Tuesday 17 March 2015

KHK 7580 structure cracked

WP_000286


KHK 7580 .....example no 3.001  R1-X- group\
  Figure imgb0352 Figure imgb0353 Figure imgb0354
HCl salt
MS · APCI: 361 [M+H] +
Figure imgb0350
in EP1757582
4-(3S-(1R-(1-naphthyl)ethylamino)pyrrolidin-1- yl)phenylacetic acid
4-​[(3S)​-​3-​[[(1R)​-​1-​(1-​naphthalenyl)​ethyl]​amino]​-​1-​pyrrolidinyl]​-Benzeneacetic acid,
cas will be updated
BASE ....870964-67-3
DI HCL SALT .......870856-31-8
MF C24 H26 N2 O2 BASE
MW 374.48 BASE
KHK-7580
KHK-7580; MT-4580
Mitsubishi Tanabe Pharma Corp... innovator
Kyowa Hakko Kirin Co Ltd.. licencee
4-(3S-(1R-(1-naphthyl)ethylamino)pyrrolidin-1-yl)phenylacetic acid,
useful as calcium-sensitive receptor (CaSR) agonists for treating hyperparathyroidism.  a CaSR agonist, being developed by Kyowa Hakko Kirin, under license from Mitsubishi Tanabe, for treating secondary hyperparathyroidism (phase 2 clinical, as of March 2015).
WILL BE UPDATED
WO2005115975,/EP1757582
http://www.google.co.in/patents/EP1757582A1?cl=en
Example no 3.001  R1-X- group Figure imgb0352 Figure imgb0353 Figure imgb0354
HCl salt
MS · APCI: 361 [M+H] +
Figure imgb0350



WO 2015034031A1
http://worldwide.espacenet.com/publicationDetails/biblio?DB=worldwide.espacenet.com&II=0&ND=3&adjacent=true&locale=en_EP&FT=D&date=20150312&CC=WO&NR=2015034031A1&KC=A1
Mitsubishi Tanabe Pharma Corporation
The present invention provides a novel crystal form of an arylalkylamine
compound. Specifically, a novel crystal form of
4-(3S-(1R-(1-naphthyl)ethylamino)pyrrolidin-1- yl)phenylacetic acid has
excellent stability, and is therefore useful as an active ingredient for
a medicine. The present invention also provides an industrially
advantageous method for producing an arylalkylamine compound.
WP_000287
WO 2015034031A1
http://worldwide.espacenet.com/publicationDetails/biblio?DB=worldwide.espacenet.com&II=0&ND=3&adjacent=true&locale=en_EP&FT=D&date=20150312&CC=WO&NR=2015034031A1&KC=A1
Mitsubishi Tanabe Pharma Corporation
The present invention provides a novel crystal form of an arylalkylamine compound. Specifically, a novel crystal form of 4-(3S-(1R-(1-naphthyl)ethylamino)pyrrolidin-1- yl)phenylacetic acid has excellent stability, and is therefore useful as an active ingredient for a medicine. The present invention also provides an industrially advantageous method for producing an arylalkylamine compound.

see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html
see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html
see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html
see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html
see all at   http://drugpatentsint.blogspot.in/2015/03/wo-2015034031.html
do not miss out on above click


 http://www.kyowa-kirin.com/research_and_development/pipeline/
KHK7580 -Secondary Hyperparathyroidism
JP
CompanyMitsubishi Tanabe Pharma Corp.
DescriptionCalcium receptor agonist
Molecular Target
Mechanism of ActionCalcium-sensing receptor (CaSR) agonist
Therapeutic ModalitySmall molecule
Latest Stage of DevelopmentPhase II
Standard IndicationThyroid disease
Indication DetailsTreat hyperparathyroidism in patients receiving hemodialysis; Treat secondary hyperparathyroidism (SHPT)
Regulatory Designation
Partner
August 29, 2014

Kyowa Hakko Kirin Announces Commencement of Phase 2b Clinical Study of KHK7580 in Patients with Secondary Hyperparathyroidism in Japan

Tokyo, Japan, August 29, 2014 --- Kyowa Hakko Kirin Co., Ltd. (Tokyo: 4151, President and CEO: Nobuo Hanai, "Kyowa Hakko Kirin") today announced the initiation of a phase 2b clinical study evaluating KHK7580 for secondary hyperparathyroidism patients receiving hemodialysis in Japan.
This randomized, placebo-controlled, double-blind, parallel-group, multi-center study is designed to evaluate efficacy and safety in cohorts comprising KHK7580, its placebo and cinacalcet and initial dose of KHK7580 for secondary hyperparathyroidism patients receiving hemodialysis.
KHK7580 is a small molecular compound produced by Mitsubishi Tanabe Pharma Corporation (President & Representative Director, CEO: Masayuki Mitsuka, "Mitsubishi Tanabe Pharma"). Kyowa Hakko Kirin signed a license agreement of KHK7580 with Mitsubishi Tanabe Pharma for the rights to cooperative research, develop, market and manufacture the product in Japan and some part of Asia on March 2008.
The Kyowa Hakko Kirin Group is contributing to the health and prosperity of the world's people by pursuing advances in life sciences and technology and creating new value.
Outline of this study
ClinicalTrials.gov IdentifierNew window opensNCT02216656
Target PopulationSecondary hyperparathyroidism patients receiving hemodialysis
Trial DesignRandomized, placebo-controlled, double-blind (included open arm of cinacalcet), parallel-group, multi-center study
Administration GroupKHK7580, Placebo, cinacalcet
Target Number of Subjects150
Primary ObjectiveEfficacy
Trial LocationJapan
Trial DurationJul. 2014 to Jun. 2015

Contact:

Kyowa Hakko Kirin
Media Contact:
+81-3-3282-1903
or
Investors:
+81-3-3282-0009




Update on march 2016

New comment waiting approval on New Drug Approvals

M.F. Balandrin commented on KHK 7580 structure cracked
KHK 7580 .....example 3.008 2HCl MS · APCI: 375[M + H]+ in …
The calcimimetic agent, KHK-7580, currently entering Phase III clinical trials, has now been given the INN (WHO) generic name, evocalcet. Its chemical structure has also now been published and it is, in fact, correct as proposed by Dr. Crasto (Well Done!!):
http://www.drugspider.com/drug/evocalcet
https://tripod.nih.gov/ginas/app/substance/f580b9fd
http://www.medkoo.com/products/6729
(Etymologically, in classical Latin, "evolutio" refers to "the unrolling of a scroll" and "evocare" refers to a "call out"...).

