DR ANTHONY MELVIN CRASTO,WorldDrugTracker, helping millions, A 90 % paralysed man in action for you, I am suffering from transverse mylitis and bound to a wheel chair, With death on the horizon, this will not stop me, Only God and death can..........
DR ANTHONY MELVIN CRASTO Ph.D ( ICT, Mumbai), INDIA, worlddrugtracker, 29Yrs Exp. in the feld of Organic Chemistry,Working for GLENMARK PHARMA at Navi Mumbai, INDIA. Serving chemists around the world. Helping them with websites on Chemistry.8 Million hits on google, world acclamation from industry, academia, drug authorities for websites, blogs and educational contribution
n, सुकून उतना ही देना प्रभू, जितने से जिंदगी चल जाये।औकात बस इतनी देना,कि औरों का भला हो जाये।...........P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

Wednesday 16 September 2015

TRAMETINIB


Mekinist (trametinib)
JTP-74057, GSK212, GSK1120212
N-{3- [3-cyclopropyl-5- (2-fluoro-4- iodophenylamino) -6 , 8-dimethyl-2 , 4 , 7-trioxo-3 ,4,6, 7-tetrahydro-2H- pyrido [4, 3-d] pyrimidin-1-yl] phenyl}acetamide
Molecular Weight 615.39
Formula C26H23FIN5O4
CAS Number 871700-17-3


Trametinib (GSK1120212) is experimental cancer drug. It is a MEK inhibitor drug with anti-cancer activity.[1]
It inhibits MEK1 and MEK2.[1]
Trametinib had good results for V600E mutated metastatic melanoma in a phase III clinical trial.[2]
  1. Trametinib, NCI Drug Dictionary
  2. METRIC phase III study: Efficacy of trametinib (T), a potent and selective MEK inhibitor (MEKi), in progression-free survival (PFS) and overall survival (OS), compared with chemotherapy (C) in patients (pts) with BRAFV600E/K mutant advanced or metastatic melanoma (MM).
GSK1120212 (JTP-74057) is a potent and selective allosteric inhibitor of the MEK1 and MEK2 (MEK1/2) enzymes with promising antitumor activity in a phase I clinical trial (ASCO 2010). GSK1120212 (JTP-74057) inhibits MEK1/2 kinase activity and prevents Raf-dependent MEK phosphorylation (S217 for MEK1), producing prolonged p-ERK1/2 inhibition. Potent cell growth inhibition was evident in most tumor lines with mutant BRAF or Ras. In xenografted tumor models, GSK1120212 orally dosed once daily had a long circulating half-life and sustained suppression of p-ERK1/2 for more than 24 hours; GSK1120212 also reduced tumor Ki67, increased p27 (Kip1/CDKN1B), and caused tumor growth inhibition in multiple tumor models.
May 29, 2013 —
GlaxoSmithKline plc announced today that the U.S. Food and Drug Administration (FDA) has approved Mekinist (trametinib) as a single-agent oral treatment for unresectable or metastatic melanoma in adult patients with BRAF V600E or V600K mutations. Mekinist is not indicated for the treatment of patients who have received a prior BRAF inhibitor therapy. The mutation must be detected by an FDA-approved test, such as the companion diagnostic assay from bioMérieux S.A., THxID™-BRAF.
 Among those with metastatic melanoma, approximately half have a BRAF mutation, which is an abnormal change in a gene that can enable some melanoma tumours to grow and spread.
Mekinist is approved for patients with the BRAF V600E mutation, which accounts for approximately 85 percent of all BRAF V600 mutations in metastatic melanoma. It is also approved for patients with the V600K mutation, which makes up approximately 10 percent of all BRAF V600 mutations in metastatic melanoma.
Melanoma is the most serious and deadly form of skin cancer.[iii] According to statistics from the National Cancer Institute, in 2013 there will be an estimated 9,480 deaths resulting from melanoma in the United States.[iv] When melanoma spreads in the body, the disease is called metastatic melanoma.[v] Approximately half of all people with metastatic melanoma have a BRAF mutation, which is an abnormal change in a gene that can enable some melanoma tumours to grow and spread.2 One in two patients worldwide with metastatic melanoma is expected to survive for a year after diagnosis,while in the U.S., the five-year survival rate was 16 percent (2003-2009). The median age of a newly diagnosed metastatic melanoma patient is almost a decade younger than other cancers.

Mekinist (trametinib) is now approved for the treatment of adult patients with unresectable or metastatic melanoma with BRAF V600E and V600K mutations as detected by an FDA-approved test. Limitation of use: Mekinist is not indicated for the treatment of patients who have received a prior BRAF inhibitor therapy.


(WO2005121142A1). Aniline a reaction with CDI was added cyclopropylamine get two , two and malonic acid cyclization get 3 . 3 chlorination with phosphorus oxychloride reaction with methylamine 4 , as well as byproducts 5 (ratio of 2:1). Mixture 4 + 5 and acid 6 crystals obtained after cyclization compound 7 (pure substance). 7 with activated trifluoromethanesulfonyl chloride to the amide 8 SNAr reaction occurs 9 , 9 in alkaline conditions rearrangement trimetazidine imatinib.
nmr5

nmr4



…………………
http://www.google.com/patents/WO2014039375A1?cl=en
The term “trametinib” as used herein means the MEK inhibitor represented by the structure of formula (I):
Figure imgf000005_0002
or a pharmaceutically acceptable salt or solvate thereof. Trametinib is preferably administered as a solvate in the form of N-{3-[3- cyclopropyl-5-(2-fluoro-4-iodo-phenylamino)6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydro- 2H-pyrido[4,3-d]pyrimidin-1 -yl]phenyl}acetamide dimethyl sulfoxide (solvate).
Depending on naming convention, the compound of formula (I) may also properly be referred to as N-{3-[3-cyclopropyl-5-[(2-fluoro-4-iodophenyl)amino]-6,8-dimethyl-2,4,7- trioxo-3,4,6,7-tetrahydropyrido[4,3-d]pyrimidin-1(2H)-yl]phenyl}acetamide.
Trametinib is disclosed and claimed, along with pharmaceutically acceptable salts thereof, and also as solvates thereof, as being useful as an inhibitor of MEK activity, particularly in treatment of cancer, in WO 2005/121 142. Trametinib can be prepared as described in WO 2005/121 142.
Suitably, trametinib is in the form of a dimethyl sulfoxide solvate. Suitably, trametinib is in the form of a sodium salt. Suitably, trametinib is in the form of a solvate selected from: hydrate, acetic acid, ethanol, nitromethane, chlorobenzene, 1 -pentancol, isopropyl alcohol, ethylene glycol and 3-methyl-1 -butanol. These solvates and salt forms can be prepared by one of skill in the art from the description in WO2005/121 142.

