirred for 15 hours at room temperature. To the reaction mixture, added potassium carbonate (64.8kg) and a mixed solution of water (217kg), and extracted. Activated carbon (10.8 kg) was added to the obtained organic layer and stirred for 17 hours at room temperature, filtered and washed activated carbon with tetrahydrofuran (96.0kg). The resulting filtrate was concentrated under reduced pressure until the total volume of about 380L at 40 ° C.. After the resultant mixture was added acetone (257.1Kg), as a seed crystal, 1- [3-methoxy-4 – ({4- [2- (propane-2-sulfonyl) anilino] 1,3,5 – after stirring for 1 hour was inoculated triazin-2-yl} amino) phenyl] piperidin-4-one crystals (approximately 11g), the addition of water (865Kg) was stirred for 15 hours, the precipitated crystals were collected by filtration did. The obtained crystals were washed with a mixed solution of acetone (50.9kg) and Tsunemizu (173 kg), and dried in vacuo at 50 ℃, 1- [3- methoxy-4 – ({4- [2- (propane 2-sulfonyl) anilino] -1,3,5-triazine-2-yl} amino) phenyl] piperidine-4-one (82.9kg, 89.0% yield (yield in a continuous two steps)) as crystals Obtained.

 

D2,343K: 1.17 (6H, d, J = 6.8Hz), 2.46-2.50 (4H, m), 3.40 (1H, sept, J = 6.8Hz), 3.61 (4H, dd, J = 6.1,6.2Hz) , 3.79 (3H, s), 6.57 (1H, dd, J = 2.6,8.7Hz), 6.70 (1H, d, J = 2.6Hz), 7.25-7.29 (1H, m), 7.38 (1H, d, J 8.7 Hz =), 7.61 (1H, br), 7.77-7.80 (1H, yd), 8.28 (1H, s), 8.50 (1H, br), 8.66 (1H, br), 9.25 (1H, br)
ESI +: 497

 

Seventh Step N- {2- methoxy-4- [4- (4-methylpiperazin-1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] – 1,3,5-triazine-2,4-diamine (formula (1) compounds) synthesis
of 1- [3-methoxy-4 – ({4- [2- (propane-2-sulfonyl) anilino] -1 , 3,5-triazin-2-yl} amino) phenyl] piperidin-4-one (60.1kg), methylpiperazine (24.2kg), was mixed with toluene (500 kg) and acetic acid (28.4kg), 1 hour at room temperature It stirred. To this mixture solution was added sodium triacetoxyborohydride (51.4kg), and stirred at room temperature for 17 hours. To the reaction mixture, methanol (47.5kg) and water (180.1kg) was added, extraction to give an organic layer and an aqueous layer 1. The organic layer was re-extracted by adding water (60.0kg), to obtain an aqueous layer 2. After mixing the aqueous layer 1 and aqueous layer 2 was extracted by adding isopropyl acetate (523.4kg). The resulting aqueous layer methanol (522.3kg), a mixed solution of 48% sodium hydroxide (60.6kg) and water (112.7kg) was added, as a seed crystal N- {2- methoxy-4- [4- (4 – methyl-1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-crystal of diamine (about 6 g) were inoculated, after stirring at room temperature for 2 hours, added water (660.2kg), further stirred for 3.5 hours at room temperature, the precipitated crystals were collected by filtration. The obtained crystals were washed with a mixed solution of methanol (104.4kg) and water (132.0kg), and dried in vacuo at 50 ℃, N- {2- methoxy-4- [4- (4-methylpiperazin- 1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (54.2kg, yield: 77.1 %) was obtained as A06-form crystals.