http://www.medkoo.com/products/6729
img
Name: Evocalcet
CAS#: 870964-67-3
Chemical Formula: C24H26N2O2
Exact Mass: 374.19943
Evocalcet is a calcium-sensing receptor agonist. The calcium-sensing receptor (CaSR) is a Class C G-protein coupled receptor which senses extracellular levels of calcium ion. The calcium-sensing receptor controls calcium homeostasis by regulating the release of parathyroid hormone (PTH). CaSR is expressed in all of the organs of the digestive system. CaSR plays a key role in gastrointestinal physiological function and in the occurrence of digestive disease. High dietary Ca2+ may stimulate CaSR activation and could both inhibit tumor development and increase the chemotherapeutic sensitivity of cancer cells in colon cancer tissues. (Last update: 12/15/2015).
Synonym: MT-4580; MT 4580; MT4580; KHK-7580; KHK7580; KHK 7580; Evocalcet
IUPAC/Chemical Name: 2-(4-((S)-3-(((R)-1-(naphthalen-1-yl)ethyl)amino)pyrrolidin-1-yl)phenyl)acetic acid

2
https://tripod.nih.gov/ginas/app/substance/f580b9fd
Structure of EVOCALCET
http://www.drugspider.com/drug/evocalcet
INN name
Evocalcet
Lab Code(s)
MT-4580
KHK-7580
Chemical name
{4-[(3S)-3-{[(1R)-1-(Naphthalen-1-yl)ethyl]amino}pyrrolidin-1-yl]phenyl}acetic acid
Chemical structure
Molecular formula
C24H26N2O2
SMILES
O=C(O)CC1=CC=C(N2C[C@@H](N[C@@H](C3=C4C=CC=CC4=CC=C3)C)CC2)C=C1
CAS registry number
870964-67-3
Orphan Drug Status
No
On Fast track
No
New Molecular Entity
Yes
Originator
Developer(s)
Class
Mechanism of action
WHO ATC code(s)
EPhMRA code(s)
Clinical trial(s)
ConditionsInterventionsPhasesRecruitmentSponsor/Collaborators
Secondary HyperparathyroidismDrug: KHK7580Phase 3RecruitingKyowa Hakko Kirin Company, Limited
Secondary HyperparathyroidismDrug: KHK7580Phase 3RecruitingKyowa Hakko Kirin Company, Limited
Secondary HyperparathyroidismDrug: KHK7580|Drug: KRN1493Phase 2|Phase 3RecruitingKyowa Hakko Kirin Company, Limited
Secondary HyperparathyroidismDrug: Placebo|Drug: KHK7580 low dose|Drug: KHK7580 middle dose|Drug: KHK7580 high dose|Drug: KRN1493Phase 2CompletedKyowa Hakko Kirin Company, Limited
HyperparathyroidismDrug: KHK7580Phase 1|Phase 2CompletedKyowa Hakko Kirin Company, Limited
Secondary HyperparathyroidismDrug: KHK7580Phase 1CompletedKyowa Hakko Kirin Company, Limited
Updated on
11 Oct 2015
///////////////
SMILES Code: O=C(O)CC1=CC=C(N2C[C@@H](N[C@@H](C3=C4C=CC=CC4=CC=C3)C)CC2)C=C1

GSK 2269557 In Phase 1....Asthma , COPD, is it COMPD A OR B?


COMPD A  

COMPD B
Compd A OR B IS GSK 2269557
Phosphatidylinositol 3-Kinase (PI3K)
PHASE 1....asthma & COPD
ASHTHMA COPD
DATA FOR COMPD A
6-​(1H-​indol-​4-​yl)​-​4-​[5-​[[4-​(1-​methylethyl)​-​1-​piperazinyl]​methyl]​-​2-​oxazolyl]​-1H-​Indazole, 6-(1 H-lndol-4-yl)-4-(5-{[4-(1-methylethyl)-1-piperazinyl]methyl}-1,3-oxazol-2-yl)-1 H- indazole
CAS 1254036-77-5 hcl salt
base 1254036-71-9, 440.54, C26 H28 N6 O
Formula C26H28N6O.HCl
EMAIL ME amcrasto@gmail.com

 
DATA FOR COMPD B
Methanesulfonamide, N-​[5-​[4-​[5-​[[(2R,​6S)​-​2,​6-​dimethyl-​4-​morpholinyl]​methyl]​-​2-​oxazolyl]​-​1H-​indazol-​6-​yl]​-​2-​methoxy-​3-​pyridinyl]​-​,
N-[5-[4-(5-{[(2R,6S)-2,6-dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1H-indazol-6-yl]-2-(methyloxy)-3-pyridinyl]methanesulfonamide
1254036-66-2 CAS
C24 H28 N6 O5 S, 512.58
Compound B may be prepared according to known procedures, such as those disclosed in international patent application PCT/EP2010/055666 (publication number WO02010/125082)
EMAIL ME amcrasto@gmail.com

Phosphoinositide 3ΌΗ kinases (hereinafter PI3Ks) are a family of signal transducer enzymes which are involved in various cellular functions including cell growth, proliferation and differentiation. A wide variety of retroviruses and DNA-based viruses activate the PI3K pathway as a way of preventing host cell death during viral infection and ultimately exploiting the host cell synthesis machinery for its replication (Virology 344(1) p. 131-8 (2006) by Vogt et al.; and Nat. Rev. Microbiol. 6(4) p. 265-75 (2008) by Buchkovich et al). It has therefore been postulated that PI3K inhibitors may have potential therapeutic benefit in the treatment of viral infections such as influenza virus infection, in addition to the more established treatment of cancer and inflammatory diseases. The Influenza NS1 protein activates Class la PI3Ks by binding to their regulatory subunit p85beta but not to other Class la regulatory subunits such as p85alpha. The recent crystal structure of the NS1-p85beta complex (Hale et al. Proc. Natl. Acad. Sci. U S A. 107(5) p.1954-1959 (2010)) is also suggestive of an interaction with the p110 kinase subunit providing a mechanism for catalytic activation of the kinase domain.