…………………….
http://www.google.com/patents/WO2005121142A1?cl=en
Example 3-10 By treating N-{3- [3-cyclopropyl-5- (2-fluoro-4- iodophenylamino) -6 , 8-dimethyl-2 , , 7-trioxo-3 ,4,6, 7-tetrahydro-2H- pyrido [4 , 3-d]pyrimidin-1-yl] phenyl Jmethanesulfonamide 46 according to conventional methods, sodium salt and potassium salt thereof were obtained.
N-{3- [3-cyclopropyl-5- (2-fluoro-4-iodophenylamino) -6 , 8-dimethyl- 2,4, 7-trioxo-3 ,4,6, 7-tetrahydro-2H-pyrido [4 , 3-d]pyrimidin-1- yl]phenylJmethanesulfonamide sodium salt:
^-NMR (DMSO-de, 300 MHz) δ 0.47 (brs, 2H) , 0.70-0.90 (m, 2H) , 1.23(s, 3H) , 2.35(brs, IH) , 2.82(s, 3H) , 3.22(s, 3H) , 6.69(t, J=8.8Hz, IH) , 6.81 (d, J=8.1Hz , IH) , 6.98 (s, IH) , 7.02 (d, J=8.8Hz , IH) , 7.10-7.30 (m, 2H) , 7.38(d, J=9.2Hz , IH) , 10.22(brs, IH) . MS (ESI) m/z 652 [MH]+.
N-{3-[3-cyclopropyl-5- (2-fluoro-4-iodophenylamino) -6 , 8-dimethyl- 2,4, 7-trioxo-3 ,4,6, 7-tetrahydro-2H-pyrido [4 , 3-d]pyrimidin-1- yl]phenylJmethanesulfonamide potassium salt: Example 4-1
N-{3- [3-cyclopropyl-5- (2-fluoro-4-iodophenylamino) -6 ,8-dimethyl- 2,4, 7-trioxo-3 ,4,6, 7-tetrahydro-2H-pyrido [4 , 3-d]pyrimidin-1-yl] – phenyl}-acetamide Step 1 Synthesis of l-cyclopropyl-3- (2-fluoro-4-iodo-phenyl) rea
Figure imgf000146_0001
47 48 Under a nitrogen atmosphere, to N,N-carbonyldiimidazole (39.9 g) were added N,N-dimethylformamide (200 ml) and triethylamine (34.3 ml) and a solution of 2-fluoro-4-iodoaniline 47 (48.5 g) in N,N-dimethylformamide (50 ml) was added dropwise with stirring under ice-cooling. After the completion of the dropwise addition, the mixture was stirred at room temperature for 18 hrs. The reaction mixture was ice-cooled, and cyclopropylamine (21.3 ml) was added dropwise. The reaction mixture was stirred at room temperature for 1 hr and added dropwise to water-toluene [2:1 (volume ratio), 750 ml] with stirring. The precipitated crystals were collected by filtration and dried to give l-cyclopropyl-3- (2-fluoro-4-iodophenyl) urea 48 (61.3 g, yield 93.4%) as colorless crystals. Step 2 Synthesis of l-cyclopropyl-3- (2-fluoro-4- iodophenyl) pyrimidine-2 , 4 , 6-trione
Figure imgf000147_0001
To l-cyclopropyl-3- (2-fluoro-4-iodophenyl) urea 48 (61.0 g) obtained in Step 1 and malonic acid 4 (19.9 g) were added acetic anhydride (300 ml) and acetyl chloride (27.2 ml), and the mixture was stirred under a nitrogen atmosphere at 60°C for 3 hrs. After allowing to cool to room temperature, the reaction mixture was added dropwise to water-toluene [2:1 (volume ratio), 900 ml] with stirring. The precipitated crystals were collected by filtration and dried to give l-cyclopropyl-3- (2-fluoro-4- iodophenyl)pyrimidine-2,4, 6-trione 49 (60.9 g, yield 82%) as pale-yellow crystals.
Step 3 Synthesis of 6-chloro-3-cyclopropyl-l- (2-fluoro-4- iodophenyl) -lH-pyrimidine-2 , -dione
Figure imgf000147_0002
49 50 51 To l-cyclopropyl-3- (2-fluoro-4-iodophenyl) -pyrimidine-
2, 4, 6-trione 49 (59.0 g) obtained in Step 2 were added phosphorus oxychloride (85.0 ml) and dimethylaniline (29.0 ml), and water (8.3 ml) was added dropwise to the mixture at room temperature with stirring. After the completion of the dropwise addition, the mixture was stirred with heating at 110°C for 1 hr. After allowing to cool to room temperature, the reaction mixture was added dropwise to ice water-toluene [2:1 (volume ratio), 900 ml] with stirring. The mixture was stirred at room temperature for 1 hr. The organic layer was separated, and washed successively with water (300 ml) and brine (300 ml) . Anhydrous magnesium sulfate and activated carbon were added and the mixture was stirred. Anhydrous magnesium sulfate and activated carbon were filtered off, and the filtrate was concentrated under reduced pressure to give a 1:2 mixture (62.9 g) of 6-chloro-3-cyclopropyl-l- (2- fluoro-4-iodophenyl) -lH-pyrimidine-2 , 4-dione 50 and 6-chloro-l- cyclopropyl-3- (2-fluoro-4-iodophenyl) -lH-pyrimidine-2 , 4-dione 51 as a yellow foamy oil, which was used for the next step without purification.
Step 4 Synthesis of 3-cyclopropyl-l- (2-fluoro-4-iodophenyl) -6- methylamino-lH-pyrimidine-2 , 4-dione
Figure imgf000148_0001
To a 1:2 mixture (62.9 g) of 6-chloro-3-cyclopropyl-l- (2- fluoro-4-iodophenyl) -lH-pyrimidine-2 ,4-dione 50 and 6-chloro-l- cyclopropyl-3- (2-fluoro-4-iodophenyl) -lH-pyrimidine-2 , -dione 51 obtained in Step 3 were added methanol (189 ml) and a solution (126 ml) of 40% methylamine in methanol, and the mixture was stirred at room temperature for 2 hrs . The precipitated crystals were filtered off and the filtrate was concentrated under reduced pressure. The residue was extracted with chloroform (200 ml) and water (200 ml) , and the organic layer was washed with brine (200 ml) and dried over anhydrous magnesium sulfate. Anhydrous magnesium sulfate was filtered off and the filtrate was concentrated under reduced pressure to give a 2:1 mixture (34.55 g) of 3-cyclopropyl-l- (2-fluoro-4-iodophenyl) -6-methylamino-lH- pyrimidine-2 ,4-dione 52 and l-cyclopropyl-3- (2-fluoro-4- iodophenyl) -6-methylamino-lH-pyrimidine-2,4 ,-dione 53 as yellow crystals, which were used for the next step without purification. Step 5 Synthesis of 3-cyclopropyl-l- (2-fluoro-4-iodophenyl) -5- hydroxy-6 , 8-dimethyl-lH, 8H-pyrido [2 , 3-d] pyrimidine-2 , 4 , 7-trione
Figure imgf000149_0001
To a 2:1 mixture (34.6 g) of 3-cyclopropyl-l- (2-fluoro-4- iodophenyl) -6-methylamino-lH-pyrimidine-2, 4-dione 52 and 1- cyclopropyl-3- (2-fluoro-4-iodo-phenyl) 6-methylamino-lH- pyrimidine-2,4,-dione 53 obtained in Step 4, and 2-methylmalonic acid 54 (10.2 g) was added acetic anhydride (173 ml) , and the mixture was stirred at 100°C for 2 hrs. After allowing to cool to room temperature, the reaction mixture was concentrated under reduced pressure. Acetone (104 ml) was added to the residue, and the mixture was stirred with heating under reflux for 30 min. After allowing to cool to room temperature, the precipitated crystals were collected by filtration and dried to give 3- cyclopropyl-1- (2-fluoro-4-iodophenyl) -5-hydroxy-6 , 8-dimethyl- lH,8H-pyrido [2, 3-d] pyrimidine-2, 4, 7-trione 55 (15.1 g, yield from 48, 21%) as colorless crystals.
Step 6 Synthesis of trifluoromethanesulfonic acid 3-cyclopropyl- 1- (2-fluoro-4-iodophenyl) -6 , 8-dimethyl-2 , 4 , 7-trioxo-l ,2,3,4,7,8- hexahydro-pyrido [2 , 3-d]pyrimidin-5-yl ester
Figure imgf000149_0002
55 56 43 Under a nitrogen atmosphere, to 3-cyclopropyl-l- (2-fluoro-
4-iodophenyl) -5-hydroxy-6 , 8-dimethyl-lH, 8H-pyrido [2,3- d] pyrimidine-2 ,4 ,7-trione 55 (33.0 g) obtained in Step 5 were added chloroform (165 ml) and 2 , 6-lutidine (10.4 ml), and trifluoromethanesulfonic anhydride 56 (14.4 ml) was added dropwise under ice-cooling with stirring. After the completion of the dropwise addition, the mixture was stirred at same temperature for 30 min and at room temperature for 2 hrs. The reaction mixture was washed successively with aqueous sodium hydrogen carbonate (165 ml) , IN hydrochloric acid (165 ml) and brine (165 ml) and dried over anhydrous magnesium sulfate. Anhydrous magnesium sulfate was filtered off and the filtrate was concentrated under reduced pressure. 2-Propanol (198 ml) was added to the residue, and the mixture was stirred with heating under reflux, and allowed to return to room temperature. The crystals were collected by filtration and dried to give trifluoromethanesulfonic acid 3-cyclopropyl-l- (2-fluoro-4- iodophenyl) -6 , 8-dimethyl-2 , 4 , 7-trioxo-l ,2,3,4,7, 8-hexahydro- pyrido [2 , 3-d] pyrimidin-5-yl ester 43 (31.9 g, yield 93%) as colorless crystals.
Step 7 Synthesis of N-{3- [3-cyclopropyl-l- (2-fluoro-4- iodophenyl) -6 , 8-dimethyl-2 ,4 , 7-trioxo-l ,2,3,4,7 , 8-hexahydro- pyrido [2 , 3-d] pyrimidin-5-ylamino] phenyl } acetamide
Figure imgf000150_0001
To trifluoromethanesulfonic acid 3-cyclopropyl-l- (2-fluoro-
4-iodophenyl) -6 , 8-dimethyl-2 , 4 , 7-trioxo-l ,2,3,4,7, 8-hexahydro- pyrido [2 , 3-d] pyrimidin-5-yl ester 43 (25.0 g) obtained in Step 6 and 3 ‘-aminoacetanilide 57 (7.33 g) were added N,N- dimethylacetamide (50.0 ml) and 2,6-lutidine (5.68 ml), and the mixture was stirred at 130°C for 5 hrs. After allowing to cool to room temperature, methanol-water [1:2 (volume ratio), 150 ml] was added with stirring. The crystals were collected by filtration and dried to give N- {3- [3-cyclopropyl-l- (2-fluoro-4-iodophenyl) – 6 , 8-dimethyl-2 , 4 , 7-trioxo-l ,2,3,4,7, 8-hexahydro-pyrido [2,3- d]pyrimidin-5-ylamino] phenyl}acetamide 58 (24.8 g, yield 99%) as colorless crystals. Step 8 Synthesis of N- { 3- [3-cyclopropyl-5- (2-fluoro-4- iodophenylamino) -6 , 8-dimethyl-2 , 4″, 7-trioxo-3 , 4 , 6 , 7-tetrahydro-2H- pyrido [4 , 3 -d]pyrimidin-l-yl] phenyl} acetamide
Figure imgf000151_0001
Under a nitrogen atmosphere, to a solution (1.57 g) of 28% sodium methoxide in methanol was added tetrahydrofuran (40 ml) , N- {3- [3-cyclopropyl-l- (2-fluoro-4-iodophenyl) -6 , 8-dimethyl-2 ,4,7- trioxo-1 ,2,3,4,7, 8-hexahydro-pyrido [2 , 3-d] pyrimidin-5- ylamino]phenyl}acetamide 58 (5.00 g) obtained in Step 7 was added, and the mixture was stirred at room temperature for 4 hrs. Acetic acid (0.56 ml) was added, and the mixture was stirred at room temperature for 30 min. Water (40 ml) was added and the mixture was further stirred for 1 hr. The crystals were collected by filtration and dried to give N-{3- [3-cyclopropyl-5- (2-fluoro-4- iodophenylamino) -6 , 8-dimethyl-2 , 4 , 7-trioxo-3 ,4,6, 7-tetrahydro-2H- pyrido [4, 3-d] pyrimidin-1-yl] phenyl}acetamide 59 (4.75 g, yield 95%) as colorless crystals. MS ESI m/e: 616 (M+H) , 614 (M-H) .
1H-NMR(DMSO-d6, 400MHz) δ 0.63-0.70 (m, 2H) , 0.91-1.00 (m, 2H) , 1.25(s, 3H) , 2.04(s, 3H) , 2.58-2.66(m, IH) , 3.07(s, 3H) , 6.92(t,
J=8.8Hz, IH) , 7.00-7.05 (m, IH) , 7.36 (t, J=8.2Hz , IH) , 7.52-7.63 (m,
3H) , 7.79(dd, J=2.0 , 10.4Hz, IH) , 10.10(s, IH) , 11.08(s, IH) .