 

D1: 1.31 (6H, d, J = 6.8Hz), 1.59-1.78 (2H, m), 1.90-2.01 (2H, m), 2.24-2.80 (11H, m), 2.30 (3H, s), 3.19- 3.32 (1H, m), 3.65-3.75 (2H, m), 3.88 (3H, s), 6.50-6.59 (2H, m), 7.18-7.30 (1H, m), 7.53-7.70 (2H, m), 7.88 (1H, dd, J = 1.5,8.3Hz), 8.10 (1H, br), 8.37 (1H, br), 8.53 (1H, br), 9.29 (1H, s)
ESI +: 581

 

 N- {2- methoxy-4- [4- (4-methylpiperazin-1-yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3 , purification step by recrystallization 5-triazine-2,4-diamine (compound of formula (1))
N-{2-methoxy-4- [4- (4-methylpiperazin-1-yl) piperidine-1 yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (54.3kg), activated charcoal (5.4 kg), 2-butanone (1046.1 kg) were mixed, stirred for 1 hour at 75 ° C., was subjected to active carbon filtration. The filtrate activated carbon (5.4kg) in addition to, and the mixture was stirred for 1 hour at 75 ℃, was activated carbon filtration. The filtrate activated carbon (5.4kg) in addition to, and the mixture was stirred for 1 hour at 75 ℃, was activated carbon filtration. After filtrate was total volume approximately until 430L normal pressure concentrated and cooled to 70 ° C., as a seed crystal N- {2- methoxy-4- [4- (4-methylpiperazin-1-yl) piperidine-1 yl] phenyl} -N ‘- inoculated with [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-crystalline diamine (approximately 5 g), after stirring for 3 hours, It was cooled to 5 ℃ at a rate of 20 ℃ / h, and the precipitated crystals were collected by filtration. After washing with the resulting crystals were cooled in 5 of 5 ° C. 2-butanone (220L), and dried in vacuo at 50 ℃, N- {2- methoxy-4- [4- (4-methylpiperazin-1- yl) piperidin-1-yl] phenyl} -N ‘- [2- (propane-2-sulfonyl) phenyl] -1,3,5-triazine-2,4-diamine (42.6kg, 78.5% yield, purity 99.5%) was obtained as A04-form crystals (A04 type ratio 100%).
Ken Jones, president and chief executive officer, Astellas Pharma Europe

Paper

Organic Process Research & Development (2015), 19(12), 1966-1972

Strategy for Controlling Polymorphism of Di(Arylamino) Aryl Compound ASP3026 and Monitoring Solution Structures via Raman Spectroscopy

Technology Process Chemistry Laboratories, Astellas Pharma Inc., 160-2 Akahama, Takahagi, Ibaraki 318-0001,Japan
Astellas Pharma Tech Co., Ltd., 160-2 Akahama, Takahagi, Ibaraki 318-0001, Japan
§ Department of Chemical Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo 184-8588, Japan
Org. Process Res. Dev., 2015, 19 (12), pp 1966–1972
DOI: 10.1021/acs.oprd.5b00208
Publication Date (Web): October 23, 2015
Copyright © 2015 American Chemical Society
*E-mail:kazuhiro.takeguchi@astellas.com. Tel.: +81-293-23-5459. Fax: +81-293-23-5993.

Abstract

Abstract Image

ASP3026(N-{2-Methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}-N′-[2-(propane-2-sulfonyl)phenyl]-1,3,5-triazine-2,4-diamine) was developed as a novel and selective inhibitor of the fusion protein EML4-ALK. Five polymorphs of ASP3026 (A01, A02, A03, A04, and A05) as well as a hydrate have been identified to date, and the most stable polymorph (A04) was selected for designing solid formulations. The influence of crystallization process parameters on nucleation of A03 and A04 was clarified for process development. A04 was obtained at relatively high temperatures and A03 at relatively low temperatures, regardless of the superaturation ratio. A03 and A04 were therefore able to be selectively obtained via temperature control, possibly due to temperature-dependent variations in the concentrations of conformers in solution. The relationship between polymorphs and solution structures before nucleation was investigated using in situ Raman spectroscopy. The relationship with the intensity ratios of nine Raman bands of both polymorphs and ASP3026 solution structures was investigated in detail. Our findings suggest that the solution structure shifted from a structure similar to that of A04 to one similar to that of A03 with decreasing temperature.