This observation provides a rationale for isoform specificity not only with the p85 regulatory subunit but also potentially with the p110 catalytic subunit too. The function of PI3K during influenza virus infection has also been investigated by, for example, Ehrhardt et al. (Cell. Microbiol. 8(8) p. 1336-1348 (2006)), and the role of PI3K5 signalling in morbidity and lung pathology induced by influenza virus infection has been reported in WO 2010/083163. There remains a need to provide compounds which are inhibitors of the activity or function of PI3K5 which may be useful in the treatment or prevention of influenza virus infection.   GSK 2269557 is an inhaled phosphatidylinositol 3-kinase delta (PI3Kdelta) inhibitor in early clinical trials at GlaxoSmithKline for the treatment of patients with asthma and also for the treatment of chronic obstructive pulmonary disease (COPD) in patients who smoke cigarettes.
  • 18 Nov 2014GlaxoSmithKline plans a phase II trial in Chronic obstructive pulmonary disease in Belgium, Denmark, the Netherlands and Russia (NCT02294734)
  • 01 Jun 2014Phase-II clinical trials in Chronic obstructive pulmonary disease in Germany (Inhalation)
  • 01 May 2014GlaxoSmithKline plans a phase II trial for Chronic obstructive pulmonary disease in Germany (NCT02130635)
Study ID Status Title Patient Level Data
115117 Completed A single-centre. double-blind, placebo controlled three part study to evaluate the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of single and repeat doses of nebulised GSK2269557 in healthy male subjects
115119 Active not recruiting A Double-Blind, Placebo Controlled, Randomised, Parallel Group Study to Evaluate the Safety, Tolerability and Pharmacokinetics of Multiple Doses of GSK2269557 Administered as a Dry Powder to COPD Patients
116617 Completed A Single-Centre, Double-Blind, Placebo Controlled Two Part Study to Evaluate the Safety, Tolerability and Pharmacokinetics of Single and Repeat Doses of GSK2269557 as a Dry Powder in Healthy Subjects who Smoke Cigarettes
116678 Not yet recruiting A Randomised, Double-blind (Sponsor Unblinded), Placebo-controlled, Parallel-group, Multicentre Study to Evaluate the Efficacy and Safety of GSK2269557 Administered in Addition to Standard of Care in Adult Subjects Diagnosed With an Acute Exacerbation of Chronic Obstructive Pulmonary Disease
 


 EMAIL ME amcrasto@gmail.com

CLICK ON IMAGES TO VIEW SIMILAR ROUTES FOR COMPD A AND B EO2 EO1A   CLICK ON IMAGE TO VIEW

EO1B ...............................................................................    


COMPD A

WO 2012032065 http://www.google.com/patents/WO2012032065A1?cl=en

Example 68 6-(1 H-lndol-4-yl)-4-(5-{[4-(1-methylethyl)-1-piperazinyl]methyl}-1,3-oxazol-2-yl)-1 H- indazole
Method A 6-Chloro-4-(5-{[4-(1-methylethyl)-1-piperazinyl]methyl}-1 ,3-oxazol-2-yl)-1-(phenylsulfonyl)- 1/-/-indazole (97 mg, 0.194 mmol), 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 H- indole (61.3 mg, 0.252 mmol, available from Frontier Scientific Europe), chloro[2'- (dimethylamino)-2-biphenylyl]palladium-(1 ,4S)-bicyclo[2.2.1]hept-2-yl[(1 S,4 )- bicyclo[2.2.1]hept-2-yl]phosphane (10.87 mg, 0.019 mmol) and potassium phosphate tribasic (124 mg, 0.582 mmol) were dissolved in 1 ,4-dioxane (1 ml) and water (0.1 ml) and heated in a Biotage Initiator microwave at 100°C for 30 min. Additional 4-(4,4,5,5- tetramethyl-1 ,3,2-dioxabotolan-2-yl)-1 H-indole (61.3 mg, 0.252 mmol) and chloro[2'- (dimethylamino)-2-biphenylyl]palladium-(1 ,4S)-bicyclo[2.2.1]hept-2-yl[(1 S,4 )- bicyclo[2.2.1]hept-2-yl]phosphane (5 mg) were added and the reaction heated at 1 10°C for 30 min, then 140°C for 30 min. The solvent was removed in vacuo and the residue purified by silica gel chromatography, eluting with 0-25% methanol in dichloromethane. The appropriate fractions were combined and concentrated to give a brown solid which was dissolved in MeOH:DMSO (1 ml, 1 : 1 , v/v) and purified by MDAP (method H). The appropriate fractions were concentrated in vacuo to give the title compound as a white solid (30 mg). LCMS (Method A): Rt 0.57 mins, MH+ 441. Method B 6-Chloro-4-(5-{[4-(1-methylethyl)-1-piperazinyl]methyl}-1 ,3-oxazol-2-yl)-1-(phenylsulfonyl)- 1 H-indazole (75.17 g, 150 mmol), 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 H- indole (73.1 g, 301 mmol), sodium bicarbonate (37.9 g, 451 mmol), and chloro[2'- (dimethylamino)-2-biphenylyl]palladium-(1 ,4S)-bicyclo[2.2.1]hept-2-yl[(1 S,4 )- bicyclo[2.2.1]hept-2-yl]phosphane (8.43 g, 15.03 mmol) were suspended in nitrogen purged 1 ,4-dioxane (1200 ml_) and water (300 ml_). The reaction vessel was placed under alternating vacuum and nitrogen five times with overhead stirring, then finally placed under a nitrogen atmosphere and heated to 120°C for 2.5 h. The reaction mixture was cooled to 45°C and then treated with 2M aqueous sodium hydroxide (376 ml_, 752 mmol). After stirring at 45°C overnight (~ 13h), the mixture was cooled to RT and DCM (600 ml) and water (400 ml) were added. The layers were separated and the aqueous re-extracted with DCM: 1 ,4-dioxane (1 : 1). Brine was added and the mixture filtered through Celite, washing with DCM: 1 ,4-dioxane (1 : 1). The layers were separated and 2M HCI (1000 ml) added to the organic. The mixture was again filtered through Celite washing with 500 ml 2M HCI keeping the washings separate. The filtrate layers were then separated and the organic layer was washed with the acid washings from the Celite. Layers were separated and the acidic aqueous combined. This was then back-washed with 2x500 ml of DCM; each wash requiring a Celite filtration. The acidic aqueous was then given a final filtration through Celite washing the Celite pad with 150 ml of 2M HCI. The acidic aqueous was transfered to a beaker (5000 ml) and with vigorous stirring 2M NaOH was added to basify the mixture to pH 10-11. The mixture was then extracted using 1 ,4-dioxane: DCM (1 : 1) (5 x 500 ml). The combined organics were washed with brine, dried over magnesium sulphate, filtered and evaporated to yield a brown foam that was dried in vacuo at 50°C overnight. This material was split into three batches and each was purified by reverse phase column chromatography (3x 1.9 kg C18 column), loading in DMF/TFA (1 : 1 , 30 ml) then eluting with 3-40% MeCN in Water + 0.25% TFA (Note: Columns 2 & 3 used a different gradient starting with 10% MeCN). Appropriate fractions were combined, the acetotnitrile removed in vacuo and the acidic aqueous basified to pH10 by addition of saturated aqueous sodium carbonate solution to the stirred solution. The resultant solid was collected by filtration, washed with water then dried in vacuo at 65°C overnight to give the title compound (28.82 g) as a pale brown foam. LCMS (Method A): Rt 0.68 mins, MH+ 441. 1 H NMR (400MHz ,DMSO-d6) d = 13.41 (br. s., 1 H), 11.35 (br. s., 1 H), 8.59 (br. s., 1 H), 8.07 (d, J = 1.5 Hz, 1 H), 7.90 (br. s., 1 H), 7.51 - 7.44 (m, 2 H), 7.32 (s, 1 H), 7.27 - 7.21 (m, 2 H), 6.61 - 6.58 (m, 1 H), 3.73 (br. s., 2 H), 2.64 - 2.36 (m, 9 H), 0.97 - 0.90 (m, 6 H) Method C Potassium hydroxide (145.6 g) was added to a suspension of 6-(1 H-indol-4-yl)-4-(5-{[4-(1- methylethyl)-1-piperazinyl]methyl}-1 ,3-oxazol-2-yl)-1-(phenylsulfonyl)-1 H-indazole (300.7 g) and cetyltrimethylammonium bromide (9.3 g) in tetrahydrofuran (6.0 L) and water (30 ml) stirring under nitrogen at ambient temperature. The mixture was heated at reflux for 17 hours and was then cooled to 20-25°C. Ethyl acetate (3.0 L) and water (3.0 L) were added, stirred for 10 minutes and then separated. The organic layer was extracted with hydrochloric acid (1 M, 1 x 3.0 L, 2 x 1.5L) and the acidic extracts combined and basified to ~pH 8 by the addition of saturated sodium carbonate solution (2.1 L). After ageing for 30 minutes the resultant suspension was filtered, washed with water (300 ml) and the solid dried under vacuum at 65°C to give the title compound as a pale yellow solid (127.9 g). LCMS (Method B): Rt 2.44 min, MH+ 441. ....................................................................................

WO 2010125082 http://www.google.co.in/patents/WO2010125082A1?cl=en

 Example 6 6-(1 H-lndol-4-yl)-4-(5-{[4-(1 -methylethyl)-1 -piperazinyl]methyl}-1 ,3-oxazol-2-yl)-1 H- indazole
Method A 6-Chloro-4-(5-{[4-(1-methylethyl)-1-piperazinyl]methyl}-1 ,3-oxazol-2-yl)-1-(phenylsulfonyl)- 1H-indazole (97 mg, 0.194 mmol), 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 H- indole (61.3 mg, 0.252 mmol, available from Frontier Scientific Europe), chloro[2'- (dimethylamino)-2-biphenylyl]palladium-(1 R,4S)-bicyclo[2.2.1]hept-2-yl[(1 S,4R)- bicyclo[2.2.1]hept-2-yl]phosphane (10.87 mg, 0.019 mmol) and potassium phosphate tribasic (124 mg, 0.582 mmol) were dissolved in 1 ,4-dioxane (1 ml) and water (0.1 ml) and heated in a Biotage Initiator microwave at 1000C for 30 min. Additional 4-(4, 4,5,5- tetramethyl-1 ,3,2-dioxabotolan-2-yl)-1 H-indole (61.3 mg, 0.252 mmol) and chloro[2'- (dimethylamino)-2-biphenylyl]palladium-(1 R,4S)-bicyclo[2.2.1]hept-2-yl[(1 S,4R)- bicyclo[2.2.1]hept-2-yl]phosphane (5 mg) were added and the reaction heated at 1 1O0C for 30 min, then 14O0C for 30 min. The solvent was removed in vacuo and the residue purified by silica gel chromatography, eluting with 0-25% methanol in dichloromethane. The appropriate fractions were combined and concentrated to give a brown solid which was dissolved in MeOH:DMSO (1 ml, 1 :1 , v/v) and purified by MDAP (method A). The appropriate fractions were concentrated in vacuo to give the title compound as a white solid (30 mg). LCMS (Method A): Rt 0.57 mins, MH+ 441. Method B 6-Chloro-4-(5-{[4-(1-methylethyl)-1-piperazinyl]methyl}-1 ,3-oxazol-2-yl)-1-(phenylsulfonyl)- 1 H-indazole (75.17 g, 150 mmol), 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 H- indole (73.1 g, 301 mmol), sodium bicarbonate (37.9 g, 451 mmol), and chloro[2'- (dimethylamino)-2-biphenylyl]palladium-(1 R,4S)-bicyclo[2.2.1]hept-2-yl[(1 S,4R)- bicyclo[2.2.1]hept-2-yl]phosphane (8.43 g, 15.03 mmol) were suspended in nitrogen purged 1 ,4-dioxane (1200 ml.) and water (300 ml_). The reaction vessel was placed under alternating vacuum and nitrogen five times with overhead stirring, then finally placed under a nitrogen atmosphere and heated to 1200C for 2.5 h. The reaction mixture was cooled to 45°C and then treated with 2M aqueous sodium hydroxide (376 ml_, 752 mmol). After stirring at 450C overnight (~ 13h), the mixture was cooled to RT and DCM (600 ml) and water (400 ml) were added. The layers were separated and the aqueous re-extracted with DCM: 1 ,4-dioxane (1 :1 ). Brine was added and the mixture filtered through Celite, washing with DCM: 1 ,4-dioxane (1 :1 ). The layers were separated and 2M HCI (1000 ml) added to the organic. The mixture was again filtered through Celite washing with 500 ml 2M HCI keeping the washings separate. The filtrate layers were then separated and the organic layer was washed with the acid washings from the Celite. Layers were separated and the acidic aqueous combined. This was then back-washed with 2x500 ml of DCM; each wash requiring a Celite filtration. The acidic aqueous was then given a final filtration through Celite washing the Celite pad with 150 ml of 2M HCI. The acidic aqueous was transfered to a beaker (5000 ml) and with vigorous stirring 2M NaOH was added to basify the mixture to pH 10-11. The mixture was then extracted using 1 ,4-dioxane:DCM (1 :1 ) (5 x 500 ml). The combined organics were washed with brine, dried over magnesium sulphate, filtered and evaporated to yield a brown foam that was dried in vacuo at 500C overnight. This material was split into three batches and each was purified by reverse phase column chromatography (3x 1.9 kg C18 column), loading in DMF/TFA (1 :1 , 30 ml) then eluting with 3-40% MeCN in Water + 0.25% TFA (Note: Columns 2 & 3 used a different gradient starting with 10% MeCN). Appropriate fractions were combined, the acetotnitrile removed in vacuo and the acidic aqueous basified to pH10 by addition of saturated aqueous sodium carbonate solution to the stirred solution. The resultant solid was collected by filtration, washed with water then dried in vacuo at 65°C overnight to give the title compound (28.82 g) as a pale brown foam. LCMS (Method A): Rt 0.68 mins, MH+ 441. 1H NMR (400MHz ,DMSOd6) d = 13.41 (br. s., 1 H), 11.35 (br. s., 1 H), 8.59 (br. s., 1 H), 8.07 (d, J = 1.5 Hz, 1 H), 7.90 (br. s., 1 H), 7.51 - 7.44 (m, 2 H), 7.32 (s, 1 H), 7.27 - 7.21 (m, 2 H), 6.61 - 6.58 (m, 1 H), 3.73 (br. s., 2 H), 2.64 - 2.36 (m, 9 H), 0.97 - 0.90 (m, 6 H) EMAIL ME amcrasto@gmail.com    

COMPD B WO2010125082 http://www.google.co.in/patents/WO2010125082A1?cl=en

Example 1 Λ/-[5-[4-(5-{[(2/?,6S)-2,6-Dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1H-indazol- 6-yl]-2-(methyloxy)-3-pyridinyl]methanesulfonamide
Method A To a solution of 6-chloro-4-(5-{[(2R,6S)-2,6-dimethyl-4-morpholinyl]methyl}-1 ,3-oxazol-2- yl)-1-(phenylsulfonyl)-1 H-indazole (0.20 g, 0.411 mmol) and N-[2-(methoxy)-5-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3-pyridyl]methanesulfonamide (0.175 g, 0.534 mmol) in 1 ,4-dioxane (2 ml) was added chloro[2'-(dimethylamino)-2-biphenylyl]palladium- 1 (1 /?,4S)-bicyclo[2.2.1]hept-2-yl[(1 S,4/?)-bicyclo[2.2.1]hept-2-yl]phosphane (11.5 mg, 0.021 mmol), potassium phosphate tribasic (0.262 g, 1.23 mmol) and water (0.2 ml). The reaction mixture was heated and stirred at 12O0C under microwave irradiation for 1 h. Additional chloroP'^dimethylamino^-biphenylyOpalladium-^I R^S^bicycloP^.ilhept^- yl[(1 S,4/?)-bicyclo[2.2.1]hept-2-yl]phosphane (11.5 mg, 0.021 mmol) and potassium phosphate tribasic (80 mg) were added and the reaction heated to 12O0C under microwave irradiation for 1 h. Additional potassium phospate tribasic (80 mg) was added and the reaction heated under the same conditions for a further 1 h. The reaction mixture was filtered through a silica SPE and eluted with methanol. The solvent was removed in vacuo and the residue partitioned between dichloromethane (5 ml) and water (5 ml). The layers were separated and the aqueous extracted with further dichloromethane (2x 2 ml). The combined organics were concentrated under a stream of nitrogen and the residue dissolved in MeOH:DMSO (3ml, 1 :1 , v/v) and purified by MDAP (method A) in 3 injections. The appropriate fractions were combined and concentrated to give a white solid which was dissolved in MeOH:DMSO (1 ml, 1 :1 , v/v) and further purified by MDAP (method B). The appropriate fractions were basified to pH 6 with saturated sodium bicarbonate solution and extracted with ethyl acetate (2x 25 ml). The combined organics were dried and evaporated in vacuo to give a white solid which was further dried under nitrogen at 4O0C for 3 h to give the title compound as a white solid (26 mg). LCMS (Method A): Rt 0.53 mins, MH+ 513. Method B N-[2-(Methyloxy)-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3- pyridinyl]methanesulfonamide (101 g, 308 mmol), 6-chloro-4-(5-{[(2R,6S)-2,6-dimethyl-4- morpholinyl]methyl}-1 ,3-oxazol-2-yl)-1-(phenylsulfonyl)-1 H-indazole (83.3 g, 154 mmol) and sodium bicarbonate (38.8 g, 462 mmol) were suspended in 1 ,4-dioxane (1840 ml) and water (460 ml) under nitrogen and heated to 800C. Chloro[2'-(dimethylamino)-2- biphenylyl]palladium-1 (1 R,4S)-bicyclo[2.2.1]hept-2-yl[(1 S,4R)-bicyclo[2.2.1]hept-2- yl]phosphane (8.63 g, 15.40 mmol) was added and the mixture stirred overnight at 800C. The reaction mixture was cooled to 450C, sodium hydroxide 2M aq. (770 ml, 1540 mmol) added and the reaction heated to 45 0C for 4 hours. The mixture was cooled to RT and diluted with water (610 ml_). Dichloromethane (920 ml.) was added, and the mixture was filtered twice through Celite (washed with 200 ml. 1 ,4-dioxane/DCM 2:1 each time). The phases were separated, and aqueous washed with 1 ,4-dioxane/DCM 2:1 (500 ml_). The aqueous phase was neutralised with hydrochloric acid to pH -7 and extracted with 1 ,4- dioxane/DCM 2:1 (1 L), then 1 ,4 dioxane/DCM 1 :1 (2x500 ml_). The organics were washed with brine (500 ml_), and filtered through Celite (washed with 200 ml. 1 ,4 dioxane/DCM 2:1 ), and evaporated to yield a dark black solid, which was purified in 4 batches: Batch 1 : 28g was dissolved in Toluene/Ethanol/Ammonia 80:20:2 (100 ml.) and purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (14.78 g). Batch 2: 3Og was dissolved in methanol and mixed with Fluorisil. The solvent was then removed by evaporation and the solid purified by column chromatography (1.5 kg silica column, solid sample injection module), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (9.44 g). Batch 3: 31 g was dissolved in Toluene/Ethanol/Ammonia 80:20:2 (100 ml.) and purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (17 g). Batch 4: 29g was dissolved in Toluene/Ethanol/Ammonia 80:20:2 (100 ml.) and purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (21 g). The mixed fractions from the 4 columns were combined and evaporated to yield 19 g which was dissolved in 200 ml. of Toluene/Ethanol/Ammonia 80:20:2 (+ additional 4ml of 0.88 NH3 to help solubility) then purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (6.1 g). All pure batches were combined (68 g) and recrystallised from ethanol (1200 ml_). The suspension was heated to reflux and a solution formed. The resulting solution was then cooled to room temperature overnight. The resulting solid was then collected by filtration, washed sparingly with ethanol and dried under vacuum to give the title compound as an off-white solid (56 g). This material was recrystallised again from ethanol (1 100 ml_). The suspension was heated to reflux and a solution formed. The resulting solution was then cooled to room temperature overnight with stirring. The resulting solid was collected by filtration and washed sparingly with ethanol. The solid was dried in vacuo at 600C for 5hrs to give the title compound as an off-white solid (45.51 g). LCMS (Method A): Rt 0.61 mins, MH+ 513. The filtrate from the two recrystallisations was evaporated to yield -23 g of a solid residue that was dissolved in 200 ml. of Toluene/Ethanol/Ammonia 80:20:2 (+ additional 4ml of 0.88 NH3 to help solubility) then purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give a further crop of the title compound as an off-white solid (18.5 g). This solid was then recrystallised from ethanol (370 ml_). The suspension was heated to reflux then the resulting solution stirred for 20 mins before being allowed to cool to room temperature naturally overnight. The solid was then dried in vacuo at 65°C overnight to give the title compound as an off-white solid (11.9O g). LCMS (Method A): Rt 0.62 mins, MH+ 513. ...............................................

  http://www.google.co.in/patents/US8735390

 Example 1N-[5-[4-(5-{[(2R,6S)-2,6-Dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1H-indazol-6-yl]-2-(methyloxy)-3-pyridinyl]methanesulfonamide
Method A To a solution of 6-chloro-4-(5-{[(2R,6S)-2,6-dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1-(phenylsulfonyl)-1H-indazole (0.20 g, 0.411 mmol) and N-[2-(methoxy)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3-pyridyl]methanesulfonamide (0.175 g, 0.534 mmol) in 1,4-dioxane (2 ml) was added chloro[2′-(dimethylamino)-2-biphenylyl]palladium-1(1R,4S)-bicyclo[2.2.1]hept-2-yl[(1S,4R)-bicyclo[2.2.1]hept-2-yl]phosphane (11.5 mg, 0.021 mmol), potassium phosphate tribasic (0.262 g, 1.23 mmol) and water (0.2 ml). The reaction mixture was heated and stirred at 120° C. under microwave irradiation for 1 h. Additional chloro[2′-(dimethylamino)-2-biphenylyl]palladium-1(1R,4S)-bicyclo[2.2.1]hept-2-yl[(1S,4R)-bicyclo[2.2.1]hept-2-yl]phosphane (11.5 mg, 0.021 mmol) and potassium phosphate tribasic (80 mg) were added and the reaction heated to 120° C. under microwave irradiation for 1 h. Additional potassium phospate tribasic (80 mg) was added and the reaction heated under the same conditions for a further 1 h. The reaction mixture was filtered through a silica SPE and eluted with methanol. The solvent was removed in vacuo and the residue partitioned between dichloromethane (5 ml) and water (5 ml). The layers were separated and the aqueous extracted with further dichloromethane (2×2 ml). The combined organics were concentrated under a stream of nitrogen and the residue dissolved in MeOH:DMSO (3 ml, 1:1, v/v) and purified by MDAP (method A) in 3 injections. The appropriate fractions were combined and concentrated to give a white solid which was dissolved in MeOH:DMSO (1 ml, 1:1, v/v) and further purified by MDAP (method B). The appropriate fractions were basified to pH 6 with saturated sodium bicarbonate solution and extracted with ethyl acetate (2×25 ml). The combined organics were dried and evaporated in vacuo to give a white solid which was further dried under nitrogen at 40° C. for 3 h to give the title compound as a white solid (26 mg). LCMS (Method A): Rt 0.53 mins, MH+ 513. Method B N-[2-(Methyloxy)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3-pyridinyl]methanesulfonamide (101 g, 308 mmol), 6-chloro-4-(5-{[(2R,6S)-2,6-dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1-(phenylsulfonyl)-1H-indazole (83.3 g, 154 mmol) and sodium bicarbonate (38.8 g, 462 mmol) were suspended in 1,4-dioxane (1840 ml) and water (460 ml) under nitrogen and heated to 80° C. Chloro[2′-(dimethylamino)-2-biphenylyl]palladium-1(1R,4S)-bicyclo[2.2.1]hept-2-yl[(1S,4R)-bicyclo[2.2.1]hept-2-yl]phosphane (8.63 g, 15.40 mmol) was added and the mixture stirred overnight at 80° C. The reaction mixture was cooled to 45° C., sodium hydroxide 2M aq. (770 ml, 1540 mmol) added and the reaction heated to 45° C. for 4 hours. The mixture was cooled to RT and diluted with water (610 mL). Dichloromethane (920 mL) was added, and the mixture was filtered twice through Celite (washed with 200 mL 1,4-dioxane/DCM 2:1 each time). The phases were separated, and aqueous washed with 1,4-dioxane/DCM 2:1 (500 mL). The aqueous phase was neutralised with hydrochloric acid to pH ˜7 and extracted with 1,4-dioxane/DCM 2:1 (1 L), then 1,4 dioxane/DCM 1:1 (2×500 mL). The organics were washed with brine (500 mL), and filtered through Celite (washed with 200 mL 1,4 dioxane/DCM 2:1), and evaporated to yield a dark black solid, which was purified in 4 batches:
  • Batch 1: 28 g was dissolved in Toluene/Ethanol/Ammonia 80:20:2 (100 mL) and purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (14.78 g).
  • Batch 2: 30 g was dissolved in methanol and mixed with Fluorisil. The solvent was then removed by evaporation and the solid purified by column chromatography (1.5 kg silica column, solid sample injection module), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (9.44 g).
  • Batch 3: 31 g was dissolved in Toluene/Ethanol/Ammonia 80:20:2 (100 mL) and purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (17 g).
  • Batch 4: 29 g was dissolved in Toluene/Ethanol/Ammonia 80:20:2 (100 mL) and purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (21 g).
The mixed fractions from the 4 columns were combined and evaporated to yield 19 g which was dissolved in 200 mL of Toluene/Ethanol/Ammonia 80:20:2 (+additional 4 ml of 0.88 NH3 to help solubility) then purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give the title compound as an off-white solid (6.1 g). All pure batches were combined (68 g) and recrystallised from ethanol (1200 mL). The suspension was heated to reflux and a solution formed. The resulting solution was then cooled to room temperature overnight. The resulting solid was then collected by filtration, washed sparingly with ethanol and dried under vacuum to give the title compound as an off-white solid (56 g). This material was recrystallised again from ethanol (1100 mL). The suspension was heated to reflux and a solution formed. The resulting solution was then cooled to room temperature overnight with stirring. The resulting solid was collected by filtration and washed sparingly with ethanol. The solid was dried in vacuo at 60° C. for 5 hrs to give the title compound as an off-white solid (45.51 g). LCMS (Method A): Rt 0.61 mins, MH+ 513. The filtrate from the two recrystallisations was evaporated to yield ˜23 g of a solid residue that was dissolved in 200 mL of Toluene/Ethanol/Ammonia 80:20:2 (+additional 4 ml of 0.88 NH3 to help solubility) then purified by column chromatography (1.5 kg silica column), eluting with Toluene/Ethanol/Ammonia 80:20:2 to give a further crop of the title compound as an off-white solid (18.5 g). This solid was then recrystallised from ethanol (370 mL). The suspension was heated to reflux then the resulting solution stirred for 20 mins before being allowed to cool to room temperature naturally overnight. The solid was then dried in vacuo at 65° C. overnight to give the title compound as an off-white solid (11.90 g). LCMS (Method A): Rt 0.62 mins, MH+ 513. Method C 10M Sodium hydroxide solution (0.70 ml) was added to a stirred suspension of N-[5-[4-(5-{[(2R,6S)-2,6-dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1-(phenylsulfonyl)-1H-indazol-6-yl]-2-(methyloxy)-3-pyridinyl]methanesulfonamide (1.17 g) in water (5.8 ml). The resulting mixture was stirred at room temperature for 3.75 hours and was then washed with ethyl acetate (2×6 ml). The layers were separated and the aqueous phase was acidified to pH 6 with 2M hydrochloric acid (0.8 ml). The acidified aqueous layer was extracted twice with ethyl acetate (11 ml then 5 ml). The combined ethyl acetate extracts were dried by azeotropic distillation and diluted with further ethyl acetate (11 ml). The misture was stirred at room temperature for 112 hours. The slurry was seeded and then stirred at room temperature for 48 hours. The resultant suspension was filtered, washed with ethyl acetate (2×2 ml) and the solid dried under vacuum at 40° C. to give the title compound as a pale yellow solid (0.58 g). LCMS (Method B): Rt 1.86 min, MH+ 513. Method D To a suspension of N-[5-[4-(5-{[(2R,6S)-2,6-dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1-(phenylsulfonyl)-1H-indazol-6-yl]-2-(methyloxy)-3-pyridinyl]methanesulfonamide (596.5 g, 0.91 mol) in water (3.8 L) is added 5M sodium hydroxide (715 ml, 3.56 mol) over 20 mins at <25° C. The mixture is stirred at 20±3° C. for 2 h 45 min then washed with EtCN (3 L). The pH of the basic aqueous phase is adjusted to pH 6.6 using 2M hydrochloric acid (1.4 L), maintaining the temperature below 30° C. The mixture is then extracted with MeTHF (2×4.8 L), and the combined MeTHF extracts are washed with water (1.2 L). The mixture is concentrated to approx 2.4 L and EtOAc (3 L) is added. This put and take distillation is repeated a further 3 times. The mixture is adjusted to 60±3° C. and seeded twice (2×3 g) 35 mins apart. The resultant is aged for 1 h 10 mins then cooled over 2 h to 20-25° C., and aged for a further 15 h 50 min. The slurry is filtered, washed with EtOAc (2×1.2 L) and dried in vacuo at 45±5° C. for approx 3 day to give the title compound. Preparation of Polymorphs of Compound A Form (II) Ethyl acetate (15 ml) was added to N-[5-[4-(5-{[(2R,6S)-2,6-dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1H-indazol-6-yl]-2-(methyloxy)-3-pyridinyl]methanesulfonamide (2.1 g) and was stirred at ambient conditions overnight. The resultant slurry was filtered and dried under vacuum at 50° C. to give a new solid state form (91 ckw/w). 1H NMR (400 MHz, DMSO d6) d=13.49 (br s, 1H), 9.39 (s, 1H), 8.58 (s, 1H), 8.42 (d, J=2.2 Hz, 1H), 7.99 (d, J=2.2 Hz, 1H), 7.93 (d, J=1.2 Hz, 1H), 7.88 (s, 1H), 7.35 (s, 1H), 4.00 (s, 3H), 3.74 (s, 2H), 3.58 (m, 2H), 3.11 (s, 3H), 2.80 (d, J=10.3 Hz, 2H), 1.78 (t, J=10.3 Hz, 2H), 1.05 (d, J=6.4 Hz, 6H)   SODIUM SALT OF COMPD B http://www.google.com/patents/US20140256721 Method D To a suspension of N-[5-[4-(5-{[(2R,6S)-2,6-dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1-(phenylsulfonyl)-1H-indazol-6-yl]-2-(methyloxy)-3-pyridinyl]methanesulfonamide (596.5 g, 0.91 mol) in water (3.8 L) is added 5M sodium hydroxide (715 ml, 3.56 mol) over 20 mins at <25° C. The mixture is stirred at 20±3° C. for 2 h 45 min then washed with EtCN (3 L). The pH of the basic aqueous phase is adjusted to pH 6.6 using 2M hydrochloric acid (1.4 L), maintaining the temperature below 30° C. The mixture is then extracted with MeTHF (2×4.8 L), and the combined MeTHF extracts are washed with water (1.2 L). The mixture is concentrated to approx 2.4 L and EtOAc (3 L) is added. This put and take distillation is repeated a further 3 times. The mixture is adjusted to 60±3° C. and seeded twice (2×3 g) 35 mins apart. The resultant is aged for 1 h 10 mins then cooled over 2 h to 20-25° C., and aged for a further 15 h 50 min. The slurry is filtered, washed with EtOAc (2×1.2 L) and dried in vacuo at 45±5° C. for approx 3 day to give the title compound. http://www.google.com/patents/US20140256721 Preparation of Salts of Compound ASodium Salt Methanol (2 ml) was added to N-[5-[4-(5-{[(2R,6S)-2,6-dimethyl-4-morpholinyl]methyl}-1,3-oxazol-2-yl)-1H-indazol-6-yl]-2-(methyloxy)-3-pyridinyl]methanesulfonamide (0.3 g) followed by aqueous sodium hydroxide (0.129 ml) to give a solution. Tert-butylmethylether (4 ml) was added to the solution followed by seed crystals of the sodium salt and this suspension was stirred overnight at ambient conditions. The suspension was filtered, washed with tert-butylmethylether (2 ml) and air dried to give the sodium salt (0.2312 g) as a hydrate. NMR: Consistent with salt formation 1H NMR (400 MHz, DMSO d6) d=13.35 (br s, 1H), 8.53 (s, 1H), 7.90 (d, J=1.2 Hz, 1H), 7.73 (s, 1H), 7.65 (d, J=2.5 Hz, 1H), 7.62 (d, J=2.2 Hz, 1H), 7.33 (s, 1H), 4.00 (s, 3H), 3.80 (s, 3H), 3.59 (m, 2H). 2.83 (d, J=10.3, 2H), 2.61 (s, 3H), 1.78 (t, J=10.5 Hz, 2H), 1.05 (d, J=6.1 Hz, 6H)    

EMAIL ME amcrasto@gmail.com


 EMAIL ME amcrasto@gmail.com
US20100280029 * 28 Apr 2010 4 Nov 2010 Julie Nicole Hamblin Novel compounds
WO2010125082A1 28 Apr 2010 4 Nov 2010 Glaxo Group Limited Oxazole substituted indazoles as pi3-kinase inhibitors
US20140256721 * 14 Apr 2014 11 Sep 2014 Glaxosmithkline Intellectual Property Development Limited Novel Polymorphs and Salts
WO2012032065A1 6 Sep 2011 15 Mar 2012 Glaxo Group Limited Indazole derivatives for use in the treatment of influenza virus infection
WO2012032067A1 6 Sep 2011 15 Mar 2012 Glaxo Group Limited Polymorphs and salts of n- [5- [4- (5- { [(2r,6s) -2, 6 - dimethyl - 4 -morpholinyl] methyl} - 1, 3 - oxazol - 2 - yl) - 1h- inda zol-6-yl] -2- (methyloxy) - 3 - pyridinyl] methanesulfonamide
WO2012055846A1 25 Oct 2011 3 May 2012 Glaxo Group Limited Polymorphs and salts of 6-(1h-indol-4-yl)-4-(5- { [4-(1-methylethyl)-1-pi perazinyl] methyl} -1,3-oxazol-2-yl)-1h-indazole as pi3k inhibitors for use in the treatment of e.g. respiratory disorders
WO2012064744A2 * 8 Nov 2011 18 May 2012 Lycera Corporation Tetrahydroquinoline and related bicyclic compounds for inhibition of rorϒ activity and the treatment of disease
WO2013088404A1 14 Dec 2012 20 Jun 2013 Novartis Ag Use of inhibitors of the activity or function of PI3K
WO2014068070A1 31 Oct 2013 8 May 2014 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for preventing antiphospholipid syndrome (aps)
US8524751 5 Mar 2010 3 Sep 2013 GlaxoSmithKline Intellecutual Property Development 4-oxadiazol-2-YL-indazoles as inhibitors of P13 kinases
US8536169 3 Jun 2009 17 Sep 2013 Glaxo Group Limited Compounds
US8575162 28 Apr 2010 5 Nov 2013 Glaxosmithkline Intellectual Property Development Limited Compounds
US8580797 28 Apr 2010 12 Nov 2013 Glaxo Smith Kline Intellectual Property Development Limited Compounds
US8586583 2 Oct 2012 19 Nov 2013 Glaxosmithkline Intellectual Property Development Limited Compounds
US8586590 2 Oct 2012 19 Nov 2013 Glaxosmithkline Intellectual Property Development Limited Compounds
US8609657 2 Oct 2012 17 Dec 2013 Glaxosmithkline Intellectual Property Development Limited Compounds
US8658635 3 Jun 2009 25 Feb 2014 Glaxosmithkline Intellectual Property Development Limited Benzpyrazol derivatives as inhibitors of PI3 kinases
US8735390 6 Sep 2011 27 May 2014 Glaxosmithkline Intellectual Property Development Limited Polymorphs and salts
US8765743 3 Jun 2009 1 Jul 2014 Glaxosmithkline Intellectual Property Development Limited Compounds

Saturday 14 March 2015

AMG 925 from Amgen

AMG 925

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

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

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

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

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

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

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

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


…………………………

PAPER

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

References:

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