Example 4-1 (alternative method)

N-{3- [3-cyclopropyl-5- (2-fluoro-4-iodophenylamino) -6 , 8-dimethyl- 2,4, 7-trioxo-3 ,4,6, 7-tetrahydro-2H-pyrido [4 , 3-d] pyrimidin-1-yl] – phenyl } -acetamide
Step 1 Synthesis of l-cyclopropyl-3- (2-fluoro-4-iodo-phenyl) -urea
Figure imgf000152_0001
48 47 Under a nitrogen atmosphere, to N,N-carbonyldiimidazole (82.1 g) were added N, N-dimethylformamide (400 ml) and triethylamine (70.5 ml) , and a solution of 2-fluoro-4-iodoaniline 47 (100 g) in N, N-dimethylformamide (100 ml) was added dropwise under ice-cooling. After the completion of the dropwise addition, the mixture was stirred at room temperature for 5 hrs . The reaction mixture was ice-cooled, and cyclopropylamine (44.0 ml) was added dropwise. The mixture was stirred at room temperature for 1 hr, and the reaction mixture was added dropwise to water- toluene [2:1 (volume ratio) , 1500 ml] with stirring. The precipitated crystals were collected by filtration and dried to give l-cyclopropyl-3- (2-fluoro-4-iodo-phenyl) -urea 48 (129 g, yield 95.5%) as colorless crystals. Step 2 Synthesis of 1- (2-cyano-acetyl) -l-cyclopropyl-3- (2-fluoro- 4-iodo-phenyl) -urea
Figure imgf000152_0002
48 73 74 Under a nitrogen atmosphere, to l-cyclopropyl-3- (2-fluoro- 4-iodo-phenyl) -urea 48 (167 g) and cyanoacetic acid 73 (80.0 g) , was added N, N-dimethylformamide (836 ml) , and methanesulfonyl chloride (72.8 ml) was added dropwise with stirring at room temperature. The mixture was stirred at room temperature for 4 hrs. The reaction mixture was cooled with water, and water- isopropanol [2:1 (volume ratio) , 1670 ml] was added dropwise. The mixture was stirred under water-cooling for 1 hr, and the precipitated crystals were collected by filtration and dried to give 1- (2-cyano-acetyl) -l-cyclopropyl-3- (2-fluoro-4-iodo-phenyl) – urea 74 (192 g) .
Step 3 Synthesis of 6-amino-3-cyclopropyl-l- (2-fluoro-4-iodo- phenyl) -lH-pyrimidine-2 , 4-dione
Figure imgf000153_0001
74 75 To 1- (2-cyano-acetyl) -l-cyclopropyl-3- (2-fluoro-4-iodo- phenyl)-urea 74 (192 g) were added water (962 ml) and 2N aqueous sodium hydroxide solution (24.9 ml) , and the mixture was stirred with heating at 80°C for 1 hr. After allowing to cool to room temperature, the crystals were collected by filtration and dried to give 6-amino-3-cyclopropyl-l- (2-fluoro-4-iodo-phenyl) -1H- pyrimidine-2, 4-dione 75 (178g, yield from 48, 88%) as pale-yellow crystals . Step 4 Synthesis of N’- [l-cyclopropyl-3- (2-fluoro-4-iodo-phenyl) – 2 , 6-dioxo-l ,2,3, 6-tetrahydro-pyrimidin-4-yl] -N ,N-dimethyl- formamidine
Figure imgf000153_0002
75 76 Under a nitrogen atmosphere, to 6-amino-3-cyclopropyl-l- (2- fluoro-4-iodo-phenyl) -lH-pyrimidine-2 , 4-dione 75 (178 g) were added N,N-dimethylformamide (356 ml) and N,N-dimethylformamide dimethylacetal (178 ml) , and the mixture was stirred at room temperature for 2 hrs. Isopropanol (178 ml) was added with stirring at room temperature, and water (1068 ml) was added dropwise. The mixture was stirred at room temperature for 2 hrs, and the precipitated crystals were collected by filtration and dried to give N’- [l-cyclopropyl-3- (2-fluoro-4-iodo-phenyl) -2,6- dioxo-1 ,2,3, 6-tetrahydro-pyrimidin-4-yl] -N,N-dimethyl-formamidine 76 (188 g, yield 92%) as yellow crystals.
Step 5 Synthesis of 3-cyclopropyl-l- (2-fluoro-4-iodo-phenyl) -6- methylamino- lH-pyrimidine-2 ,4-dione
Figure imgf000154_0001
76 52 Under a nitrogen atmosphere, to t-butanol-ethanol [2:1 (volume ratio) , 250 ml] was added sodium borohydride (6.41 g) , and the mixture was stirred at room temperature for 1 hr. Under water-cooling, N’- [l-cyclopropyl-3- (2-fluoro-4-iodo-phenyl) -2 ,6- dioxo-1 ,2,3, 6-tetrahydro-pyrimidin-4-yl] -N,N-dimethy1-formamidine 76 (50.0 g) was added, and the mixture was stirred for 2.5 hrs. Under water-cooling, water (225 ml) and 10% aqueous citric acid solution (175 ml) were successively added dropwise, and the mixture was stirred for 3 hrs . The precipitated crystals were collected by filtration and dried to give crude crystals (34.5 g, LC purity 91%) of 3-cyclopropyl-l- (2-fluoro-4-iodo-phenyl) -6- methylamino- lH-pyrimidine-2 ,4-dione 52, which were used for the next reaction without purification. Step 6 Synthesis of 3-cyclopropyl-l- (2-fluoro-4-iodo-phenyl) -5- hydroxy-6 , 8-dimethyl-lH, 8H-pyrido [2 , 3-d] pyrimidine-2 ,4 ,7-trione
Figure imgf000154_0002
Under a nitrogen atmosphere, to 3-cyclopropyl-l- (2-fluoro- 4-iodo-phenyl) -6-methylamino-lH-pyrimidine-2, -dione 52 (34.4 g) and 2-methyl-malonic acid 54 (15.2 g) was added acetic anhydride (34.4 ml) , and the mixture was stirred with heating at 100°C for 3 hrs. After allowing to cool to 50°C, acetone (68.8 ml) was added dropwise, and the mixture was stirred as it was for 30 min. Water (172 ml) was further added dropwise, and the mixture was stirred for 1 hr. After allowing to cool to room temperature with stirring, the precipitated crystals were collected by filtration and dried to give crude crystals (37.7 g, LC purity 91%) of 3- cyclopropyl-1- (2-fluoro-4-iodo-phenyl) -5-hydroxy-6 , 8-dimethyl- lH,8H-pyrido [2 ,3-d] pyrimidine-2 ,4 ,7-trione 55. Isopropanol (92.0 ml) was added to the obtained crude crystals (30.7 g) , and the mixture was stirred at room temperature for 4 hrs . The crystals were collected by filtration and dried to give 3-cyclopropyl-l- (2-fluoro-4-iodo-phenyl) -5-hydroxy-6 , 8-dimethyl-lH, 8H-pyrido [2 ,3- d] pyrimidine-2 ,4 ,7-trione 55 (25.9 g, yield from 76, 58%) as pale-yellow crystals.
Step 7 Synthesis of p-toluenesulfonic acid 3-cyclopropyl-l- (2- fluoro-4-iodo-phenyl) -6 , 8-dimethyl-2 , , 7-trioxo-l ,2,3,4,7,8- hexahydro-pyrido [2 , 3-d]pyrimidin-5-yl ester
Figure imgf000155_0001
55 11 77 Under a nitrogen atmosphere, to 3-cyclopropyl-l- (2-fluoro- 4-iodo-phenyl) -5-hydroxy-6 ,8-dimethyl-lH,8H-pyrido [2,3- d] pyrimidine-2 ,4,7-trione 55 (23.9 g) was added acetonitrile (167 ml) , and the mixture was stirred under ice-cooling. Triethylamine (11.0 ml) and trimethylamine hydrochloride (2.37 g) were added, and a solution of p-toluenesulfonyl chloride 11 (12.3 g) in acetonitrile (72.0 ml) was added dropwise. The mixture was stirred under ice-cooling for 1 hr, and stirred at room temperature for 3 hrs. Methanol (239 ml) was added, and the mixture was stirred at room temperature for 1 hr. The crystals were collected by filtration and dried to give p-toluenesulfonic acid 3-cyclopropyl-l- (2-fluoro-4-iodo-phenyl) -6 , 8-dimethyl-2 ,4,7- trioxo-1 ,2,3,4,7, 8-hexahydro-pyrido [2 ,3-d]pyrimidin-5-yl ester 77 (28.7 g, yield 91%) as colorless crystals.
Step 8 Synthesis of N-{3- [3-cyclopropyl-l- (2-fluoro-4-iodo- phenyl) -6 , 8-dimethyl-2 , 4 , 7-trioxo-l ,2,3,4,7, 8-hexahydro- pyrido [2 ,3-d]pyrimidin-5-ylamino] -phenyl}-acetamide
Figure imgf000156_0001
To p-toluenesulfonic acid 3-cyclopropyl-l- (2-fluoro-4-iodo- phenyl) -6 , 8-dimethyl-2 ,4,7- trioxo-1 ,2,3,4,7, 8-hexahydro- pyrido [2,3-d]pyrimidin-5-yl ester 77 (28.0 g) and 3′- aminoacetanilide 57 (13.2 g) were added N,N-dimethylacetamide (84.0 ml) and 2,6-lutidine (15.3 ml), and the mixture was stirred at 130°C for 4 hrs. After allowing to cool with stirring, methanol (196 ml) was added dropwise, and the mixture was stirred at room temperature. The crystals were collected by filtration and dried to give N-{3- [3-cyclopropyl-l- (2-fluoro-4-iodo-phenyl) – 6 , 8-dimethyl-2 , , 7-trioxo-l ,2,3,4,7, 8-hexahydro-pyrido [2,3- d]pyrimidin-5-ylamino] -phenyl}-acetamide 58 (25.2 g, yield 93%) as colorless crystals. Step 9 Synthesis of N-{3- [3-cyclopropyl-5- (2-fluoro-4-iodo- phenylamino) -6 , 8-dimethyl-2 ,4 , 7-trioxo-3 ,4,6, 7-tetrahydro-2H- pyrido [4 ,3-d] pyrimidin-1-yl] -phenyl}-acetamide
Figure imgf000156_0002
58 59 Under a nitrogen atmosphere, to N-{3- [3-cyclopropyl-l- (2- fluoro-4-iodo-phenyl) -6 , 8-dimethyl-2 ,4 , 7-trioxo-l ,2,3,4,7,8- hexahydro-pyrido [2 ,3-d]pyrimidin-5-ylamino] -phenyl }-acetamide 58 (45.7 g) was added tetrahydrofuran (366 ml), and a solution (15.7 g) of 28% sodium methoxide in methanol was added dropwise with stirring at room temperature and the mixture was stirred at room temperature for 4 hrs. Acetic acid (5.61 ml) was added, and the mixture was stirred at room temperature for 30 min. With stirring at 70°C in an oil bath, water (366 ml) was added dropwise, and the mixture was stirred for 1 hr. After allowing to cool with stirring, the crystals were collected by filtration and dried to give crystal 1 (46.0 g) of N-{3- [3-cyclopropyl-5- (2-fluoro-4- iodo-phenylamino) -6 , 8-dimethyl-2 ,4 , 7-trioxo-3 ,4,6, 7-tetrahydro- 2H-pyrido [4 , 3-d]pyrimidin-1-yl] -phenyl}-acetamide 59. N,N-Dimethylacetamide (184 ml) was added to crystal 1 (46.0 g) , and the mixture was stirred with heating at 130°C. After complete dissolution, the solution was filtered by suction using with paper (5B) , and washed with N,N-dimethylacetamide (92.0 ml).
The filtrate was stirred under heating at 130°C, 1-butanol (138 ml) and water (96.0 ml) were successively added dropwise, and the mixture was stirred for 30 min. Water (46.0 ml) was further added dropwise, and the mixture was stirred for 30 min allowed to cool with stirring. The crystals were collected by filtration and dried to give crystal 2 (41.7 g) of N-{3- [3-cyclopropyl-5- (2- fluoro-4-iodo-phenylamino) -6 , 8-dimethyl-2 , , 7-trioxo-3 ,4,6,7- tetrahydro-2H-pyrido [ ,3-d]pyrimidin-l-yl] -phenyl}-acetamide 59 as colorless crystals. To crystal 2 (41.5 g) was added 1-butanol-water [19:1 (volume ratio) , 415 ml] , and the mixture was stirred at 130°C for 18 hrs. After allowing to cool with stirring, the crystals were collected by filtration and dried to give N-{3- [3-cyclopropyl-5- (2-fluoro-4-iodo-phenylamino) -6 , 8-dimethyl-2 , 4 , 7-trioxo-3 ,4,6,7- tetrahydro-2H-pyrido [4 ,3-d]pyrimidin-1-yl] -phenyl}-acetamide 59 (40.7 g, yield 89%) as colorless crystals
References
Combination Small Molecule MEK and PI3K Inhibition Enhances Uveal Melanoma Cell Death in a Mutant GNAQ- and GNA11-Dependent Manner.
Khalili JS et al. Clin Cancer Res. 2012 Aug 15;18(16):4345-55. PMID: 22733540.
Comprehensive predictive biomarker analysis for MEK inhibitor GSK1120212.
Jing J et al. Mol Cancer Ther. 2012 Mar;11(3):720-9. PMID: 22169769.
Antitumor activities of JTP-74057 (GSK1120212), a novel MEK1/2 inhibitor, on colorectal cancer cell lines in vitro and in vivo.
Yamaguchi T et al. Int J Oncol. 2011 Jul;39(1):23-31. PMID: 21523318.
GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition.
Gilmartin AG et al. Clin Cancer Res. 2011 Mar 1;17(5):989-1000. PMID: 21245089.
//////
nmr5

Share this:

 



 DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE




Join me on Linkedin

View Anthony Melvin Crasto Ph.D's profile on LinkedIn

Join me on Facebook FACEBOOK
Join me on twitterFollow amcrasto on Twitter     
Join me on google plus Googleplus

 amcrasto@gmail.com

09b37-misc2b027LIONEL MY SON
He was only in first standard in school when I was hit by a deadly one in a million spine stroke called acute transverse mylitis, it made me 90% paralysed and bound to a wheel chair, Now I keep him as my source of inspiration and helping millions, thanks to millions of my readers who keep me going and help me to keep my son happy

GSK 1059615, GSK 615

GSK1059615.png
GSK1059615; 958852-01-2; GSK-1059615; UNII-07YMO87363;
  • GSK 615
(5Z)-5-[(4-pyridin-4-ylquinolin-6-yl)methylidene]-1,3-thiazolidine-2,4-dione
5-[[4-(4-Pyridinyl)-6-quinolinyl]methylene]-2,4-thiazolidenedione
C18H11N3O2S
MOLECULAR WEIGHT:333.36384
CAS 958852-01-2
GSK1059615 is a potent, ATP-competitive inhibitor of PI 3-kinase alpha (PI3Kα) with IC50 of 2 nM. Phosphatidylinositol-3 kinases (PI3K) are critical for malignant cellular processes including growth, proliferation, and survival. GSK1059615 is also a novel inhibitor of PI3Kβ, PI3Kδ, PI3Kγ and mTOR with IC50 of 0.6 nM, 2 nM, 5 nM and 12 nM, respectively. GSK1059615 (25 mg/kg) effectively inhibits tumor growth in xenograft mice models of BT474 or HCC1954 breast cancer cells and attenuates MAPK signaling.
GSK1059615 is a  phosphoinositide 3-kinase (PI3K) inhibitor with potential antineoplastic activity. PI3K inhibitor GSK1059615 inhibits PI3K in the PI3K/AKT kinase signaling pathway, which may trigger the translocation of cytosolic Bax to the mitochondrial outer membrane and an increase in mitochondrial membrane permeability, followed by apoptosis. Bax is a member of the proapoptotic Bcl-2 family of proteins. PIK3, an enzyme often overexpressed in cancer cells, plays a crucial role in tumor cell regulation and survival.
GSK1059615 Structure

GSK 1059615 sodium salt hydrate
Patent
Figure US20090306074A1-20091210-C00010
Figure US20090306074A1-20091210-C00017
Example 1 (5Z)-5-{[4-(4-pyridinyl)-6-quinolinyl]methylidene}-1,3-thiazolidine-2,4-dione
Figure US20090306074A1-20091210-C00007
a) 4-chloro-6-ethenylquinoline
A mixture of 6-bromo-4-chloroquinoline (6.52 g, 26.88 mmol; see J. Med. Chem., 21, 268 (1978)), tributyl(vinyl)tin (8.95 g, 28.22 mmol), and tetrakistriphenylphosphine palladium (0) (0.62 g, 0.54 mmol) in 1,4-dioxane (150 mL) was refluxed for 2.0 h, cooled to room temperature, and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (0-4% MeOH:CH2Cl2) to give the title compound (5.1 g) as a pale yellow solid. MS (ES)+m/e 190 [M+H]+. This material was used directly in the next step.
b) 4-chloro-6-quinolinecarbaldehyde
A mixture of 4-chloro-6-ethenylquinoline (5.1 g, 26.88 mmol), 2,6-lutidine (5.76 g, 53.75 mmol), sodium (meta) periodate (22.99 g, 107.51 mmol), and osmium tetroxide (5.48 g of a 2.5% solution in tert-butanol, 0.538 mmol) in 1,4-dioxane:H2O (350 mL of 3:1 mixture) was stirred for 3.5 h at room temperature and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (CH2Cl2) to give the title compound (4.26 g, 83% for 2 steps) as a pale yellow solid. MS (ES)+ m/e 192 [M+H]+.
c) 4-(4-pyridinyl)-6-quinolinecarbaldehyde
A mixture of 4-chloro-6-quinolinecarbaldehyde (3.24 g, 16.92 mmol), 4-pyridylboronic acid (3.12 g, 25.38 mmol), tetrakistriphenylphosphine palladium (0) (0.978 g, 0.846 mmol), and 2M aqueous K2CO(7.02 g, 50.76 mmol, 25.4 mls of 2M solution) in DMF (100 mL) was heated at 100° C. for 3.0 h and cooled to room temperature. The mixture was filtered through celite and the celite was washed with EtOAc. The filtrate was transferred to a separatory funnel, washed with water and saturated NaCl, dried (Na2SO4), filtered and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (5% MeOH:CH2Cl2) to give the title compound (2.03 g, 51%) as a tan solid. MS (ES)+ m/e 235 [M+H]+.
d) (5Z)-5-{[4-(4-pyridinyl)-6-quinolinyl]methylidene}-1,3-thiazolidine-2,4-dione
A mixture of 4-(4-pyridinyl)-6-quinolinecarbaldehyde (0.108 g, 0.463 mmol), 2,4-thiazolidinedione (0.0417 g, 0.356 mmol), piperidine (0.0303 g, 0.356 mmol), and acetic acid (0.0214 g, 0.356 mmol) in EtOH (5 mL) was heated at 150° C. for 30 minutes in a microwave oven. The reaction was cooled to room temperature and the resulting precipitate was filtered and dried in a Buchner funnel to give the title compound (0.0594 g, 50%) as a tan solid. MS (ES)+ m/e 334 [M+H]+1H NMR (400 MHz, DMSO-d6) □ ppm 9.08 (d, J=4.42 Hz, 1H) 8.80-8.88 (m, 2H) 8.25 (d, J=8.72 Hz, 1H) 8.00-8.07 (m, 2H) 7.98 (s, 1H) 7.65-7.68 (m, 2H) 7.63 (d, J=4.42 Hz, 1H).
……………..
Schemes/Experimentals
Scheme I:
Figure imgf000040_0001
Conditions: a) Tributyl(vinyl)tin, Pd(PPh3)4, dioxane, reflux; b) OsO4, NaIO4, 2,6- lutidine, f-BuOH, dioxane, H2O, rt; c) heteroaryl (R) boronic acid, Pd(PPh3)4, 2 M K2CO3, DMF, 10O 0C; d) 2,4-thiazolidinedione, piperidine, AcOH, EtOH, μwave, 150 0C.
Examples:
Example 1 : (5Z)-5-ff4-(4-pyridinyl)-6-quinolinvnmethylidene}-1 ,3-thiazolidine-
2,4-dione
Figure imgf000041_0001
a) 4-chloro-6-ethenylquinoline
A mixture of 6-bromo-4-chloroquinoline (6.52 g, 26.88 mmol; see J. Med. Chem., 21_, 268 (1978) ), tributyl(vinyl)tin (8.95 g, 28.22 mmol), and tetrakistriphenylphosphine palladium (0) (0.62 g, 0.54 mmol) in 1 ,4-dioxane (150 ml.) was refluxed for 2.0 h, cooled to room temperature, and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (0-4% MeOH:CH2CI2) to give the title compound (5.1 g) as a pale yellow solid. MS(ES)+ m/e 190 [M+H]+. This material was used directly in the next step.
b) 4-chloro-6-quinolinecarbaldehyde
A mixture of 4-chloro-6-ethenylquinoline (5.1 g, 26.88 mmol), 2,6-lutidine (5.76 g, 53.75 mmol), sodium (meta) periodate (22.99 g, 107.51 mmol), and osmium tetroxide (5.48 g of a 2.5% solution in tert-butanol, 0.538 mmol) in 1 ,4- dioxane:H2O (350 ml. of 3:1 mixture) was stirred for 3.5 h at room temperature and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (CH2CI2) to give the title compound (4.26 g, 83% for 2 steps) as a pale yellow solid. MS(ES)+ m/e 192 [M+H]+.
c) 4-(4-pyridinyl)-6-quinolinecarbaldehyde
A mixture of 4-chloro-6-quinolinecarbaldehyde (3.24 g, 16.92 mmol), 4- pyridylboronic acid (3.12 g, 25.38 mmol), tetrakistriphenylphosphine palladium (0) (0.978 g, 0.846 mmol), and 2M aqueous K2CO3 (7.02 g, 50.76 mmol, 25.4 mis of 2M solution) in DMF (100 ml.) was heated at 1000C for 3.0 h and cooled to room temperature. The mixture was filtered through celite and the celite was washed with EtOAc. The filtrate was transferred to a separatory funnel , washed with water and saturated NaCI, dried (Na2SO4), filtered and concentrated in vacuo. The residue was purified by flash chromatography on silica gel (5% MeOHiCH2CI2) to give the title compound (2.03 g, 51%) as a tan solid. MS(ES)+ m/e 235 [M+H]+.
d) (5Z)-5-{[4-(4-pyridinyl)-6-quinolinyl]methylidene}-1 ,3-thiazolidine-2,4-dione
A mixture of 4-(4-pyridinyl)-6-quinolinecarbaldehyde (0.108 g, 0.463 mmol), 2,4-thiazolidinedione (0.0417 g, 0.356 mmol), piperidine (0.0303 g, 0.356 mmol), and acetic acid (0.0214 g, 0.356 mmol) in EtOH (5 ml.) was heated at 15O0C for 30 minutes in a microwave oven. The reaction was cooled to room temperature and the resulting precipitate was filtered and dried in a Buchner funnel to give the title compound (0.0594 g, 50%) as a tan solid. MS(ES)+ m/e 334 [M+H]+. 1 H NMR (400 MHz, DMSO-d6) D ppm 9.08 (d, J=4.42 Hz, 1 H) 8.80 – 8.88 (m, 2 H) 8.25 (d, J=8.72 Hz, 1 H) 8.00 – 8.07 (m, 2 H) 7.98 (s, 1 H) 7.65 – 7.68 (m, 2 H) 7.63 (d, J=4.42 Hz, 1 H).



PATENTSUBMITTEDGRANTED
THIAZOLIDINEDIONE DERIVATIVES AS PI3 KINASE INHIBITORS [US2008255115]2008-10-16
THIAZOLIDINEDIONE DERIVATIVES AS P13 KINASE INHIBITORS [US2009306074]2009-12-10
Role of PI3K p110 delta Signaling in Retroviral Infection and Replication [US2011135655]2011-06-09
PI3 KINASE INHIBITORS AND USES THEREOF [US2011230476]2011-09-22
Identification of druggable targets for radiation mitigation using a small interfering RNA screening assay.
Zellefrow CD,et al. Radiat Res. 2012 Sep;178(3);150-9. PMID: 22747550.
Saadia et al (2009) Phosphatidylinositol-3-kinase as a therapeutic target in melanoma. Clin.Cancer Res. 15 3029. PMID: 19383818.
Knight et al (2010) Discovery of GSK2126458, a highly potent inhibitor of PI3K and the mammalian target of rapamycin. ACS Med.Chem.Lett. 1 39.
////////GSK 1059615,  GSK 615

GSK1904529A, GSK 4529

GSK1904529A Structure
GSK1904529A, GSK 4529
GSK1904529A is a selective inhibitor of IGF1R with IC50 of 27 nM.
851.96
FormulaC44H47F2N9O5S
CAS Number1089283-49-7
N-(2,6-difluorophenyl)-5-[3-[2-[5-ethyl-2-methoxy-4-[4-(4-methylsulfonylpiperazin-1-yl)piperidin-1-yl]anilino]pyrimidin-4-yl]imidazo[1,2-a]pyridin-2-yl]-2-methoxybenzamide,
N-(2,6-Difluorophenyl)-5-[3-[2-[[5-ethyl-2-(methyloxy)-4-[4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl]phenyl]amino]-4-pyrimidinyl]imidazo[1,2-a]pyridin-2-yl]-2-(methyloxy)benzamide
Figure US20080300242A1-20081204-C00613
GSK1904529A, selectively inhibits IGF-IR and IR with IC50s of 27 and 25 nmol/L, respectively. It is a promising candidate for therapeutic use in solid and hematologic cancers. IC50s for GSK1904529A in tumor cell lines ranged from 35 nmol/L to >30 umol/L. The tumor histologic types showing the greatest sensitivity to this compound were Ewing’s sarcoma and multiple myeloma, where IC50s in three of five Ewing’s sarcoma cell lines were <100 nmol/L and IC50s in five of eight multiple myeloma cell lines were <200 nmol/L.
GSK1904529A is a small-molecule inhibitor of the insulin-like growth factor-I receptor (IGF-IR) with IC50 value of 27 nM 1.
GSK1904529A is a reversible and ATP-competitive inhibitor with Ki value of 1.6 nM. In NIH-3T3/LISN cells, GSK1904529A potently inhibited phosphorylation of IGF-IR with IC50 value of 22 nM. It also demonstrated to be a selective inhibitor since it showed poor inhibitory activity against 45 other serine/threonine and tyrosine kinases. When treated with whole-cell extracts, GSK1904529A significantly inhibited the ligand-induced phosphorylation of IGF-IR and decreased phosphorylation of downstream signaling including AKT, IRS-1 and ERK at concentrations > 0.01μM. GSK1904529A suppressed cell proliferation in a variety of tumor cells. The IC50 values for NCI-H929, TC-71, SK-N-MC, COLO 205, MCF7 and PREC are 81, 35, 43, 124, 137 and 68 nM, respectively. In COLO 205, MCF-7, and NCI-H929 cells, GSK1904529A treatment resulted in cell accumulation in G1 and decrease in S and G2-M phases. Moreover, in NIH-3T3/LISN xenograft model, once daily administration of GSK1904529A at 30 mg/kg inhibited 56% of tumor growt
NMR3

…………..
Intermediates
Figure US20080300242A1-20081204-C00061Figure US20080300242A1-20081204-C00062Figure US20080300242A1-20081204-C00063Figure US20080300242A1-20081204-C00064

Figure US20080300242A1-20081204-C00065,


Figure US20080300242A1-20081204-C00060
Figure US20080300242A1-20081204-C00535Figure US20080300242A1-20081204-C00536Figure US20080300242A1-20081204-C00537
Figure US20080300242A1-20081204-C00542Figure US20080300242A1-20081204-C00543
Figure US20080300242A1-20081204-C00613u can construct your synthesis


Intermediate Example 2 5-[3-(2-chloro-4-pyrimidinyl)imidazo[1,2-a]pyridin-2-yl]-N-(2,6-difluorophenyl)-2-(methyloxy)benzamide
Figure US20080300242A1-20081204-C00060
Step A: Methyl 3-formyl-4-hydroxybenzoate
Figure US20080300242A1-20081204-C00061
Methyl 4-hydroxybenzoate (3.00 g, 19.7 mmol) and magnesium chloride (2.81 g, 29.5 mmol) were stirred in 100 mL of acetonitrile. TEA (10.3 mL, 73.9 mmol) was added via syringe. Paraformaldehyde (12.0 g, 133 mmol) was added in a single portion and the reaction was heated to reflux. The reaction was stirred at reflux for 24 hours and cooled to rt. The reaction was quenched by the addition of approximately 100 mL of 1N HCl and poured into EtOAc. The layers were separated, and the organic layer was washed with brine. The combined aqueous layers were extracted with EtOAc. The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The crude product was purified by flash chromatography. The clean fractions (by TLC) were concentrated in vacuo to afford 2.06 g (58%) of the desired product. 1H NMR (400 MHz, DMSO-d6): δ 11.54 (s, 1H), 10.27 (s, 1H), 8.21 (d, J=2.4 Hz, 1H), 8.03 (dd, J=8.8, 2.4 Hz, 1H), 7.07 (d, J=8.8 Hz, 1H), 3.79 (s, 3H).
Step B: methyl 3-formyl-4-(methyloxy)benzoate
Figure US20080300242A1-20081204-C00062
Methyl 3-formyl-4-hydroxybenzoate (2.06 g, 11.4 mmol) and K2CO(2.36 g, 17.1 mmol) were stirred in 50 mL of DMF. Methyl iodide (1.42 mL, 22.8 mmol) was added via syringe, and the reaction was stirred for 6 hours at rt. The reaction was poured into H2O and diethyl ether, and the layers were separated. The organic layer was washed with brine, and the combined aqueous layers were extracted with diethyl ether. The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo to afford 2.24 g of crude desired product. 1H NMR (400 MHz, DMSO-d6): δ 10.33 (s, 1H), 8.23 (d, J=2.2 Hz, 1H), 8.20 (dd, J=8.8, 2.2 Hz, 1H), 7.36 (d, J=8.8 Hz, 1H), 3.99 (s, 3H), 3.83 (s, 3H).
Step C: 2-(methyloxy)-5-[(methyloxy)carbonyl]benzoic acid
Figure US20080300242A1-20081204-C00063
Crude methyl 3-formyl-4-(methyloxy)benzoate from the previous step was dissolved in 40 mL of dioxane with stirring. Sulfamic acid (5.87 g, 60.5 mmol) in 20 mL of H2O was added to the stirring solution. Sodium chlorite (1.68 g, 80% by weight, 18.6 mmol) in 20 mL of H2O was added dropwise via addition funnel. The reaction was stirred for 40 min and poured into EtOAc and H2O. The layers were separated, and the organic layer was washed with brine. The combined aqueous layers were extracted with EtOAc, and the combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The solid was transferred to an Erlenmeyer flask with the aid of 30-40 mL of DCM. Approximately 50 mL of hexanes was added. Air was blown over the solution to allow most of the DCM to evaporate. Diethyl ether was added (20-30 mL), and the suspension was filtered. The solid was washed with hexanes, collected, and dried to afford 1.96 g (82% over 2 steps) of the desired compound. 1H NMR (400 MHz, DMSO-d6): δ 12.92 (brs, 1H), 8.22 (d, J=2.2 Hz, 1H), 8.07 (dd, J=8.8, 2.2 Hz, 1H), 7.24 (d, J=8.8 Hz, 1H), 3.88 (s, 3H), 3.82 (s, 3H).
Step D: methyl 3-{[(2,6-difluorophenyl)amino]carbonyl}-4-(methyloxy)benzoate
Figure US20080300242A1-20081204-C00064
2-(Methyloxy)-5-[(methyloxy)carbonyl]benzoic acid (1.96 g, 9.33 mmol) was suspended in 60 mL of DCM with stirring. DMF (0.036 mL, 0.46 mmol) was added via syringe. Oxalyl chloride (7.0 mL, 2.0M in dichloromethane, 14 mmol) was added dropwise via addition funnel. The addition funnel was rinsed with 10 mL of DCM. The reaction was stirred for 2 hours and concentrated in vacuo. The resultant solid was further dried under high vacuum pressure. The solid was dissolved in 60 mL of DCM with stirring. Pyridine (3.8 mL, 47 mmol), (4-dimethylamino)pyridine (0.0570 g, 0.467 mmol), and 2,6-difluoroaniline (3.0 mL, 28 mmol) were added to the solution. The reaction was stirred for 18 hours and poured into 1N HCl. The layers were separated, and the aqueous layer was washed once with DCM and once with diethyl ether. The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The crude product was purified by flash chromatography. The clean fractions (by TLC) were concentrated in vacuo to afford 1.56 g (52%) of the desired product. 1H NMR (400 MHz, DMSO-d6): δ 9.81 (s, 1H), 8.31 (d, J=2.0 Hz, 1H), 8.10 (dd, J=8.8, 2.0 Hz, 1H), 7.38 (m, 1H), 7.31 (d, J=88 Hz, 1H), 7.22-7.13 (m, 2H), 3.97 (s, 3H), 3.82 (s, 3H).
Step E: 5-[(2-Chloro-4-pyrimidinyl)acetyl]-N-(2,6-difluorophenyl)-2-(methyloxy)benzamide and 5-[(E)-2-(2-chloro-4-pyrimidinyl)-1-hydroxyethenyl]-N-(2,6-difluorophenyl)-2-(methyloxy)benzamide
Figure US20080300242A1-20081204-C00065
Methyl 3-{[(2,6-difluorophenyl)amino]carbonyl}-4-(methyloxy)benzoate (1.56 g, 4.86 mmol) was dissolved in 50 mL of THF with stirring and cooled to 0° C. Lithium bis(trimethylsilyl)amide (14.6 mL, 1.0M in THF, 14.6 mmol) was added slowly via syringe. 2-Chloro-4-methylpyrimidine (0.750 g, 5.83 mmol) was dissolved in 10 mL of THF and added dropwise via addition funnel. The addition funnel was rinsed with 10 mL of THF. The reaction was stirred at 0° C. for 1 hour and quenched with saturated ammonium chloride solution. The mixture was poured into H2O and EtOAc, and the layers were separated. The organic layer was washed with brine, and the combined aqueous layers were extracted with EtOAc. The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The crude product was purified by flash chromatography. The clean fractions (by TLC) were concentrated in vacuo to afford 1.26 g (62%) of the desired product. The proton NMR is a mixture of the keto and enol tautomers (˜2:1). 1H NMR (400 MHz, DMSO-d6): δ 13.58 (s, 1H, enol), 9.83 (s, 1H, keto), 9.82 (s, 1H, enol), 8.72 (m, 1H, keto), 8.54 (m, 1H, enol), 8.34 (s, 1H, keto), 8.22 (m, 1H, both), 8.06 (m, 1H, enol), 7.56 (m, 1H, keto), 7.42-7.31 (m, 2H, both+1H, enol), 7.22-7.14 (m, 2H, both), 6.55 (s, 1H, enol), 4.66 (s, 2H, keto), 4.00 (s, 3H, keto), 3.97 (s, 3H, enol).
Step F: 5-[3-(2-chloro-4-pyrimidinyl)imidazo[1,2-a]pyridin-2-yl]-N-(2,6-difluorophenyl)-2-(methyloxy)benzamide
A tautomeric mixture of 5-[(2-Chloro-4-pyrimidinyl)acetyl]-N-(2,6-difluorophenyl)-2-(methyloxy)benzamide and 5-[(E)-2-(2-chloro-4-pyrimidinyl)-1-hydroxyethenyl]-N-(2,6-difluorophenyl)-2-(methyloxy)benzamide (1.26 g, 3.02 mmol) was dissolved in 60 mL of DCM with stirring. NBS (0.538 g, 3.02 mmol) was added in a single portion. The reaction was stirred for 20 minutes and concentrated in vacuo. The residue was dissolved in 60 mL of dioxane with stirring, and 2-aminopyridine (0.853 g, 9.06 mmol) was added in a single portion. The reaction was heated at 60° C. with an oil bath for 24 hours and cooled to rt. The reaction was stirred at rt for an additional 40 hours. The reaction was poured into half-saturated NaHCOsolution and EtOAc, and the layers were separated. The organic layer was washed with brine, and the combined aqueous layers were extracted twice with EtOAc. The combined organic layers were dried over MgSO4, filtered, and concentrated in vacuo. The crude product was purified by flash chromatography. Impure fractions were concentrated and further purified by flash chromatography. The combined clean fractions (by TLC) from both runs were combined and concentrated in vacuo to afford 1.07 g (72%) of the desired product. 1H NMR (400 MHz, DMSO-d6): δ 9.80 (s, 1H), 9.40 (d, J=7.0 Hz, 1H), 8.57 (d, J=5.1 Hz, 1H), 8.10 (d, J=1.5 Hz, 1H), 7.84-7.77 (m, 2H), 7.57 (m, 1H), 7.39 (m, 1H), 7.33-7.26 (m, 2H), 7.24-7.14 (m, 3H), 3.99 (s, 3H).
Step A: 1,1-dimethylethyl 4-(methylsulfonyl)-1-piperazinecarboxylate
Figure US20080300242A1-20081204-C00535
To 1,1-dimethylethyl 1-piperazinecarboxylate (568 g, 3.05 mol) in DCM (4 L) was added TEA (617 g, 6.10 mol). After stirring for 10 min at 0° C., methanesulfonyl chloride (384 g, 3.35 mol) was added via addition funnel. The mixture was stirred at rt overnight. The mixture was poured into H2O (1 L) and extracted with DCM (1 L). The organic layer was separated, washed with H2O (1 L), dried (Na2SO4), and rotovapped down to provide the title compound of step A (720 g, 2.72 mol, 90%) which was used without further purification. 1H NMR (400 MHz, CDCl3) δ 1.44 (s, 9H), 2.76 (s, 3H), 3.11-3.17 (m, 4H), 3.50-3.53 (m, 4H).
Step B: 1-(methylsulfonyl)piperazine hydrochloride
Figure US20080300242A1-20081204-C00536
To 1,1-dimethylethyl 4-(methylsulfonyl)-1-piperazinecarboxylate (360 g, 1.36 mol) in MeOH (1 L) was added HCl (6 M in MeOH, 2 L) dropwise. The mixture was stirred at rt for 1 h. About 1 L of MeOH was rotovapped off. The resultant precipitate was filtered, washed with MeOH, and dried on high vacuum to provide the title compound of Step B (A combination of 2 batches, 570 g) which was used without further purification. 1H NMR (400 MHz, D2O) δ 2.95 (s, 3H), 3.27-3.29 (m, 4H), 3.42-3.46 (m, 4H).
Step C: 1-(methylsulfonyl)-4-(4-piperidinyl)piperazine dihydrochloride
Figure US20080300242A1-20081204-C00537
To 1-(methylsulfonyl)piperazine hydrochloride (150 g, 632 mmol) in DCE (3.5 L) was added TEA (192 g, 1.90 mol). The mixture was stirred at rt for 1 h and then acetic acid (94.8 g, 1.58 mol) and 1,1-dimethylethyl 4-oxo-1-piperidinecarboxylate (251 g, 1.26 mol) was added. After stirring another h, the reaction was cooled with an ice water bath and NaBH(OAc)(294 g, 1.39 mol) was added in four portions. The mixture was stirred overnight at rt. The reaction mixture was neutralized with saturated Na2COto pH 8-9. The organic phase was washed with brine and H2O, dried (Na2SO4), and rotovapped down to provide the crude Boc-protected amine (A combination of 3 batches, 720 g). This amount was split into 2 batches and used without further purification. To 1,1-dimethylethyl 4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinecarboxylate (360 g, 1.04 mol) in MeOH (1 L) was added HCl (6 M in MeOH, 2 L). The mixture was stirred at rt for 30 min. About 1 L of MeOH was rotovapped off. The resultant precipitate was filtered, washed with MeOH, and dried on high vacuum to provide the title compound of Step C (A combination of 2 batches, 600 g, 1.87 mol, 89% over 2 steps). 1H NMR (400 MHz, D2O) δ 1.87-1.91 (m, 2H), 2.33-2.36 (m, 2H), 2.97 (s, 3H), 2.99-3.05 (m, 2H), 3.45-3.59 (m, 11H).
Step A: 1-{1-[2-ethyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4-(methylsulfonyl)piperazine
Figure US20080300242A1-20081204-C00542
A mixture of 1-ethyl-2-fluoro-4-(methyloxy)-5-nitrobenzene (Example 187, step C) (0.93 g, 4.67 mmol), 1-(methylsulfonyl)-4-(4-piperidinyl)piperazine (Example 204, step C) (1.16 g, 4.67 mmol) and K2CO(0.774 g, 5.60 mmol) in DMSO (20 mL) was heated at 90° C. for 48 h. The reaction had not progressed sufficiently so the reaction was then heated at 120° C. for an additional 4 h. The reaction was cooled to rt, poured into H2O and extracted with DCM. Some saturated brine solution was added and the resultant was exhaustively extracted with DCM. The combined organics were washed with H2O then dried over MgSO4. The resultant solution was concentrated onto silica and purified by flash chromatography to afford 1-{1-[2-ethyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4-(methylsulfonyl)piperazine (1.12 g, 56%). 1H NMR (400 MHz, DMSO-d6) δ ppm 7.73-7.80 (m, 1H), 6.75 (s, 1H), 3.91 (s, 3H), 3.23-3.30 (m, 1H), 3.05-3.19 (m, 3H), 2.87 (s, 2H), 2.70-2.84 (m, 2H), 2.53-2.67 (m, 5H), 1.77-1.94 (m, 2H), 1.48-1.67 (m, 2H), 1.19 (t, J=7.42 Hz, 3H).
Step B: 5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}aniline
Figure US20080300242A1-20081204-C00543
A mixture of 1-{1-[2-ethyl-5-(methyloxy)-4-nitrophenyl]-4-piperidinyl}-4-(methylsulfonyl)piperazine (1.12 g, 2.63 mmol) and sulfided platinum on carbon (0.410 g, 0.105 mmol) in EtOAc (40 mL) was sealed in a round bottom flask with a rubber septum. The reaction mixture was purged with Ngas and then a balloon of Hgas was connected and the vessel was flushed with the H2gas. The reaction was stirred at rt for 2 d. TLC analysis showed the complete consumption of the starting nitro compound so the reaction mixture was filtered through celite to remove the catalyst. The filtrate was concentrated onto silica gel and purified by flash chromatography to afford 5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}aniline (0.479 g, 46%).
1H NMR (400 MHz, DMSO-d6) δ ppm 6.60 (s, 1H), 6.46 (s, 1H), 4.35 (br. s., 2H), 3.71 (s, 3H), 3.03-3.16 (m, 4H), 2.81-2.93 (m, 5H), 2.56-2.68 (m, 6H), 2.29-2.42 (m, 1H), 1.72-1.89 (m, 2H), 1.44-1.62 (m, 2H), 1.09 (t, J=7.51 Hz, 3H).
Example 237 N-(2,6-difluorophenyl)-5-(3-{2-[(5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}phenyl)amino]-4-pyrimidinyl}imidazo[1,2-a]pyridin-2-yl)-2-(methyloxy)benzamide
Figure US20080300242A1-20081204-C00613
A mixture of 5-[3-(2-chloro-4-pyrimidinyl)imidazo[1,2-a]pyridin-2-yl]-N-(2,6-difluorophenyl)-2-(methyloxy)benzamide (Intermediate Example 2) (0.60 g, 1.22 mmol), 5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}aniline (Example 206, Step B) (0.48 g, 1.22 mmol) and HCl (4N,1,4-Dioxane, 0.61 mL, 2.44 mmol) in trifluoroethanol (15 mL) was heated at 170° C. for 40 min in the microwave. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography. Recrystallization from DCM and EtOH afforded the title compound N-(2,6-difluorophenyl)-5-(3-{2-[(5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}phenyl)amino]-4-pyrimidinyl}imidazo[1,2-a]pyridin-2-yl)-2-(methyloxy)benzamide (0.61 g, 56%).

1H NMR (400 MHz, DMSO-d6)

δ ppm 9.80 (s, 1H), 9.36 (br. s., 1H), 8.50 (s, 1H), 8.26 (d, J=5.22 Hz, 1H), 8.12 (d, J=2.11 Hz, 1H), 7.80 (dd, J=8.80, 2.02 Hz, 1H), 7.71 (d, J=9.07 Hz, 1H), 7.53 (s, 1H), 7.36-7.50 (m, 2H), 7.30 (d, J=8.80 Hz, 1H), 7.14-7.25 (m, 2H), 6.91-7.00 (m, 1H), 6.83 (s, 1H), 6.58 (d, J=5.22 Hz, 1H), 4.00 (s, 3H), 3.80 (s, 3H), 3.08-3.15 (m, 4H), 3.00-3.07 (m, 2H), 2.88 (s, 3H), 2.67-2.76 (m, 2H), 2.61-2.66 (m, 4H), 2.56 (q, J=7.51 Hz, 2H), 2.38-2.46 (m, 1H), 1.80-1.91 (m, 2H), 1.50-1.68 (m, 2H), 1.11 (t, J=7.51 Hz, 3H).

MS (M+H, ES+) 852.

Separately, the Title Compound was Prepared in the Following Manner:
A mixture of 5-[3-(2-chloro-4-pyrimidinyl)imidazo[1,2-a]pyridin-2-yl]-N-(2,6-difluorophenyl)-2-(methyloxy)benzamide (Intermediate Example 2) (23.0 g, 46.8 mmol), 5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}aniline (Example 206, Step B) (18.6 g, 46.8 mmol) and HCl (4N,1,4-Dioxane, 23.4 mL, 93.6 mmol) in trifluoroethanol (200 mL) was heated in a sealed vessel at 85° C. for 48 h. After cooling to rt, the reaction mixture was treated with an excess of 7N NHin MeOH and then subjected to filtration. The filtrate was concentrated onto silica gel and purified by flash chromatography. The chromatographed product was dissolved in DCM and treated with an excess of diethyl ether. The resultant bright yellow precipitate was collected by filtration and then recrystallized from DCM and EtOH to afford the title compound N-(2,6-difluorophenyl)-5-(3-{2-[(5-ethyl-2-(methyloxy)-4-{4-[4-(methylsulfonyl)-1-piperazinyl]-1-piperidinyl}phenyl)amino]-4-pyrimidinyl}imidazo[1,2-a]pyridin-2-yl)-2-(methyloxy)benzamide (28.2 g, 67%).

……………..
Discovery and optimization of imidazo[1,2-a]pyridine inhibitors of insulin-like growth factor-1 receptor (IGF-1R)
Bioorg Med Chem Lett 2009, 19(3): 1004……http://www.sciencedirect.com/science/article/pii/S0960894X08014376
Image for unlabelled figure
Reagents and conditions: (a) (ClCO)2, DMF, CH2Cl2; (b) 2,6-difluoroaniline, ...
Scheme 1.
Reagents and conditions: (a) (ClCO)2, DMF, CH2Cl2; (b) 2,6-difluoroaniline, pyridine, CH2Cl2 (84%, 2 steps); (c) LiN(SiMe3)2, THF (83%); (d) NBS, CH2Cl2, then 2-aminopyridine, dioxane, 60 °C (77%); (e) HCl or p-TSA·H2O, trifluoroethanol or isopropanol, 80–100 °C or 140–180 °C (μw) (50–90%).

References

Antitumor activity of GSK1904529A, a small-molecule inhibitor of the insulin-like growth factor-I receptor tyrosine kinase.
Sabbatini et al. Clin Cancer Res. 2009 May 1;15(9):3058-67. PMID: 19383820.


/////////////GSK1904529A, IGF1R, GSK 4529, preclinical