Chairman of Astellas Pharma Inc. Mr. Masafumi Nogimori is conferred with Netherlands Honor – ‘Officer in the Order of Oranje-Nassau’

PAPER

Effect of Temperature and Solvent of Solvent-Mediated Polymorph Transformation on ASP3026 Polymorphs and Scale-up

Technology Process Chemistry Laboratories, Astellas Pharma Inc., 160-2 Akahama, Takahagi, Ibaraki 318-0001,Japan
Astellas Pharma Tech Co., Ltd., 160-2 Akahama, Takahagi, Ibaraki 318-0001, Japan
§ Department of Chemical Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo 184-8588, Japan
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00068
Publication Date (Web): April 28, 2016
Copyright © 2016 American Chemical Society
*Telephone: +81-293-23-5459. Fax: +81-293-23-5993; e-mail:kazuhiro.takeguchi@astellas.com.

Abstract

Abstract Image

ASP3026 (N-{2-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}-N′-[2-(propane-2-sulfonyl)phenyl]-1,3,5-triazine-2,4-diamine) was developed as a novel and selective inhibitor of the fusion protein EML4-ALK. Five polymorphs of ASP3026 (A01, A02, A03, A04, and A05) as well as a hydrate have been identified to date. Process development was conducted for large-scale pilot plant manufacturing, and obtaining the desired polymorph A04 was key after a synthetic route of ASP3026 was selected for scale-up. The effects of temperature and solvent species on induction time of polymorph transformation were investigated using in situ Raman spectroscopy, and selective transformation conditions of A02 to A03 and A04 were examined in detail. A04 was obtained at high temperatures using highly polar non-hydrogen-bond-donating solvents, while A03 was obtained at low temperatures using low-polarity or hydrogen-bond-donating solvents. Further, the desired polymorph A04 was successfully obtained in high purity in first stage scale-up manufacturing. Given these findings, this method of solvent-mediated polymorph transformation may aid in process development for obtaining desired polymorphs.

http://pubs.acs.org/doi/full/10.1021/acs.oprd.6b00068

REFERENCES

1: Awad MM, Shaw AT. ALK Inhibitors in Non-Small Cell Lung Cancer: Crizotinib and Beyond. Clin Adv Hematol Oncol. 2014 Jul;12(7):429-39. PubMed PMID: 25322323.

2: George SK, Vishwamitra D, Manshouri R, Shi P, Amin HM. The ALK inhibitor ASP3026 eradicates NPM-ALK⁺ T-cell anaplastic large-cell lymphoma in vitro and in a systemic xenograft lymphoma model. Oncotarget. 2014 Jul 30;5(14):5750-63. PubMed PMID: 25026277; PubMed Central PMCID: PMC4170597.

3: Mori M, Ueno Y, Konagai S, Fushiki H, Shimada I, Kondoh Y, Saito R, Mori K, Shindou N, Soga T, Sakagami H, Furutani T, Doihara H, Kudoh M, Kuromitsu S. The selective anaplastic lymphoma receptor tyrosine kinase inhibitor ASP3026 induces tumor regression and prolongs survival in non-small cell lung cancer model mice. Mol Cancer Ther. 2014 Feb;13(2):329-40. doi: 10.1158/1535-7163.MCT-13-0395. Epub 2014 Jan 13. PubMed PMID: 24419060.

Patent ID Date Patent Title
US2015150850 2015-06-04 TREATING CANCER WITH HSP90 INHIBITORY COMPOUNDS
US8906885 2014-12-09 Treating cancer with HSP90 inhibitory compounds
US2013338358 2013-12-19 METHOD FOR PRODUCING DI(ARYLAMINO)ARYL COMPOUND AND SYNTHETIC INTERMEDIATE THEREFOR
US2013096100 2013-04-18 DI(ARYLAMINO)ARYL COMPOUND
US2013059855 2013-03-07 CRYSTAL OF DI(ARYLAMINO)ARYL COMPOUND
US2010099658 2010-04-22 DI(ARYLAMINO)ARYL COMPOUND

////ASP3026, EML4-ALK, ASP 3026, ASTELLAS

CC(C)S(=O)(=O)C1=CC=CC=C1NC2=NC=NC(=N2)NC3=C(C=C(C=C3)N4CCC(CC4)N5CCN(CC5)C)OC

 

  • 灌水
  • 赞
  • 美好
  • 顶
  • 顶
  • 顶
  • 开心
  • 路过

 

Share
Follow

Get every new post on this blog delivered to your Inbox.

Join other followers: