AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

AMG 900, An aurora kinase (ARK) inhibitor potentially for the treatment of leukemia and solid tumours

 phase 1, Uncategorized  Comments Off on AMG 900, An aurora kinase (ARK) inhibitor potentially for the treatment of leukemia and solid tumours
Jun 152016
 

AMG-900

N-(4-((3-(2-aminopyrimidin-4-yl)pyridin-2-yl)oxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine.

N-(4-(3-(2-Aminopyrimidin-4-yl)pyridin-2-yloxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine

Phase I

Amgen Inc. INNOVATOR

Inventors Victor J. Cee, Holly L. Deak, Bingfan Du,Stephanie D. Geuns-Meyer, Brian L. Hodous,Hanh Nho Nguyen, Philip R. Olivieri, Vinod F. Patel, Karina Romero, Laurie Schenkel,Less «
Applicant Amgen Inc.

An aurora kinase (ARK) inhibitor potentially for the treatment of leukemia and solid tumours.

CAS No. 945595-80-2

In 2014, orphan drug designation was assigned in the U.S. for the treatment of ovarian cancer

Molecular Formula: C28H21N7OS
Molecular Weight: 503.57764 g/mo
AMG 900; AMG-900; 945595-80-2; AMG900; UNII-9R2G075611; N-(4-((3-(2-aminopyrimidin-4-yl)pyridin-2-yl)oxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine;

AMG 900 is a small-molecule inhibitor of Aurora kinases A, B and C with potential antineoplastic activity. Aurora kinase inhibitor AMG 900 selectively binds to and inhibits the activities of Aurora kinases A, B and C, which may result in inhibition of cellular division and proliferation in tumor cells that overexpress these kinases. Aurora kinases are serine-threonine kinases that play essential roles in mitotic checkpoint control during mitosis and are overexpressed by a wide variety of cancer cell types. Check for active clinical trials or closed clinical trials using this agent

AMG 900 is a potent and highly selective pan-Aurora kinases inhibitor for Aurora A/B/C with IC50 of 5 nM/4 nM /1 nM;  >10-fold selective for Aurora kinases than p38α, Tyk2, JNK2, Met and Tie2.
IC50 Value: 5 nM(Aurora A); 4 nM(Aurora B); 1 nM(Aurora C)
Target: pan-Aurora
in vitro: AMG 900 is a novel class of ATP-competitive phthalazinamine small molecule inhibitors of aurora kinases. In HeLa cells, AMG 900 inhibits autophosphorylation of aurora-A and -B as well as phosphorylation of histone H3 on Ser, a proximal substrate of aurora-B. The predominant cellular response of tumor cells to AMG 900 treatment is aborted cell division without a prolonged mitotic arrest, which ultimately results in cell death. AMG 900 inhibits the proliferation of 26 tumor cell lines, including cell lines resistant to the antimitotic drug paclitaxel and to other aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358), at low nanomolar concentrations (about 2- 3 nM). Furthermore, AMG 900 is active in an AZD1152-resistant HCT116 variant cell line that harbors an aurora-B mutation (W221L) [1].
in vivo: Oral administration of AMG 900 blocks the phosphorylation of histone H3 in a dose-dependent manner and significantly inhibited the growth of HCT116 tumor xenografts. AMG 900 is broadly active in multiple xenograft models, including 3 multidrugresistant xenograft models, representing 5 tumor types [1]. AMG 900 exhibits a low-to-moderate clearance and a small volume of distribution. Its terminal elimination half-life ranged from 0.6 to 2.4 hours. AMG 900 is well-absorbed in fasted animals with an oral bioavailability of 31% to 107%. Food intake has an effect on rate (rats) or extent (dogs) of AMG 900 oral absorption. The clearance and volume of distribution at steady state in humans are predicted to be 27.3 mL/h/kg and 93.9 mL/kg, respectively. AMG 900 exhibits acceptable PK properties in preclinical species and is predicted to have low clearance in humans [2].

In mammalian cells, the aurora kinases (aurora-A, -B, and -C) play essential roles in regulating cell division. The expression of aurora-A and -B is elevated in a variety of human cancers and is associated with high proliferation rates and poor prognosis, making them attractive targets for anticancer therapy. AMG 900 is an orally bioavailable, potent, and highly selective pan-aurora kinase inhibitor that is active in taxane-resistant tumor cell lines. In tumor cells, AMG 900 inhibited autophosphorylation of aurora-A and -B as well as phosphorylation of histone H3 on Ser(10), a proximal substrate of aurora-B. The predominant cellular response of tumor cells to AMG 900 treatment was aborted cell division without a prolonged mitotic arrest, which ultimately resulted in cell death. AMG 900 inhibited the proliferation of 26 tumor cell lines, including cell lines resistant to the antimitotic drug paclitaxel and to other aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358), at low nanomolar concentrations. Furthermore, AMG 900 was active in an AZD1152-resistant HCT116 variant cell line that harbors an aurora-B mutation (W221L). Oral administration of AMG 900 blocked the phosphorylation of histone H3 in a dose-dependent manner and significantly inhibited the growth of HCT116 tumor xenografts. Importantly, AMG 900 was broadly active in multiple xenograft models, including 3 multidrug-resistant xenograft models, representing 5 tumor types. AMG 900 has entered clinical evaluation in adult patients with advanced cancers and has the potential to treat tumors refractory to anticancer drugs such as the taxanes.

MG 900 is an orally bioavailable, potent, and highly selective pan-aurora kinase inhibitor that is active in taxane-resistant tumor cell lines. In tumor cells, AMG 900 inhibited autophosphorylation of aurora-A and -B as well as phosphorylation of histone H3 on Ser10, a proximal substrate of aurora-B. The predominant cellular response of tumor cells to AMG 900 treatment was aborted cell division without a prolonged mitotic arrest, which ultimately resulted in cell death. AMG 900 inhibited the proliferation of 26 tumor cell lines, including cell lines resistant to the antimitotic drug paclitaxel and to other aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358), at low nanomolar concentrations. Furthermore, AMG 900 was active in an AZD1152-resistant HCT116 variant cell line that harbors an aurora-B mutation (W221L). Oral administration of AMG 900 blocked the phosphorylation of histone H3 in a dose-dependent manner and significantly inhibited the growth of HCT116 tumor xenografts. Importantly, AMG 900 was broadly active in multiple xenograft models, including 3 multidrug-resistant xenograft models, representing 5 tumor types. AMG 900 has entered clinical evaluation in adult patients with advanced cancers and has the potential to treat tumors refractory to anticancer drugs such as the taxanes. (Source: Cancer Res; 70(23); 9846–54.)

Clinical Information of AMG 900

Product Name Sponsor Only Condition Start Date End Date Phase Last Change Date
AMG 900 Amgen Inc Leukemia 31-JUL-11 31-JUL-14 Phase 1 14-SEP-13
Amgen Inc Advanced solid tumor 30-APR-09 30-JUN-13 Phase 1 10-SEP-13

AMG 900.png

PATENT

WO 2007087276

http://www.google.co.in/patents/WO2007087276A1?cl=en

PATENT

WO 2015084649

https://google.com/patents/WO2015084649A1?cl=en

The compound, N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)- 4-(4-methyl-2-thienyl)-l-phthalazinamine, also chemically named as 4-((3-(2-amino- pyrimidin-4-yl)-pyridin-2-yl)oxy)phenyl-(4-(4-methyl-thiophen-2-yl)-phthalazin-l- yl)amine, and is referred to herein as “AMG 900” has a chemical structure of

AMG 900 is an ATP competitive small molecule Aurora kinase inhibitor that is highly potent and selective for Aurora kinases A, B and C. AMG 900 is disclosed in US patent publication no. 20070185111, which published on August 9, 2007 and issued as U.S. Patent No. 7,560,551. AMG 900 is further disclosed in US patent publication no.

20090163501, now US patent no 8,022,221. Various uses and applications of AMG 900 are described in patent publications US20120028917 and WO2013149026. AMG 900 is being clinically evaluated primarily for its safety, tolerability and pharmacokinetic (PK) profile in human phase I trials for (1) advanced solid tumors (US Clinical Trial Id No. NCT00858377), and (2) for acute leukemias (US Clinical Trial Id No. NCT1380756).

Different solid state forms of a given compound are typically investigated to determine whether or not a particular form possesses and/or exhibits desirable properties allowing that compound to be clinically and/or commercially developed. Such beneficial and advantageous properties, by way of example, include without limitation, crystallinity, improved thermodynamic stability, non-hygroscopicity, high purity, minimal to total absence of moisture and/or residual solvents, chemical stability, high yielding synthetic process and/or manufacturability and reproducibility, desirable biopharmaceutical properties including improved dissolution characteristics and increased bioavailability, absence or reduced toxicities due to reduced or limited exposure, rate of exposure or release, or related to counter ions, good bulk and formulation properties including good flow, bulk density, desirable particle size and the like, or a combination of the aforementioned characteristic attributes.

Generally when a compound, also referred to herein as drug substance (DS), has been identified as a developmental candidate, the DS is screened to identify potentially beneficial polymorphic, crystalline or solid state forms of the compound and/or a pharmaceutically acceptable salt thereof. X-ray diffraction, Raman, solid state NMR and a melting point temperature and/or a melting point temperature range have been typically used to monitor or screen and identify the different polymorphic form of the DS.

Different polymorphic forms of a given DS can have an impact on that compound’s solubility, stability and bioavailability. Also, it is important to monitor possible changes in polymorphic forms of the DS during stability studies.

AMG 900 was previously isolated and identified as a free base compound. This compound exhibited rather lack-luster pharmacokinetic (PK) and/or pharmacodynamic (PD) properties, including poor aqueous solubility, poor bioavailability, poor absorption, poor target exposure and overall, a not-so-attractive in-vivo efficacy profile. Thus, there is a need to address and solve the technical problem of identifying alternative forms of AMG 900 to achieve substantially the same effect or an improved effect, including improved PK and PD profiles, as that of AMG 900 known in the art.

 

Example 1

Synthesis of N-(4-((3-(2-amino-4-pyrimidinylN)-2-pyridinylN)oxyN)phenylN)-4-(4-methyl-2-thienvD-l-phthalazinamine (AMG 900)

Step 1 : 4-(2-chloropyridin-3-yl)pyrimidin-2 -amine

In an argon purged 500 mL round bottom flask placed in an isopropanol bath, was added sodium metal (3.40g, 148mmol) slowly to methanol (180mL). The mixture was stirred at room temperature (RT) for about 30 minutes. To this was added guanidine hydrochloride (12.0 mL, 182 mmol) and the mixture was stirred at RT for 30 minutes, followed by addition of (E)-l-(2-chloropyridin-3-yl)-3-(dimethylamino)prop-2-en-l-one (12.0 g, 57.0 mmol), attached air condenser, moved reaction to an oil bath, where it was heated to about 50 °C for 24 hr. Approximately half of the methanol was evaporated under reduced pressure and the solids were filtered under vacuum, then washed with saturated sodium bicarbonate (NaHCO and H^O, air dried to yield 4-(2-chloropyridin-3-yl)pyrimidin-2-amine as off white solid. MS m/z = 207 [M+l]+. Calc’d for C9H7C1N4: 206.63.

Step 2: 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2-amine

To a resealable tube was added 4-aminophenol (1.3 g, 12 mmol), cesium carbonate (7.8 g, 24 mmol), and DMSO (16 ml, 0.75 M). The mixture was heated to 100 °C for 5 minutes, and then 4-(2-chloropyridin-3-yl)pyrimidin-2 -amine (2.5 g, 12 mmol) was added, and the reaction mixture was heated to 130 °C overnight. Upon completion, as judged by LCMS, the reaction mixture was allowed to cool to RT and diluted with water. The resulting precipitate was filtered, and the solid washed with water and diethyl ether. The solid was then taken up in 9: 1 CH2Cl2:MeOH and passed through a pad of silica gel with 9:1 CH2Cl2:MeOH as eluent. The solvent was concentrated in vacuo to provide the desired product, 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2-amine. MS m/z = 280

[M+l]+. Calc’d for Ci5H13N50: 279.30.

Step 3: l-Chloro-4-(4-methylthiophen-2-yl)phthalazine

1 ,4-Dichlorophthalazine (1.40 g, 7.03 mmol), 4-methyltmophen-2-ylboronic acid (999 mg, 7.03 mmol), and PdCl2(DPPF) (721 mg, 985 μιηοΐ) were added into a sealed tube. The tube was purged with Argon. Then sodium carbonate (2.0 M in water) (7.74 ml, 15.5 mmol) and 1,4-dioxane (35.2 ml, 7.03 mmol) were added. The tube was sealed, stirred at RT for 5 min, and placed in a preheated oil bath at 110 °C. After 1 hr, LC-MS showed product and byproduct (double coupling), and starting material

dichlorophthalazme. The reaction was cooled to RT, filtered through a pad of celite with an aid of ethyl acetate (EtOAc), concentrated, and loaded onto column. The product was purified by column chromatography using Hex to remove the top spot, then 80:20 hexanes:EtOAc to collect the product. The product, 1 -chloro-4-(4-methylthiophen-2-yl)phthalazine was obtained as yellow solid. LC-MS showed that the product was contaminated with a small amount of dichlorophthalazme and biscoupling byproduct. MS m/z = 261 [M+l]+. Calcd for Ci3H9ClN2S: 260.12.

Step 4: N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)- 1 -phthalazinamine

To 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2 -amine and l-chloro-4-(4-methyl-2-thienyl)phthalazine was added tBuOH. The resulting mixture was heated at 100 °C in a sealed tube for 16 hours. The rection was diluted with diethyl ether and saturated sodium carbonate and vigorously shaken. The resulting solids were filtered and washed with water, diethyl ether and air dried to yield N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-l -phthalazinamine as an off-white solid. MS m/z = 504 [M+H]+. Calc’d for C28 H21 N7 O S: 503.58.

Example 2

Alternative Synthesis of N-(4-((3-(2-amino-4-pyrimidinylN)-2-pyridinylN)oxyN)phenylN)-4-(4-methyl-2-thienvD-l-phthalazinamine (AMG 900)

Step 1 : 4-(2-chloropyridin-3-yl)pyrimidin-2 -amine

In an argon purged 500 mL round bottom flask placed in an isopropanol bath, was added sodium metal (3.40g, 148mmol) slowly to methanol (180mL). The mixture was stirred at room temperature (RT) for about 30 minutes. To this was added guanidine hydrochloride (12.0 mL, 182 mmol) and the mixture was stirred at RT for 30 minutes, followed by addition of (E)-l-(2-chloropyridin-3-yl)-3-(dimethylamino)prop-2-en-l-one (12.0 g, 57.0 mmol), attached air condenser, moved reaction to an oil bath, where it was heated to about 50 °C for 24 hr. Approximately half of the methanol was evaporated under reduced pressure and the solids were filtered under vacuum, then washed with saturated sodium bicarbonate (NaHCO and H^O, air dried to yield 4-(2-chloropyridin-3-yl)pyrimidin-2-amine as off white solid. MS m/z = 207 [M+l]+. Calc’d for C9H7C1N4: 206.63.

Step 2: 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2-amine

To a reaction vessel at ambient temperature was added 4-aminophenol (571 g, 5.25 mol, 1.05 equiv) followed by 4-(2-chloropyridin-3-yl)pyrimidin-2-amine (1064g, 97 wt%, 5.00 mol, 1.0 equiv) and DMSO (7110 ml, 7820 g, 7x the volume of 4-(2-chloropyridin-3-yl)pyrimidin-2 -amine). The reaction mixture was agitated and sparged with nitrogen gas for at least 10 minutes. Then a 50 weight % aqueous KOH solution (593 g, 5.25 mol, 1.05 equiv.) was added to the mixture while keeping the reaction

mixture temperature below about 40°C. The mixture was sparged with nitrogen gas for more than 5 minutes, the sparging tube was removed, and the reaction mixture was heated to 110 +/- 10 °C for at least 1.5 hrs. Upon completion, as judged by HPLC, the reaction mixture was allowed to cool to RT and diluted with 6N HC1 (42 mL, 0.25 mol, 0.05 equiv). The desired product, 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2 -amine was not isolated. Rather, it was formed in-situ and combined with the product of step 3 below, in step 4 to form the desired product.

Step 3: l-Chloro-4-(4-methylthiophen-2-yl)phthalazine

A separate reaction vessel was fitted with a reflux condenser and an addition funnel, and 4-(4-methylthiophen-2-yl)phthalazin-l(2H)-one (1,537 mg, 6.34 mol, 1.0 equivalent) was added to the reaction vessel. Acetonitrile (7540 mL, 5859 g, 5 V), was added and the reaction vessel was agitated to allow the starting material to dissolve. The vessel was then charged with phosphorus oxychloride (709 ml, 1166 g, 7.44 mol, 1.2 equivalents) and the reaction was heated to about 75 +/- 5 °C for a least 4 hrs. The reaction was cooled to about about 25 +/- 5 °C and held there for more than 24 hrs. N,N-diisopropylethylamine (3046 g, 4100 mL, 3.8 equivalents) was added to the reaction vessel and the temperature was maintained at <30°C. Pyridine (97g, 1.24 mol, 0.2 equiv) was added in a single portion followed by water (4100 g, 2.7V) over more than 30 minutes. The reaction mixture was stirred at ambient temperature ofr about 24 hrs. the mixture was filtered through a <25uM polypropylene filter and the rsulting mother liquor was diluted with 1 : 1 ACN:water (9000 mL total) and stirred for a minimum of 2 minutes. Filter off product solids as they precipitate. Collect mother liquor and washes to obtain additional product. Dry the filter cake, and additional product crops, under a constant stream of nitrogen gas for at least 14 hrs. Unlike the previous method, the present method avoids contamination of impurities, such as dichlorophthalazine and biscoupling byproduct, as seen via LC-MS. Yield: 1537 g (97.2 weight %). MS m/z = 261 [M+l]+. Calcd for Ci3H9ClN2S: 260.12.

Step 4: N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)- 1 -phthalazinamine

To the reaction mixture was added l-chloro-4(4-methylthiophen-2-yl)phthalazine

(1450g, 97.2 wt%, 5.40 mol, 1.08 equiv) rinding the addition port with DMSO (520 ml, 572 g, 0.5x the volume of 4-(2-chloropyridin-3-yl)pyrimidin-2-amine). The reaction mixture was again agitated and sparged with nitrogen gas for at least 10 minutes. The sparging tube was removed, and the reaction mixture was heated to 80 +/- 20 °C for at least 2 hrs. Upon completion, as judged by HPLC, the reaction mixture was allowed to cool to RT and N,N-diisopropylethylamine (776 g, 1045 mL, 6.0 mol, 1.2 equiv) was added and the mixture was kept at about 80 +/- 10°C. Filter the mixture at about 80oC into a separate reactor vessel rinsing with DMSO (1030 mL, 1133 g, 1 V). Then adjust the raction mixture temperature to about 70+/-5 °C and add 2-propanol (13200 mL, 10360 g, 12.75 V) over more than 15 minutes at about 70°C. As the reaction mistreu cools, the product begins to precipitate out of solution. Add more 2-propanol (8780 mL, 6900 g, 8.5V) to the solution slowly over more then 60 minutes at about 70°C. The reactor vessel was cooled to about 20°C over more than 60 minutes and let sit for over 2 hrs. The product was filtered through an Aurora filter with a >25uM polypropylene filter cloth. Additional crops were obtained from the mother liquors by diluting with additional 2-propanol. The filter cakes were dried under a constant stream of nitrogen gas for at least 14 hrs to provide the desired product, N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-l-phthalazinamine as an off-white solid. Yield: 2831 g (88.8%); purity 99.7%. MS m/z = 504 [M+H]+. Calc’d for C28 H21 N7 O S: 503.58.

The starting material 1 used/shown in Example 2 was prepared as follows:

and starting material 3, thienyl substituted phthalazinone, shown in Example 2 was prepared as follows:

Starting material 3

Synthesis of 4-(5-methylthiophen-2-yl)phthalazin-l(2//)-one

Step 1 : 2-(Dimethylamino)isoindoline-1.3-dione

A solution of isobenzofuran-l,3-dione (5.00 g, 34 mmol) and N,N-dimethylhydrazine (2.9 ml, 37 mmol) in toluene (75 ml, 34 mmol) was added p-TsOH H20 (0.32 g, 1.7 mmol). The Dean-Stark apparatus and a condenser were attached. The mixture was refluxed. After 4 hr, LCMS showed mainly product. The reaction was cooled to rt. Toluene was removed under reduced pressure the crude was dissolved in DCM, washed with sat NaHC03, water, and brine. The organic was dried over MgS04, filtered, and concentrated. Light yellow solid was obtained. !H NMR showed mainly product, 2-(dimethylamino)isoindoline-l,3-dione. MS Calcd for C10H10N2O2: [M]+ = 190. Found: [M+H]+ = 191.

Step 2 : 2-(Dimethylamino)-3 -hydroxy-3 -(5 -methylthiophen-2 -vDisoindolin- 1 -one

A solution of 2-bromo-5-methylthiophene (0.60 mL, 5.3 mmol) in THF (11 mL) was purged with nitrogen and cooled to -78 °C. «-Butyllithium (2.2 mL, 5.5 mmol; 2.5 M in THF) was added and the mixture was stirred under nitrogen for 30 min. This solution was cannulated into a flask containing a solution of 2-(dimethylamino)isoindoline-l,3-dione (1.5 g, 7.9 mmol) in THF (16 mL) at -78 °C under nitrogen. The reaction was allowed to warm to -30 °C over an hour, at which point LCMS showed complete conversion of 2-bromo-5-methylthiophene to product. The reaction was quenched by careful addition of saturated aqueous NH4C1. The reaction mixture was diluted with dichloromethane and water, and the layers were separated. The aqueous portion was extracted with additional dichloromethane, and the combined organic layers were dried with MgS04, filtered, concentrated, and purified by silica gel chromatography eluting with 0-2% MeOH in dichloromethane to provide intermediate A, as a light yellow solid, 2-(dimethylamino)-3-hydroxy-3-(5-methylthiophen-2-yl)isoindolin-l-one (1.2 g, 80% yield). !H NMR (400 MHz, DMSO-4) δ 7.68-7.65 (m, 1H). 7.63-7.59 (m, 1H), 7.57-7.51 (m, 1H), 7.37 (d, 1H, J=8), 7.09 (s, 1H), 6.69-6.66 (m, 1H), 6.65-6.62 (m, 1H), 2.81 (s, 6H), 2.40 (s, 3H). 13C NMR (400 MHz, DMSO-de) δ 165.0, 147.3, 141.6, 139.3, 132.7, 129.49, 129.46, 125.0, 124.7, 123.0, 122.1, 88.4, 44.7, 14.9. FT-IR (thin film, cm ) 3347, 3215, 1673. MS Calcd for Ci2H7ClN2S: [M]+ = 288. Found: [M+H]+= 289.

HRMS Calcd for Ci5H16N202S: [M+H]+= 288.1005, [M+Na]+ = 311.0825. Found:

[M+H]+ = 289.1022, [M+Na]+= 311.0838. mp = 138-140 °C.

Step 3: 4-(5-Methylthiophen-2-yl)phthalazin-l(2//)-one

2-(Dimethylamino)-3 -hydroxy-3 -(5 -methylthiophen-2-yl)isoindolin- 1 -one (1.1 g, 0.40 mmol), EtOH (4.0 mL), and hydrazine (0.19 mL, 59 mmol) were added into a RBF fitted with a reflux condenser. A nitrogen balloon was attached on top of the condenser. After refluxing overnight, the reaction was cooled to room temperature. An off-white solid precipitated. After cooling to 0 °C, water was added. The solid was filtered off with an aid of water and dried under vacuum to afford a white solid, 4-(5-methylthiophen-2-yl)phthalazin-l(2//)-one (0.82 g, 85% yield).

!H NMR (400 MHz, CDC13) δ 10.57 (s, 1H), 8.50-8.39 (m, 1H), 8.14-8.04 (m, 1H), 7.83- 7.69 (m, 2H), 7.20-7.17 (m, 1H), 6.82-6.71 (m, 1H), 2.47 (s, 3H). 13C NMR (400 MHz,

CDC13) 8 159.9, 142.5, 141.1, 134.3, 133.7, 131.7, 129.4, 128.8, 128.3, 127.1, 126.6,

125.8, 15.4. FT-IR (thin film, cm“1) 2891, 1660, 1334. MS Calcd for Ci3H10N2OS: [M]+

= 242. Found: [M+H]+= 243. HRMS Calcd for Ci3H10N2OS: [M+H]+= 243.0587. Found:

[M+H]+ = 243.0581. mp = 191-194 °C.

Alternatively, starting material 3 was prepared as follows:

The above scheme depicts the process by which intermediate-scale synthesis of thiophene-phthalazinone 5 (shown above) was prepared. Treatment of 50 grams of 3-methylthiophene with z-PrMgCl at 66 °C in the presence of catalytic TMP-H resulted in 98% conversion to the reactive species lb with a >40:1 regioisomeric ratio. After cooling to 20 °C, this mixture was added dropwise to a -20 °C slurry of phthalic anhydride in THF to provide keto acid 3 in 94% assay yield. While this intermediate could be crystallized from toluene/heptane, the crude reaction mixture was taken directly in a through -process conversion to the phthalazinone 5. To that end, removal of THF, MTBE, and residual 3-methylthiophene was accomplished through a distillative solvent switch into ethanol. The resulting solution of 3 was exposed to aqueous hydrazine at 80 °C. After 18 hours, the reaction was cooled and the precipitated product was filtered directly at 20 °C. This process provided 82.7 grams of 98.6 wt % thiophene-phthalazinone 5 in a weight-adjusted 85% yield over the two steps.

LCMS Method:

Samples were run on a Agilent model- 1100 LC-MSD system with an Agilent Technologies XDB-C8 (3.5 μ) reverse phase column (4.6 x 75 mm) at 30 °C. The flow rate was constant and ranged from about 0.75 mL/min to about 1.0 mL/min.

The mobile phase used a mixture of solvent A (H2O/0.1% HO Ac) and solvent B

(AcCN/O.1 HOAc) with a 9 min time period for a gradient from 10%> to 90%> solvent B. The gradient was followed by a 0.5 min period to return to 10% solvent B and a 2.5 min 10% solvent B re-equilibration (flush) of the column.

Other methods may also be used to synthesize AMG 900. Many synthetic chemistry transformations, as well as protecting group methodologies, useful in synthesizing AMG 900, are known in the art. Useful organic chemical transformation literature includes, for example, R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley and Sons (1999); L. Fieser and M. Fieser, Fieser and Fieser’s Reagents for Organic Synthesis, John Wiley and Sons (1994); A. Katritzky and

A. Pozharski, Handbook of Heterocyclic Chemistry, 2nd edition (2001); M. Bodanszky, A. Bodanszky, The Practice of Peptide Synthesis, Springer- Verlag, Berlin Heidelberg (1984); J. Seyden-Penne, Reductions by the Alumino- and Borohydrides in Organic Synthesis, 2nd edition, Wiley- VCH, (1997); and L. Paquette, editor, Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995).

AMG 900 was tested for its ability to reduce or inhibit tumor progression in various cell lines (in-vitro) and multiple solid tumor types (in-vivo), some of which have previously been exposed to and developed resistance to standard-of-care antimitotic agents, including taxanes and vinca alkaloids, as well as to other chemotherapeutic agents. The following Examples and resulting data will illustrate the ability of AMG 900 to treat cancer, including cancer resistant to the presently standard-of-care therapies, including antimitotic agents, such as paclitaxel, and other drugs used in conjunction with chemotherapy, such as doxorubicin. Unless otherwise indicated, the free base form of AMG 900 was used in the Examples described hereinbelow.

The following Examples describe the efforts of identifying and characterizing various crystalline solid state forms of various salts of AMG 900. Some attempts at forming a solid state crystalline form of a given salt failed, as shown in table 1 hereinbelow. To this end, synthesizing and/or forming &isolating a crystalline solid state form of AMG 900 was not, in any way, straightforward or routine. Rather, the ability to prepare and identify a crystalline solid state form of AMG 900 depended upon the particular salt of AMG 900 and/or the crystallization conditions employed.

PAPER

Journal of Medicinal Chemistry (2015), 58(13), 5189-5207

Discovery of N-(4-(3-(2-Aminopyrimidin-4-yl)pyridin-2-yloxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine (AMG 900), A Highly Selective, Orally Bioavailable Inhibitor of Aurora Kinases with Activity against Multidrug-Resistant Cancer Cell Lines

Departments of Medicinal Chemistry, Pharmaceutical Research and Development, §Pharmacokinetics and Drug Metabolism, Molecular Structure, and Oncology Research, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States, and Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
J. Med. Chem., 2015, 58 (13), pp 5189–5207
DOI: 10.1021/acs.jmedchem.5b00183
*Phone: 617-444-5041. E-mail: MeyerS@amgen.com.

ACS Editors’ Choice – This is an open access article published under an ACS AuthorChoice License, which permits copying and redistribution of the article or any adaptations for non-commercial purposes.

Abstract

Abstract Image

Efforts to improve upon the physical properties and metabolic stability of Aurora kinase inhibitor14a revealed that potency against multidrug-resistant cell lines was compromised by increased polarity. Despite its high in vitro metabolic intrinsic clearance, 23r (AMG 900) showed acceptable pharmacokinetic properties and robust pharmacodynamic activity. Projecting from in vitro data to in vivo target coverage was not practical due to disjunctions between enzyme and cell data, complex and apparently contradictory indicators of binding kinetics, and unmeasurable free fraction in plasma. In contrast, it was straightforward to relate pharmacokinetics to pharmacodynamics and efficacy by following the time above a threshold concentration. On the basis of its oral route of administration, a selectivity profile that favors Aurora-driven pharmacology and its activity against multidrug-resistant cell lines, 23r was identified as a potential best-in-class Aurora kinase inhibitor. In phase 1 dose expansion studies with G-CSF support, 23r has shown promising single agent activity.

N-(4-(3-(2-Aminopyrimidin-4-yl)pyridin-2-yloxy)phenyl)-4-(4-methylthiophen-2-yl)phthalazin-1-amine (23r)

Applying similar SNAr conditions as for 23b, reaction of 22r and 20a in 2-butanol provided the title compound (2.08 g, 49%) as an off-white solid; mp (DSC) 216 °C.
1H NMR (400 MHz, DMSO-d6) δ ppm 9.36 (s, 1 H) 8.64–8.69 (m, 1 H) 8.41–8.44 (m, 1 H) 8.36–8.40 (m, 1 H) 8.35 (d, J = 5.2 Hz, 1 H) 8.23 (dd, J = 4.8, 2.0 Hz, 1 H) 8.00–8.10 (m, 2 H) 7.91–7.97 (m, 2 H) 7.52 (d, J = 1.0 Hz, 1 H) 7.26–7.33 (m, 3 H) 7.16–7.22 (m, 2 H) 6.74 (br s, 2 H) 2.34 (br s, 3 H).
13C NMR (150 MHz, DMSO-d6) δ 163.81, 160.72, 160.67, 158.68, 151.64, 148.50, 148.36, 147.14, 139.86, 139.24, 137.72, 137.10, 132.61, 131.74, 130.24, 125.27, 124.89, 122.92, 122.83, 122.44, 121.56, 121.52, 119.11, 118.23, 109.93, 15.6
HRMS m/z [M + H]+ Calcd for C28H21N7OS: 504.1601. Found: 504.1607.
Table 1. Aurora Kinase Inhibitors with Known Structures That Have Entered Clinical Trials

ID, compd name AKa AK cell assayb (nM) most potently inhibited non-AKs (nM)c [total kinases in panel] admin route
1 (MK-0457/VX-680/tozasertib)(7) A/B MDA-MB-231 p-HH3(12) 43 FLT3 (6), PLK4 (9), ABL (13), MLCK (15), RET (28); [317](16) IV
2 (PHA-739358/danusertib)(8) A/B MDA-MB-231 p-HH3(12) 49 ABL (25), RET (31), TrkA (31), FGFR1 (47); [35](17) IV
3a(AZD1152/barasertib)d,(9) B MDA-MB-231 p-HH3(12) 16 FLT3 (8), cKIT (17), PDGFRA (38), PDGFRB (41), RET (80); [317](16) IV
4 (AT9283)(18) A/B HCT-116 DNA ploidy ∼30 JAK2 (1), JAK3 (1) Abl (T315I) (4), 9 others ≤10 nM; [230] IV
5 (SNS-314)(19) A/B HCT-116 DNA ploidy(20) 9 TrkB (5), TrkA (12), FLT4 (14), Fms (15), DDR2 (82), Axl (84); [219] IV
6 (GSK1070916)(21) B HCT-116 p-HH3(22) 20 FLT1 (42), TIE2 (59), SIK (70), FLT4 (74), FGFR (78); [328](22) IV
7 (ENMD-2076)(23) A HCT-116 p-AurA 130 FLT3 (2), RET (10), FLT4 (16), SRC (20), TrkA (24), Fms (25); [100] PO
8 (CYC116)(24) A/B A549 p-HH3 480 VEGFR2 (44), FLT3 (44), CDK2 (390); [23] PO
9 (ABT-348)(25) A/B HCT-116 p-HH3 21 VEGFR1 (1), FLT3 (1), VEGFR2 (2), CSF-1R (3), PDGFR-α (11); [128] PO
10 (AS703569/R763)(26) A/B A549 p-HH3 14 cell-based assays: VEGFR2 (11), FLT3 (27), AMPK (201); [10] PO
11 (PF-03814735)(27) A/B MDA-MB-231 p-HH3 ∼50 FLT1 (10), FAK (22), TrkA (30), 17 others ≥90% inh@100 nM; [220] PO
12 (MK-5108)(28) A HeLa S3 ↑p-HH3+ cells <1000 TrkA (2), ABL (8), FLT4 (12), TrkB (13), VEGFR2 (30); [233] PO
13a (MLN8054)(29) A HCT-116 p-AurA 34 DRAK2 (8), BLK (68), DRAK1 (190), FGR (220); [317](16) PO
13b (MLN8237/alisertib)(30) A HeLa p-AurA 7 %inh@1 μM: EphA2 (111), FGR (97), CAMK2A (95), EphA4 (94); [220] PO

a

AK = Aurora kinase family member(s) inhibited (AurA and/or AurB; AurC potency not listed).

b

Cell line; substrate or phenotype detected.

c

Kinase activities of greatest potency listed in published literature.

d

Listed enzyme and cellular potency data is for 3b, the parent of prodrug 3a.

References on AMG 900

Patent ID Date Patent Title
US2016008316 2016-01-14 USE OF DIANHYDROGALACTITOL AND ANALOGS OR DERIVATIVES THEREOF IN COMBINATION WITH PLATINUM-CONTAINING ANTINEOPLASTIC AGENTS TO TREAT NON-SMALL-CELL CARCINOMA OF THE LUNG AND BRAIN METASTASES
US2016009785 2016-01-14 NOVEL FUSION MOLECULES AND USES THEREOF
US2015266868 2015-09-24 PHARMACEUTICALLY ACTIVE COMPOUNDS
US2015079022 2015-03-19 USE OF AMG 900 FOR THE TREATMENT OF CANCER
US2015072988 2015-03-12 USE OF N-(4-((3-(2-AMINO-4-PYRIMIDINYL)-2-PYRIDINYL)OXY)PHENYL)-4-(4-METHYL-2-THIENYL)-1-PHTHALAZINAMINE IN COMBINATION WITH HISTONE DEACETYLASE INHIBITORS FOR TREATMENT OF CANCER
US8921367 2014-12-30 Use of AMG 900 for the treatment of cancer
US2014163052 2014-06-12 FUSED TRICYCLIC DUAL INHIBITORS OF CDK 4/6 AND FLT3
US2014127271 2014-05-08 BLOCK COPOLYMERS FOR STABLE MICELLES
US2014113879 2014-04-24 BLOCK COPOLYMERS FOR STABLE MICELLES
US2014114051 2014-04-24 BLOCK COPOLYMERS FOR STABLE MICELLES
Patent ID Date Patent Title
US2014114051 2014-04-24 BLOCK COPOLYMERS FOR STABLE MICELLES
US2014066430 2014-03-06 AURORA KINASE MODULATORS AND METHOD OF USE
US8623885 2014-01-07 Fused tricyclic dual inhibitors of CDK 4/6 and FLT3
US2012028917 2012-02-02 Use Of N-(4-((3-(2-Amino-4-Pyrimidinyl)-2-Pyridinyl)Oxy)Phenyl)-4-(4-Methyl-2-Thienyl)-1-Phthalazinamine In The Treatment Of Antimitotic Agent Resistant Cancer
US2011263530 2011-10-27 Aurora Kinase Modulators and Method of Use
US8022221 2011-09-20 Aurora kinase modulators and method of use
US7560551 2009-07-14 Aurora kinase modulators and method of use
WO2003055491A1 20 Dec 2002 10 Jul 2003 Astrazeneca Ab Substituted quinazoline derivatives as inhibitors of aurora kinases
WO2004000833A1 19 Jun 2003 31 Dec 2003 Vertex Pharmaceuticals Incorporated Processes for preparing substituted pyrimidines and pyrimidine derivatives as inhibitors of protein kinase
WO2004016612A2 13 Aug 2003 26 Feb 2004 Cyclacel Limited New purine derivatives
WO2004037814A1 27 Oct 2003 6 May 2004 Vertex Pharmaceuticals Incorporated Indazolinone compositions useful as kinase inhibitors
WO2004039774A2 19 May 2003 13 May 2004 Merck & Co., Inc. Mitotic kinesin inhibitors
WO2004092607A1 30 Mar 2004 28 Oct 2004 Carbone Lorraine Composants Ventilated disc brake pads
WO2005113494A2 9 May 2005 1 Dec 2005 Amgen Inc. Nitrogenated heterocyclic derivatives as protein kinase modulators and use for the treatment of angiogenesis and cancer
EP1702919A1 28 Dec 2004 20 Sep 2006 Banyu Pharmaceutical Co., Ltd. Novel 2-heteroaryl-substituted benzimidazole derivative
US6919338 21 Jun 2001 19 Jul 2005 Astrazeneca Ab Substituted quinazoline derivatives and their use as inhibitors of aurora-2 kinase
Citing Patent Filing date Publication date Applicant Title
WO2008124083A3 * 3 Apr 2008 15 Jan 2009 Amgen Inc Aurora kinase modulators and method of use
WO2009117157A1 * 19 Mar 2009 24 Sep 2009 Amgen Inc. Aurora kinase modulators and method of use
WO2010017240A2 * 4 Aug 2009 11 Feb 2010 Amgen Inc. Aurora kinase modulators and methods of use
WO2010017240A3 * 4 Aug 2009 1 Apr 2010 Amgen Inc. Aurora kinase modulators and methods of use
WO2011031842A1 9 Sep 2010 17 Mar 2011 Amgen Inc. N-4 ( – ( ( 3- ( 2 -amino-4 pyrimidinyl) -2 -pyridinyl) oxy) phenyl) -4- (4-methyl-2-thienyl) -1-phthalazinamine for use in the treatment of antimitotic agent resistant cancer
WO2012129344A1 21 Mar 2012 27 Sep 2012 Amgen Inc. Fused tricyclic dual inhibitors of cdk 4/6 and flt3
WO2015084649A1 25 Nov 2014 11 Jun 2015 Amgen Inc. Crystalline forms of n-(4-((3-(2-amino-4-pyrimidinyl) – 2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-1 -phthalazinamine pharmaceutically acceptable salts and uses thereof
EP2818170A1 9 Sep 2010 31 Dec 2014 Amgen, Inc N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridin yl)oxy)phenyl)-4-(4-methyl-2-thienyl)-1-phthalazinamine for use in the treatment of antimitotic agent resistant cancer
EP2937349A1 21 Mar 2012 28 Oct 2015 Amgen Inc. Fused tricyclic dual inhibitors of cdk 4/6 and flt3
US7994185 4 May 2009 9 Aug 2011 Glaxo Smith Kline LLC Benzene sulfonamide thiazole and oxazole compounds
US8362241 29 Jan 2013 Amgen Inc. Inhibitors of PI3 kinase and/or mTOR
US8404694 19 Mar 2009 26 Mar 2013 Amgen Inc. Aurora kinase modulators and method of use
US8415345 4 May 2009 9 Apr 2013 Glaxo SmithKline LLC Benzene sulfonamide thiazole and oxazole compounds
US8586751 10 Jun 2010 19 Nov 2013 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
US8637500 16 Dec 2009 28 Jan 2014 Amgen Inc. Aminopyridine and carboxypyridine compounds as phosphodiesterase 10 inhibitors
US8642759 31 Jan 2013 4 Feb 2014 Glaxosmithkline Llc Benzene sulfonamide thiazole and oxazole compounds
US8772480 19 Nov 2012 8 Jul 2014 Amgen Inc. Inhibitors of PI3 kinase and/or mTOR
US9126935 7 Aug 2009 8 Sep 2015 Amgen Inc. Aurora kinase modulators and methods of use
US9233956 25 Nov 2013 12 Jan 2016 Novartis Ag Benzene sulfonamide thiazole and oxazole compounds

///////////945595-80-2, AMG 900,  aurora kinase (ARK) inhibitor,  treatment of leukemia and solid tumours, AMGEN, 2014, orphan drug designation,  U.S. for the treatment of ovarian cancer

CC1=CSC(=C1)C2=NN=C(C3=CC=CC=C32)NC4=CC=C(C=C4)OC5=C(C=CC=N5)C6=NC(=NC=C6)N

Share

GSK-2881078

 phase 1, Uncategorized  Comments Off on GSK-2881078
Jun 142016
 

GSK-2881078

(R)-1-[1-(Methylsulfonyl)propan-2-yl]-4-(trifluoromethyl)-1H-indole-5-carbonitrile

(R)-1 -(1-(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)indoline-5-carbonitrile

Phase I

A selective androgen receptor modulator potentially for the treatment of cachexia.

Inventors Philip Stewart Turnbull, Rodolfo Cadilla
Applicant Glaxosmithkline Intellectual Property (No.2) Limited
CAS Number 1539314-06-1
Chemical Name GSK-2881078
Synonyms GSK-2881078
Molecular Formula C14H13NF3N2O2S
Formula Weight 330.33
  • Originator GlaxoSmithKline
  • Mechanism of Action Selective androgen receptor modulators
  • Phase I Cachexia

Most Recent Events

  • 03 Sep 2015 GlaxoSmithKline initiates enrolment in a phase I trial for Cachexia (In volunteers) in USA (NCT02567773)
  • 01 Mar 2015 GlaxoSmithKline completes a phase I trial in Cachexia (In volunteers) in USA (NCT02045940)
  • 31 Jan 2014 Phase-I clinical trials in Cachexia (In volunteers) in USA (PO)

GSK2881078 is a selective androgen receptor modulator (SARM) that is being evaluated for effects on muscle growth and strength in subjects with muscle wasting to improve their physical function. Part A of this study will evaluate the safety, efficacy and pharmacokinetics of GSK2881078 in healthy, older men and post-menopausal women who will take daily dosing for 28 days and be followed for a total of 70 days. Part B of this study will characterize the effect of Cytochrome P450 3A4 (CYP3A4) inhibition on the GSK2881078 pharmacokinetics. Part B will only be conducted if safe and efficacious dose is identified in Part A. Part A will include healthy older males and post-menopausal females; and randomize approximately 60 subjects (about 15 per cohort [4 cohorts]) to complete approximately 48 (about 12 per cohort). Part B will enroll one cohort of approximately 15 healthy male subjects to complete approximately 12. The study duration will be approximately 115 days for Part A and 122 days for Part B.

Steroidal nuclear receptor (NR) ligands are known to play important roles in the health of both men and women. Testosterone (T) and dihydrotestosterone (DHT) are endogenous steroidal ligands for the androgen receptor (AR) that appear to play a role in every tissue type found in the mammalian body. During the development of the fetus, androgens play a role in sexual differentiation and development of male sexual organs. Further sexual development is mediated by androgens during puberty. Androgens play diverse roles in the adult, including stimulation and maintenance of male sexual accessory organs and maintenance of the musculoskeletal system. Cognitive function, sexuality, aggression, and mood are some of the behavioral aspects mediated by androgens. Androgens have a physiologic effect on the skin, bone, and skeletal muscle, as well as blood, lipids, and blood cells (Chang, C. and Whipple, G. Androgens and Androgen Receptors. Kluwer Academic Publishers: Boston, MA, 2002)

Many clinical studies with testosterone have demonstrated significant gains in muscle mass and function along with decreases in visceral fat. See, for example,

Bhasin (2003) S. J. Gerontol. A Biol. Sci. Med. Sci. 58:1002-8, and Ferrando, A. A. et al. (2002) Am. J. Phys. Endo. Met. 282: E601-E607. Androgen replacement therapy (ART) in men improves body composition parameters such as muscle mass, strength, and bone mineral density (see, for example, Asthana, S. et al. (2004) J. Ger, Series A: Biol. Sci. Med. Sci. 59: 461 -465). There is also evidence of improvement in less tangible parameters such as libido and mood. Andrologists and other specialists are increasingly using androgens for the treatment of the symptoms of androgen deficiency. ART, using T and its congeners, is available in transdermal, injectable, and oral dosage forms. All current treatment options have contraindications (e.g., prostate cancer) and side-effects, such as increased hematocrit, liver toxicity, and sleep apnoea. Side-effects from androgen therapy in women include: acne, hirsutism, and lowering of high-density lipoprotein (HDL) cholesterol levels, a notable side-effect also seen in men.

Agents that could selectively afford the benefits of androgens and greatly reduce the side-effect profile would be of great therapeutic value. Interestingly, certain NR ligands are known to exert their action in a tissue selective manner (see, for example, Smith et al. (2004) Endoc. Rev. 2545-71 ). This selectivity stems from the particular ability of these ligands to function as agonists in some tissues, while having no effect or even an antagonist effect in other tissues. The term “selective receptor modulator” (SRM) has been given to these molecules. A synthetic compound that binds to an intracellular receptor and mimics the effects of the native hormone is referred to as an agonist. A compound that inhibits the effect of the native hormone is called an antagonist. The term “modulators” refers to compounds that have a spectrum of activities ranging from full agonism to partial agonism to full antagonism.

SARMs (selective androgen receptor modulators) represent an emerging class of small molecule pharmacotherapeutics that have the potential to afford the important benefits of androgen therapy without the undesired side-effects. Many SARMs with demonstrated tissue-selective effects are currently in the early stages of development See, for example, Mohler, M. L. et al. (2009) J. Med. Chem. 52(12): 3597-617. One notable SARM molecule, Ostarine™, has recently completed phase I and II clinical studies. See, for example, Zilbermint, M. F. and Dobs, A. S. (2009) Future Oncology 5(8):121 1-20. Ostarine™ appears to increase total lean body mass and enhance functional performance. Because of their highly-selective anabolic properties and demonstrated androgenic-sparing activities, SARMs should be useful for the prevention and/or treatment of many diseases in both men and women, including, but not limited to sarcopenia, cachexias (including those associated with cancer, heart failure, chronic obstructive pulmonary disease (COPD), and end stage renal disease (ESRD), urinary incontinence, osteoporosis, frailty, dry eye and other conditions associated with aging or androgen deficiency. See, for example, Ho et al. (2004) Curr Opin Obstet Gynecol. 16:405-9; Albaaj et al. (2006) Postgrad Med J 82:693-6; Caminti et al. (2009) J Am Coll Cardiol. 54(10):919-27; lellamo et al. (2010) J Am Coll Cardiol. 56(16): 1310-6; Svartberg (2010) Curr Opin Endocrinol Diabetes Obes. 17(3):257-61 , and Mammadov et al. (201 1 ) Int Urol Nephrol 43:1003-8. SARMS also show promise for use in promoting muscle regeneration and repair (see, for example, Serra et al. (Epub 2012 Apr 12)

doi:10.1093/Gerona/gls083),in the areas of hormonal male contraception and benign prostatic hyperplasia (BPH), and in wound healing (see, for example, Demling (2009) ePIasty 9:e9).

Preclinical studies and emerging clinical data demonstrate the therapeutic potential of SARMs to address the unmet medical needs of many patients. The demonstrated advantages of this class of compounds in comparison with steroidal androgens (e.g. , tissue-selective activity, oral administration, AR selectivity, and lack of androgenic effect) position SARMs for a bright future of therapeutic applications.

Although amorphous forms of SARMs may be developed for some uses, compounds having high crystallinity are generally preferred for pharmaceutical use due to their improved solubility and stability. Accordingly, there remains a need in the art for crystalline form of SARMs for therapeutic use.

Patent

WO 2015110958

EXAMPLES

Example 1 – Synthesis of (R)-1 -(1 -(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)- -indole-5-carbonitrile

(R)-1 -(1-(methylsulfonyl)propan-2-yl)^-(trifluoromethyl)-1 H-indole-5-carbonitrile

Method 1 :

A. (R)-1 -(Methylthio)propan-2 -amine

Step 1

To a solution of commercially available (R)-2-aminopropan-1 -ol (5 g, 66.6 mmol) in MeCN (20 mL), in an ice bath, is added very slowly, dropwise, chlorosulfonic acid (4.46 mL, 66.6 mmol) (very exothermic). The reaction mixture is kept in the cold bath for ~10 min, and then at rt for ~ 30 min. After stirring for another ~ 10 minutes, the solids are collected by filtration, washed sequentially with MeCN (40 mL) and hexanes (100 mL), and dried by air suction for ~ 40 min. to produce the intermediate ((R)-2-aminopropyl hydrogen sulfate.

Step 2:

To a solution of sodium thiomethoxide (5.60 g, 80 mmol) in water (20 mL) is added solid NaOH (2.66 g, 66.6 mmol) in portions over ~ 10 min. Then the intermediate from step 1 is added as a solid over ~ 5 min. The mixture is then heated at 90 °C for ~10 h. The reaction mixture is biphasic. Upon cooling, MTBE (20 mL) is added, and the organic phase (brownish color) is separated. The aqueous phase is extracted with MTBE (2 x 20 mL). The original organic phase is washed with 1 N NaOH (15 mL). The basic aqueous phase is re-extracted with MTBE (2 x 20 mL). All the ether phases are combined, dried over Na2S04, filtered, and concentrated (carefully, since the product is volatile) to afford the crude product as a light yellow oil.

Method 2

(R)-1-(methylthio)propan-2 -amine hydrochloride

A. (R)-2-((tert-Butoxycarbonyl)amino)propyl methanesulfonate

Step 1

Commercially available (R)-2-aminopropan-1 -ol (135 g, 1797 mmol) is dissolved in MeOH 1350 mL). The solution is cooled to 5°C with an icebath, then Boc20 (392 g, 1797 mmol) is added as a solution in MeOH (1000 mL). The reaction temperature is kept below 10°C. After the addition, the cooling bath is removed, and the mixture is stirred for 3 h. The MeOH is removed under vacuum (rotavap bath: 50°C). This material is used as is for the next step.

Step 2

The residue is dissolved in CH2CI2 (1200 mL) and NEt3 (378 mL, 2717 mmol) is added, then the mixture is cooled on an ice bath. Next, MsCI (166.5 mL, 2152 mmol) is added over ~2 h, while keeping the reaction temperature below 15°C. The mixture is stirred in an icebath for 1 h then the bath was removed. The mixture is stirred for 3 d, then washed with a 10% NaOH solution (500 mL 3 x), then with water. The organic phase is dried with MgS04, filtered, then stripped off (rota, 50°C waterbath. The impure residue is dissolved in a mix of 500mL EtOAc (500 mL) and MTBE (500 mL) and then extracted with water to remove all water-soluble salts. The organic phase is dried with MgS04, filtered, then stripped off to afford a white solid residue.

B. (R)-tert-Butyl (1 -(methylthio)propan-2-yl)carbamate

NaSMe (30 g, 428 mmol) is stirred with DMF (200 mL) to afford a suspension. Next, (R)-2-((tertbutoxycarbonyl)amino)propyl methanesulfonate (97 g, 383 mmol) is added portionwise while the temperature is kept below 45°C (exothermic). After the addition, the mixture is stirred for 2 h, then toluene (100 mL) is added. The mixture is washed with water (500 mL, 4 x), then dried with MgS04, and filtered. The filtrate is stripped off (rotavap) to a pale yellow oil.

C. (R)-1 -(Methylthio)propan-2 -amine hydrochloride

Acetyl chloride (150 mL,) is added to a stirred solution of MeOH (600 mL) cooled with an icebath. The mixture is stirred for 30 min in an icebath, then added to (R)-tert-butyl (1 -(methylthio)propan-2-yl)carbamate (78 g, 380 mmol). The mixture is stirred at rt for 2 h, (C02, (CH3)2C=CI-l2 evolution) and then stripped off to a white solid.

D. 4-Fluoro-3-iodo-2-(trifluoromethyl)benzonitrile

To a freshly prepared solution of LDA (1 19 mmol) in anhyd THF (250 mL) at -45°C is added a solution of commercially available 4-fluoro-2-(trifluoromethyl)benzonitrile (21 .5 g, 1 14 mmol) in THF (30 mL), dropwise at a rate such that the internal temperature remained < -40°C (became dark brown during addition). The mixture is stirred 30 min at -45°C, cooled to -70°C and iodine (31 .7 g, 125 mmol) is added in one portion (-70°C→ -52°C). The mixture is stirred for 1 h, removed from the cooling bath and quenched by addition of 10% Na2S203 (ca. 250 mL) and 1 N HCI (ca. 125 mL). The mixture is extracted with EtOAc (x3). Combined organics are washed (water, brine), dried over Na2S04 and concentrated in vacuo. The residue is purified by low pressure liquid chromatography (silica gel, EtOAc / hexanes, gradient elution) followed by

recrystallization from heptane (30 mL), twice, affording 4-fluoro-3-iodo-2-(trifluoromethyl)benzonitrile (15.79 g, 50.1 mmol, 44.1 % yield) as a pale yellow solid.

E. 4-Fluoro-2-(trifluoromethyl)-3-((trimethylsilyl)ethynyl)benzonitrile

A 20 mL vial is charged with 4-fluoro-3-iodo-2-(trifluoromethyl)benzonitrile,(0.315 g, 1 .00 mmol), Pd(PPh3)2CI2 (0.014 g, 0.020 mmol) and Cul (0.0076 g, 0.040 mmol), and sealed with a rubber septum. Anhyd PhMe (5 mL) and DIPA (0.210 mL, 1 .500 mmol) are added via syringe and the mixture is degassed 10 min by sparging with N2while immersed in an ultrasonic bath. Ethynyltrimethylsilane (0.155 mL, 1 .100 mmol) is added dropwise via syringe and the septum is replaced by a PTFE-faced crimp top. The mixture is stirred in a heating block at 60°C. Upon cooling the mixture is diluted with EtOAc and filtered through Celite. The filtrate is washed (satd NH4CI, water, brine), dried over Na2S04 and concentrated in vacuo. The residue is purified by low pressure liquid chromatography (silica gel, EtOAc / hexanes, gradient elution) affording 4-fluoro-2-(trifluoromethyl)-3-((trimethylsilyl)ethynyl)benzonitrile .

F. (R)-1 -(1 -(methylthio)propan-2-yl)-4-(trifluoromethyl)-1 H-indole-5-carbonitrile

A mixture of 4-fluoro-2-(trifluoromethyl)-3-((trimethylsilyl)ethynyl)benzonitrile (1 .16 g, 4.07 mmol), (R)-1 -(methylthio)propan-2-amine (0.599 g, 5.69 mmol) and DIEA (1 .42 mL, 8.13 mmol) in DMSO (7 mL) is heated (sealed tube) at 100°C for 50 min. Upon cooling, the reaction mixture is diluted with EtOAc (50 mL) and washed with water (30 mL). The organic phase is washed with water and brine, dried over Na2S04, filtered and concentrated to give the intermediate aniline. This intermediate is dissolved in NMP (7 mL), treated with KOtBu (1 M in THF) (5.69 mL, 5.60 mmol) and heated at 50°C. The reaction is monitored by LCMS, and deemed complete after 40 min. Upon cooling, the reaction mixture is diluted with EtOAc (40 mL) and washed with water (30 mL). The organic phase is washed with more water and brine, dried over Na2S04, filtered and concentrated. The residue is chromatographed over silica gel using a 5-40% EtOAc-hexane gradient to give the thioether intermediate:

G. (R)-1 -(1-(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)-1 H-indole-5-carbonitrile

To an ice-cold solution of (R)-1 -(1 -(methylthio)propan-2-yl)-4-(trifluoromethyl)-1 H-indole-5-carbonitrile (0.560 g, 1.88 mmol) in MeOH (10 mL) is added a solution of Oxone (4.04 g, 6.57 mmol) in water (10 mL). After 50 min, the reaction mixture is diluted with water (30 mL) and extracted with EtOAc (50 mL). The organic phase is washed with brine, dried over Na2S04, filtered and concentrated. The residue is chromatographed over silica gel using 100% CH2CI2 to give (R)-1-(1 -(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)-l H-indole-5-carbonitrile as a white foam that is crystallized from

CH2CI2/hexanes to afford a white solid.

Example 2- Preparation of crystalline form 1 of (R)-1 -(1-(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)indoline-5-carbonitrile

(R)-1 -(1-(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)indoline-5-carbonitrile (1 .74kg, 1wt) was dissolved in ethyl acetate (12.0 Kg, 6.9 wt) at 20-30°C. The solution was transferred into a clean reaction vessel via an in-line cartridge filter. The solution was concentrated to ~3.0-5.0 volumes under reduced pressure, keeping the temperature below 50°C. The solution was cooled to 20-30°C, and n-heptane (23.0 Kg, 13.2 wt) was added slowly over ~1 hour. The solution was stirred 1 -2 hrs at 20-30°C, heated to 50-55°C for 2-3 hours, cooled back to 20-30°C and stirred for 1 -2 hours. The slurry was sampled and analyzed by XRPD. The solid was collected by filtration, washed with n-heptane (1 .4 Kg, 0.8 wt), and dried in vacuo at 40-50 °C to provide crystalline

(R)-1 -(1-(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)indoline-5-carbonitrile (1 .54 Kg, Form 1 ; 88.5 % yield, 99.5% purity) as a slightly colored solid.

Example 3- Preparation of crystalline form 2 of (R)-1 -(1-(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)indoline-5-carbonitrile

Crude (R)-1 -(1 -(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)indoline-5-carbonitrile (1 .54 g [theoretical], 1 wt) was dissolved in dichloromethane (5mL, 3.25 vol) and loaded onto a 12-g ISCO column (Si02). The column was eluted with DCM (-500 mL, 325 vol) and the product-containing fractions were combined and concentrated in vacuo. The resulting residue was triturated in n-heptane. The solid was collected by filtration, air-dried, and placed under high vacuum for 3 h to provide GSK2881078A (1 .009 g, Form 2; 65.1 % yield, 100% AUC HPLC-UV) as a white solid.

 

PATENT

https://www.google.com/patents/WO2014013309A1?cl=en22

Example 26

1-(1-(Methylsulfonyl)propan-2-yl)-4-(trifiuoromethyl)-1H-indole-5-carbonitrile Synthesized in a manner similar to Example 9 using 1-(1-(methylthio)propan-2-yl)-4-(trifluoromethyl)-1 H-indole-5-carbonitrile (Example 25): MS (ESI): m/z 331 (MH+).

Example 27

(R)-1 -(1 -(Methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)-1 H-indole-5-carbonitrile

A. (R)-1-(Methylthio)propan-2-amine

Step l

To a solution of commercially available (R)-2-aminopropan-1-ol (5 g, 66.6 mmol) in MeCN (20 mL), in an ice bath, was added very slowly, dropwise, chlorosulfonic acid (4.46 mL, 66.6 mmol) (very exothermic). A gummy beige precipitate formed. The reaction mixture was kept in the cold bath for -10 min, and then at rt for ~ 30 min. The reaction mixture was scratched with a spatula to try to solidify the gummy precipitate. After a few minutes, a beige solid formed. After stirring for another ~ 10 minutes, the solids were collected by filtration, washed sequentially with MeCN (40 mL) and hexanes (100 mL), and dried by air suction for ~ 40 min. The intermediate ((R)-2-aminopropyl hydrogen sulfate, weighed 0.46 g (~ 96% yield).

Step 2:

To a solution of sodium thiomethoxide (5.60 g, 80 mmol) in water (20 mL) was added solid NaOH (2.66 g, 66.6 mmol) in portions over – 10 min. Then the intermediate from step 1 was added as a solid over ~ 5 min. The mixture was then heated at 90 °C for -10 h. The reaction mixture was biphasic. Upon cooling, MTBE (20 mL) was added, and the organic phase (brownish color) was separated. The aqueous phase was extracted with MTBE (2 x 20 mL). The original organic phase is washed with 1 NaOH (15 mL) (this removes most of the color). The basic aqueous phase was re-extracted with MTBE (2 x 20 mL). All the ether phases are combined, dried over Na2S04, filtered, and

concentrated (carefully, since the product is volatile) to afford the crude product as a light yellow oil: 1H NMR (400 MHz, DMSO-cf6) δ 2.91-2.87 (m, 1 H), 2.43-2.31 (m, 2 H), 2.04 (s, 3 H), 1.50 (bs, 2 H), 1.01 (d, J = 6.3 Hz, 3 H).

Alternative synthesis of example 27A:

(R)-1 -(Methylthio)propan-2 -amine hydrochloride

A. (R)-2-((tert-Butoxycarbonyl)amino)propyl methanesulfonate

Step 1

Commercially available (R)-2-aminopropan-1-ol (135 g, 1797 mmol) was dissolved in MeOH 1350 mL). The solution was cooled to 5°C with an icebath, then Boc20 (392 g, 1797 mmol) was added as a solution in MeOH (1000 mL). The reaction temperature was kept below 10°C. After the addition, the cooling bath was removed, and the mixture was stirred for 3 h. The MeOH was removed under vacuum (rotavap bath: 50°C). The resulting residue was a colorless oil that solidified overnight to a white solid. This material was used as is for the next step.

Step 2

The residue was dissolved in CH2CI2 (1200 mL) and NEt3 (378 mL, 2717 mmol) was added, then the mixture was cooled on an ice bath. Next, MsCI (166.5 mL, 2152 mmol) was added over ~2 h, while keeping the reaction temperature below 15°C. The mixture was stirred in an icebath for 1 h then the bath was removed. The mixture was stirred for 3 d, then washed with a 10% NaOH solution (500 mL 3 x), then with water. The organic phase was dried with MgS0 , filtered, then stripped off (rota, 50°C waterbath. The impure residue was dissolved in a mix of 500mL EtOAc (500 mL) and MTBE (500 mL) and then, extracted with water to remove all water-soluble salts.The organic phase was dried with MgS04, filtered, then stripped off to afford a white solid residue: 1H NMR (400 MHz, DMSO-ds) δ 6.94-6.92 (m, 1 H), 4.02 (d, J = 5.8 Hz, 2 H), 3.78-3.71 (m, 1 H), 3.16 (s, 3 H), 1.38 (s, 9 H), 1.06 (d, J = 6.8 Hz, 3 H).

B. (R)-tert-Butyl (1-(methylthio)propan-2-yl)carbamate

NaSMe (30 g, 428 mmol) was stirred with DMF (200 mL) to afford a suspension. Next, (R)-2-((tertbutoxycarbonyl)amino)propyl methanesulfonate (97 g, 383 mmol) was added

portionwise while the temperature was kept below 45°C (exothermic).. After the addition, the mixture was stirred for 2 h, then toluene (100 ml_) was added. The mixture was washed with water (500 ml_, 4 x), then dried with MgS04, and filtered. The filtrate was stripped off (rotavap) to a pale yellow oil: 1H NMR (400 MHz, DMSO-d6) δ 6.77-6.75 (m, 1 H), 3.60-3.54 (m, 1 H), 2.54-2.50 (m, 1 H), 2.43-2.38 (m, 1 H), 2.05 (s, 3 H), 1.38 (s, 9 H), 1.08 (d, J = 7.8 Hz, 3 H).

C. (R)-1-(Methylthio)propan-2-amine hydrochloride

Acetyl chloride (150 mL,) was added to a stirred solution of MeOH (600 mL) cooled with an icebath. The mixture was stirred for 30 min in an icebath, then added to (R)-tert-butyl (1-(methylthio)propan-2-yl)carbamate (78 g, 380 mmol). The mixture was stirred at rt for 2 h, (C02, (CH3)2C=CH2 evolution) and then stripped off to a white solid: 1H NMR (400 MHz, DMSO-d6) δ 8.22 (bs, 3 H), 3.36-3.29 (m, 1 H), 2.80-2.75 (m, 1 H), 2.64-2.59 (m, 1 H (d, J = 6.6 Hz, 3 H).

D. (R)-1 -(1 -(Methylthio)propan-2-yl)-4-(trif luoromethy l)-1 H-indole-5-carbonitrile

A mixture of 4-fluoro-2-(trifluoromethyl)-3-((trimethylsilyl)ethynyl)benzonitrile (Example 21 D,1.16 g, 4.07 mmol), (R)-1-(methylthio)propan-2-amine (0.599 g, 5.69 mmol) and DIEA (1.42 mL, 8.13 mmol) in DMSO (7 mL) was heated (sealed tube) at 100°C for 50 min. Upon cooling, the reaction mixture was diluted with EtOAc (50 mL) and washed with water (30 mL). The organic phase was washed with water and brine, dried over Na2S04, filtered and concentrated to give the intermediate aniline. This intermediate was dissolved in NMP (7 mL), treated with KOtBu (1 M in THF) (5.69 mL, 5.60 mmol) and heated at 50°C. The reaction was monitored by LCMS, and deemed complete after 40 min. Upon cooling, the reaction mixture was diluted with EtOAc (40 mL) and washed with water (30 mL). The organic phase was washed with more water and brine, dried over Na2S04, filtered and concentrated. The residue was chromatographed over silica

gel using a 5-40% EtOAc-hexane gradient to give the thioether intermediate: MS (ESI):

E. (R)-1-(1-(Methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)-1H-indole-5-carbonitrile

To an ice-cold solution of (R)-1-(1-(methylthio)propan-2-yl)-4-(trifluoromethyl)-1 H-indole-5-carbonitrile (0.560 g, 1.88 mmol) in MeOH (10 mL) was added a solution of Oxone (4.04 g, 6.57 mmol) in water (10 mL). After 50 min, the reaction mixture was diluted with water (30 mL) and extracted with EtOAc (50 mL). The organic phase was washed with brine, dried over Na2S04, filtered and concentrated. The residue was chromatographed over silica gel using 100% CH2CI2 to give (R)-1-(1-(methylsulfonyl)propan-2-yl)-4-(trifluoromethyl)-l H-indole-5-carbonitrile as a white foam that was crystallized from CH2CI2/hexanes to afford a white solid (0.508 g, 79% yield): 1H NMR (400 MHz, DMSO-d6) δ 8.17 (d, J = 8.6 Hz, 1 H), 8.12 (d, J = 3.5 Hz, 1 H), 7.81 (d, J – 8.5 Hz, 1 H), 6.87-6.84 (m, 1 H), 5.43-5.35 (m, 1 H), 4.01 (dd, J = 14.8, 8.6 Hz, 1 H), 3.83 (dd, J = 14.8, 4.9 Hz, 1 H), 2.77 (s, 3 H), 1.59 (d, J = 6.8 Hz, 3 H); MS (ESI): m/z 331 (M+H).

 

Philip Turnbull

Philip Turnbull

Director of Chemistry

https://www.linkedin.com/in/philip-turnbull-21266a8

Experience

Director of Chemistry

Receptos, a wholly-owned subsidiary of Celgene

– Present (1 year 1 month)Greater San Diego Area

Director

GSK

(5 years 3 months)RTP

Section Head

GSK

(3 years 1 month)RTP

Group Manager

GlaxoSmithKline

(4 years 1 month)RTP

Investigator

GSK

(4 years 11 months)RTP

Research Associate

Biophysica Foundation

(3 years 8 months)La Jolla, Ca

Education

University of California, Irvine

Doctor of Philosophy (Ph.D.), Organic synthesis

////////GSK-2881078,  1539314-06-1, Phase 1, clinical trials,  Cachexia , GlaxoSmithKline

Share

GSK-2879552

 phase 1, Uncategorized  Comments Off on GSK-2879552
Jun 142016
 

GSK-2879552

CAS 1401966-69-5 (ABS),  1401966-63-9(REL)

C23 H28 N2 O2, 364.48

Benzoic acid, 4-​[[4-​[[[(1R,​2S)​-​2-​phenylcyclopropyl]​amino]​methyl]​-​1-​piperidinyl]​methyl]​-

4-((4-((((lR,2S)-2-phenylcyclopropyl)amino)methyl)piperidin-l-yl)methyl)benzoic acid

  • 4-[[4-[[[(1R,2S)-2-Phenylcyclopropyl]amino]methyl]-1-piperidinyl]methyl]benzoic acid
  • 4-[[4-[[((1R,2S)-2-Phenylcyclopropyl)amino]methyl]piperidin-1-yl]methyl]benzoic acid

4-((4-((((1R,2S)-2-phenylcyclopropyl)amino)methyl)piperidin-1-yl)methyl)benzoic acid

Phase I

Glaxosmithkline Llc  INNOVATOR

Neil W. Johnson, Jiri Kasparec, William Henry Miller, Meagan B. Rouse, Dominic Suarez, Xinrong Tian,

A LSD1 inhibitor potentially for the treatment of small cell lung cancer and acute myeloid leukemia.

GSK2879552 is an orally available, irreversible, inhibitor of lysine specific demethylase 1 (LSD1), with potential antineoplastic activity. Upon administration, GSK2879552 binds to and inhibits LSD1, a demethylase that suppresses the expression of target genes by converting the dimethylated form of lysine at position 4 of histone H3 (H3K4) to mono- and unmethylated H3K4. LSD1 inhibition enhances H3K4 methylation and increases the expression of tumor-suppressor genes. This may lead to an inhibition of cell growth in LSD1-overexpressing tumor cells. LSD1, overexpressed in certain tumor cells, plays a key role in tumor cell growth and survival. Check for active clinical trials or closed clinical trials using this agent.

GSK-2879552 chemical structure

Formula: C23H29ClN2O2
M.Wt: 400.94

GSK2879552, LSD1 Inhibitor

CAS 1902123-72-1

Molecular Weight: 437.41
Formula: C23H28N2O2.2HCl

Chromatin modification plays an essential role in transcriptional regulation (T. Kouzarides, 2007, Cell 128: 693-705). These modifications, which include DNA methylation, histone acetylation and hsitone methylation, are disregulated in tumors. This epigenetic disregulation plays an important role in the silencing of tumor suppressors and overexpression of oncogenes in cancer (M. Esteller, 2008, N Engl J Med 358: 1148-59. P. Chi et al, 2010, Nat Rev Cane 10:457-469.). The enzymes that regulate histone methylation are the histone methyl transferases and the histone demethylases.

Lysine-specific demethylase 1 (LSDl; also known as BHC110) is a histone lysine demethylase reported to demethylate H3K4mel/2 (Y. Shi et al, 2004, Cell 119: 941-953) and H3K9mel/2 (R. Schule et al.,2005, Nature 437: 436-439). LSDl is overexpressed in multiple human cancers, including prostate where it is associated with more frequent relapse (P. Kahl et al, 2006, Cane. Res. 66: 11341-11347), breast (J. Kirfel et al, 2010, Carcinogenesis 31: 512-520) neuroblastoma (J. Kirfel et al, 2009, Cane. Res. 69: 2065-2071. G. Sun et al, 2010, Mol. Cell. Biol. 28: 1997-2000). LSDl is essential for transcriptional regulation mediated by a number of nuclear hormone receptors, including androgen receptor in prostate cancer (R. Schuele et al, 2005, Nature 437: 436-439. R. Schuele et al, 2007, Nat. Cell Biol. 9: 347-353. R. Schuele et al, 2010, Nature 464: 792-796), estrogen receptor in breast carcinomas (M.G. Rosenfeld et al, 2007, Cell 128: 505-518), and TLX receptor in neuorblastoma (S. Kato et al, 2008, Mol. Cell. Biol. 28: 3995-4003). These studies have shown that knockdown of LSDl expression results in decreased cancer cell proliferation. Additionally, LSDl is overexpressed in multiple cancer types that are nuclear hormone receptor-independent. Those tumors include ER-negative breast (J. Kirfel et al, 2010, Carcinogenesis 31: 512-520), small-cell lung, bladder, head & neck, colon, serous ovary, and kidney Wilm’s tumor. Therefore, potent selective small molecule inhibitors of LSDl may be useful for treatment of cancers that are nuclear hormone receptor-dependent and/or nuclear hormone receptor-independent.

The compositions and methods provided herein can potentially be useful for the treatment of cancer including tumors such as skin, breast, brain, cervical carcinomas, testicular carcinomas, etc. More particularly, cancers that may be treated by the compositions and methods of the invention include, but are not limited to tumor types such as astrocytic, breast, cervical, colorectal, endometrial, esophageal, gastric, head and neck, hepatocellular, laryngeal, lung, oral, ovarian, prostate and thyroid carcinomas and sarcomas. More specifically, these compounds can potentially be used to treat: Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Kaposi’s sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma); Genitourinary tract: kidney (adenocarcinoma, Wilm’s tumor

(nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma,angiosarcoma, hepatocellular adenoma, hemangioma; Bone: osteogenic sarcoma(osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing’s sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, meduUoblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre -tumor cervical dysplasia), ovaries (ovarian carcinoma (serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes

(carcinoma); Hematologic: blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplasia syndrome), Hodgkin’s disease, non-Hodgkin’s lymphoma (malignant lymphoma); Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi’s sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and Adrenal glands: neuroblastoma. Thus, the term “cancerous cell” as provided herein, includes a cell afflicted by any one of or related to the above identified conditions.

SYNTHESIS

GSK-2879552

 

STR1

PATENT

WO 2012135113

https://www.google.co.in/patents/WO2012135113A2?cl=en

 

Example 2

1 , 1 -Dimethylethyl 4-( { \( 1 R,2S)-2-phenylcyclopropyl] amino I methyl)- 1 -piperidinecarboxylate

Following a procedure analogous to the procedure described in Example 1 using [(1R,2S)-2-phenylcyclopropyl]amine ((-) isomer) (94 mg, 0.703 mmol) afforded 1,1 -dimethylethyl 4-({[(lR,2S)-2-phenylcyclopropyl]amino}methyl)-l-piperidinecarboxylate (92 mg, 0.264 mmol, 56.4 % yield) as white solid. 1H NMR (400 MHz, METHANOL-d4) δ 7.29 – 7.37 (m, 2H), 7.23 – 7.28 (m, 1H), 7.17 – 7.22 (m, 2H), 4.14 (d, J= 12.63 Hz, 2H), 3.14 (d, J = 7.07 Hz, 2H), 3.01 (dt, J= 4.14, 7.64 Hz, 1H), 2.81 (br. s., 2H), 2.53 (ddd, J= 3.54, 6.63, 10.29 Hz, 1H), 1.97 (ddd, 1H), 1.80 (d, J= 12.13 Hz, 2H), 1.55 (ddd, J= 4.29, 6.63, 10.55 Hz, 1H), 1.47 (s, 9H), 1.36 – 1.45 (m, 1H), 1.23 (qd, J= 4.29, 12.38 Hz, 2H); LC-MS Rt = 0.78 min; MS (ESI): 331.3 [M+H]+.

Example 6

[(lR,2S)-2-Phenylcyclopropyll(4-piperidinylmethyl)amine

Following a procedure analogous to the procedure described in Example 4 using 1,1-dimethylethyl 4-({[(lR,2S)-2-phenylcyclopropyl]amino}methyl)-l-piperidinecarboxylate (Example 2, 60 mg, 0.182 mmol) afforded [(lR,2S)-2-phenylcyclopropyl](4-piperidinylmethyl)amine (41 mg, 0.146 mmol, 80 % yield)as white solid. 1H NMR (400 MHz, METHANOLS) δ 7.29 – 7.38 (m, 2H), 7.23 – 7.29 (m, 1H), 7.18 – 7.23 (m, 2H), 3.47 (d, J= 13.39 Hz, 2H), 3.21 (d, 2H), 2.89 – 3.13 (m, 3H), 2.60 (ddd, J= 3.79, 6.57, 10.36 Hz, 1H), 2.13 – 2.28 (m, J= 3.85, 3.85, 7.61, 11.21 Hz, 1H), 1.99 – 2.13 (m, 2H), 1.49 – 1.71 (m, 3H), 1.35 – 1.48 (m, 1H); LC-MS Rt = 0.44 min; MS (ESI): 231.2

Example 26

4-((4-(((trans-2-phenylcyclopropyl)amino)methyl)piperidin- 1 -yl)methyl)benzoic acid

To the solution of 2,2,2-trifluoro-N-(trans-2-phenylcyclopropyl)-N-(piperidin-4-ylmethyl)acetamide (200 mg, 0.613 mmol, Example l ib) and 4-(bromomethyl)benzoic acid (198 mg, 0.919 mmol) in acetonitrile (6 mL) was added potasium carbonate (254 mg, 1.838 mmol). The reaction mixture was stirred for 3 hours at the 90 °C. The reaction mixture was then filtered and evaporated. The crude oil was mixed with 10 mL of 10 % acetic acid and 10 mL of ethyl acetate. Layers were separated, and the organic layer was discharged. Aqueous layer was neutralized with 1 M Na2C03, and the product was extracted into 10 mL of ethyl acetate. The organic layer was washed with brine, dried over MgS04, filtered and evaporated. The oil was dissolved in 6 ml of EtOH and 3 ml of 1 M NaOH. The reaction mixture was stirred for 20 min, and then it was concentrated. The solution was then partioned between 2 ml of water and 5 mL of ethyl acetate. The organic layer was separated and evaporated. The oil was purified on preparatory HPLC (2 to 10 % AcCN: H20 with 0.1 % formic acid modifier). The fractions were collected. To each

fraction was added 1 ml of 1 M HCl, and the fractions were evaporated to dryness. 4-((4-(((trans-2-phenylcyclopropyl)amino)methyl)piperidin-l-yl)methyl)benzoic acid (50 mg, 0.118 mmol, 19.33 % yield) was isolated as a white solid. 1H NMR (400 MHz,

METHANOLS) δ 8.16 (d, J= 8.34 Hz, 2H), 7.70 (d, J= 8.34 Hz, 2H), 7.30 – 7.37 (m, 2H), 7.23 – 7.29 (m, 1H), 7.20 (d, J= 7.33 Hz, 2H), 4.44 (br. s., 2H), 3.57 (d, J= 11.62 Hz, 2H), 3.07 – 3.27 (m, 4H), 3.04 (dt, J= 3.95, 7.52 Hz, 1H), 2.59 (ddd, J= 3.54, 6.57, 10.11 Hz, lH), 2.12 (d, J= 13.89 Hz, 3H), 1.54 – 1.81 (m, 3H), 1.42 (q, 1H); LC-MS Rt = 0.47 min; MS (ESI): 365.3 [M+H]+.

[M+H]+.

Example 29

4-((4-((((lR,2S)-2-phenylcyclopropyl)amino)methyl)piperidin-l-yl)methyl)benzoic acid

Step 1.

tert-Butyl 4-((4-(hydroxymethyl)piperidin-l-yl)methyl)benzoate

tert-Butyl 4-(bromomethyl)benzoate (1 g, 3.13 mmol) and piperidin-4-ylmethanol (0.361 g, 3.13 mmol) were dissolved in acetonitrile (25 mL). K2CO3 (1.300 g, 9.40 mmol) was added and the reaction mixture was heated to reflux for 20 min. The reaction mixture was cooled down to room temperature, filtered and evaporated. The resulting solid was partitioned between ethyl acetate (50mL) and 1 M HC1 (50 mL). The layers were separated and the aqueous layer was washed with ethyl acetate and the organic layers were discarded. The aqueous layer was basified with 8 M NaOH to pH -10 and extracted 2 times with 50 mL of ethyl acetate. The organic layers were combined, washed with brine and dried over MgSC^, filtered and evaporated. tert-Butyl 4-((4- (hydroxymethyl)piperidin-l-yl)methyl)benzoate (0.95 g, 2.99 mmol, 95 % yield) was isolated as yellow oil. 1H NMR (400 MHz, CHLOROFORM-d) δ 7.95 (d, J= 8.34 Hz, 2H), 7.39 (d, J = 8.08 Hz, 2H), 3.56 (s, 2H), 3.51 (d, J = 6.57 Hz, 2H), 2.90 (d, J= 11.37 Hz, 2H), 1.94 – 2.04 (m, 2H), 1.73 (d, J= 14.15 Hz, 2H), 1.61 (s, 9H), 1.40 – 1.56 (m, 2H), 1.30 – 1.37 (m, 2H); LC-MS Rt = 0.67 min; MS (ESI): 306.2 [M+H]+.

Step 2.

tert-Butyl 4-((4-formylpiperidin- 1 -yl)methyl)benzoate

To a solution of oxalyl chloride (0.408 mL, 4.67 mmol) in dichloromethane (5 mL) at -60 °C was added a solution of DMSO (0.508 mL, 7.15 mmol) in 15 mL of dichloromethane over 30 minutes. The reaction was stirred for 30 minutes at -60 °C A solution of tert-butyl 4-((4-(hydroxymethyl)piperidin-l-yl)methyl)benzoate (950 mg, 3.11 mmol) in 5 mL of dichloromethane was added over 10 minutes at -60 °C. The reaction mixture was stirred for 3 hours at – 60 °C, then triethylamine (2.168 mL, 15.55 mmol) was added and after 10 minutes 10 mL of water was added. The reaction mixture was allowed to warm up to the room temperature. The layers were separated. The pH of the water layer was adjusted to ~7 with 1 M HC1 and then extracted with 20 mL of dichloromethane. The combined organic layers were washed with water and brine, then dried over MgSO, filtered and evaporated. The resulting oil was purified on a silica column eluting with EtOAc to yield tert-butyl 4-((4-formylpiperidin-l-yl)methyl)benzoate (550 mg, 1.722 mmol, 55.4 % yield) as a yellow oil. 1H NMR (400 MHz, CHLOROFORM-d) δ 9.67 (d, J= 1.26 Hz, 1H), 7.96 (d, J= 8.34 Hz, 2H), 7.38 (d, J= 8.34 Hz, 2H), 3.56 (s, 2H), 2.75 – 2.92 (m, 2H), 2.21 – 2.35 (m, 1H), 2.14 (t, J= 10.48 Hz, 2H), 1.91 (dd, J= 2.78, 13.14 Hz, 2H), 1.65 – 1.81 (m, 2H), 1.58 – 1.64 (m, 9H); LC-MS Rt = 0.69 min; MS (ESI): 304.2

[M+H]+, 322.2 [M+H20]+, 336.6 [M+Na]+

Step 3.

tert-Butyl 4-((4-(((( 1 R,2S)-2-phenylcyclopropyl)amino)methyl)piperidin- 1 -yl)methyl)benzoate

To a solution of tert-butyl 4-((4-formylpiperidin-l-yl)methyl)benzoate (6.7 g, 22.08 mmol) in methanol (50 mL) was added (lR,2S)-2-phenylcyclopropanamine (3.53 g, 26.5 mmol). The reaction mixture was refluxed for 5 minutes then cooled down to the room temperature. Sodium cyanotrihydroborate (2.082 g, 33.1 mmol) was added. The reaction mixture was stirred 1 hour at room temperature. Water (50 mL) was added. The reaction was concentrated and 50 mL of dichloromethane was added. The layers were separated. The organics were washed with 10 % acetic acid (50 mL). The layers were separated and 50 mL of brine was added slowly as a solid crashed out. The solid was filtered and suspended in isopropanol. The suspension was sonicated and filtered. tert-Butyl 4-((4-(((( 1 R,2S)-2-phenylcyclopropyl)amino)methyl)piperidin- 1 -yl)methyl)benzoate (5.8 g, 13.65 mmol, 61.8 % yield) was isolated as a white solid. 1H NMR (400 MHz,

METHANOLS) δ 8.07 (d, J= 8.34 Hz, 2H), 7.70 (d, J= 8.08 Hz, 2H), 7.28 – 7.37 (m, 2H), 7.10 – 7.28 (m, 3H), 4.43 (br. s., 2H), 3.54 (d, J= 10.86 Hz, 2H), 3.08 – 3.26 (m, 4H), 3.03 (dt, J= 3.76, 7.39 Hz, 1H), 2.54 – 2.71 (m, 1H), 2.03 – 2.29 (m, 3H), 1.67 – 1.84 (m, 2H), 1.58 – 1.67 (m, 10H), 1.40 (q, J = 6.82 Hz, lH); LC-MS Rt = 0.76 min; MS (ESI): 421.4 [M+H]+.

Step 4.

4-((4-((((lR,2S)-2-phenylcyclopropyl)amino)methyl)piperidin-l-yl)methyl)benzoic acid

A suspension of tert-butyl 4-((4-((((lR,2S)-2-phenylcyclopropyl)amino)methyl)piperidin-l-yl)methyl)benzoate (5.8 g, 13.79 mmol) in HCL – 1 M (80 ml, 80 mmol) was heated to 89 °C (internal temperature) for 2 hr. The solution was cooled down to the room temperature and held in an ice -bath for 1 hour and then filtered. 4-((4-((((lR,2S)-2-phenylcyclopropyl)amino)methyl)piperidin-l-yl)methyl)benzoic acid (3.8 g, 8.25 mmol, 59.8 % yield) was isolated as white solid. 1H NMR (400 MHz, METHANOL-d4) 5 8.15 (d, J= 8.34 Hz, 2H), 7.72 (d, J= 8.59 Hz, 2H), 7.29 – 7.37 (m, 2H), 7.14 – 7.28 (m, 3H), 4.45 (br. s., 2H), 3.55 (d, J= 10.36 Hz, 2H), 3.07 – 3.29 (m, 4H), 3.04 (dt, J= 3.98, 7.71 Hz, 1H), 2.61 (ddd, J= 3.66, 6.57, 10.23 Hz, 1H), 1.98 – 2.31 (m, 3H), 1.72 (br. s., 2H), 1.62 (ddd, J= 4.42, 6.51, 10.55 Hz, 1H), 1.41 (q, J= 6.82 Hz, lH); LC-MS Rt = 0.49 min; MS (ESI): 365.3 [M+H]+.

 

Neil Johnson

Neil Johnson

US Lead of Chemistry Talent Development, External Engagement and Recruitment at GSK

https://www.linkedin.com/in/neil-johnson-6628894

Experience

US Lead of Chemistry Talent Development, External Engagement and Recruitment

GSK

– Present (4 months)Greater Philadelphia Area

Manager

GSK

– Present (17 years)

Investgator

GlaxoSmithKline

– Present (17 years)

Senior Scientist

Cephalon

(4 years 10 months)

Education

The Johns Hopkins University

Doctor of Philosophy (PhD), Organic Chemistry

Fort Lewis College

BS, Chemistry

 

 

 

 

///////////GSK-2879552,  1401966-63-9, Phase I , A LSD1 inhibitor,  small cell lung cancer,  acute myeloid leukemia, 1401966-69-5, 1902123-72-1

O=C(O)C1=CC=C(CN2CCC(CN[C@H]3[C@H](C4=CC=CC=C4)C3)CC2)C=C1

O=C(O)c1ccc(cc1)CN2CCC(CC2)CN[C@@H]4C[C@H]4c3ccccc3

Share

GSK-2816126

 phase 1, Uncategorized  Comments Off on GSK-2816126
Jun 142016
 

STR1

GSK-2816126

N-[(1,2-Dihydro-4,6-dimethyl-2-oxo-3-pyridinyl)methyl]-3-methyl-1-[(1S)-1-methylpropyl]-6-[6-(1-piperazinyl)-3-pyridinyl]-1H-indole-4-carboxamide, GSK 126, GSK 2816126, GSK 2816126A

N-[(4,6-Dimethyl-2-oxo-1,2-dihydro-3-pyridinyl)methyl]-3-methyl-1-((1S)-1-methylpropyl)-6-[6-(1-piperazinyl)-3-pyridinyl]-1H-indole-4-carboxamide

Phase I

Formula
C31H38N6O2
Formula Wt.
526.67

An histone-lysine n-methyltransferase EZH2 inhibitor potentially for the treatment of B-cell lymphoma.

Research Code GSK-2816126; GSK-126; 2816126

CAS No. 1346574-57-9

  • Originator GlaxoSmithKline
  • Class Antineoplastics
  • Mechanism of Action EZH2 enzyme inhibitors; Histone modulators
  • Phase I Diffuse large B cell lymphoma; Follicular lymphoma
  • Preclinical Acute myeloid leukaemia

Most Recent Events

  • 31 Mar 2014 Phase-I clinical trials in Follicular lymphoma (Second-line therapy or greater) in USA and United Kingdom (IV)
  • 31 Mar 2014 Phase-I clinical trials in Diffuse large B cell lymphoma (Second-line therapy or greater) in USA and United Kingdom (IV)
  • 16 Jan 2014 Preclinical trials in Diffuse large B cell lymphoma & Follicular lymphoma in United Kingdom (IV)

GSK-126 is an inhibitor of mutant EZH2, a histone methyltransferase (HMT) that exhibits point mutations at key residues Tyr641 and Ala677; this compound does not appreciably affect WT EZH2. EZH2 is responsible for modulating expression of a variety of genes. GSK-126 competes with cofactor S-adenylmethionine (SAM) for binding to EZH2. GSK-126 displays anticancer chemotherapeutic activity by inhibiting proliferation in in vitro and in vivo models of diffuse large B-cell lymphoma.

SYNTHESIS

 

STR1

 

 

STR1

 

 

PATENT

CN 105541801

https://www.google.com/patents/CN105541801A?cl=zh

Example 79: Add (S) in a three-necked flask 100 Qiu – bromo – Shu – (isobutyl) – N – ((4,6-dimethyl-2-oxo -l, 2- dihydropyridin-3-yl) methyl) -3-methyl-1 hydrogen – indole carboxamide (365mg, 0.82mmol), 2- (piperazin-1-yl) pyridine-5-boronic acid pinacol ester (309mg, 1.07mmol, 1 · 3eq), potassium phosphate (522mg, 2.46mmol, 3eq), water, and I, 4- diepoxy-hexadecane as the solvent. Then, under nitrogen was added [I, Γ- bis (diphenylphosphino) ferrocene] dichloropalladium (II) dichloromethane complex (53.9mg, 0.066mmo 1), and at 90 ° C reaction, to give the desired product after purification 400mg (92% yield). Goo NMR (500MHz, DMSO- (I6) JO.70-0 · 78 (ιή, 3H), 1.37-1.44 (m, 4H), 1.75-1.87 (m, 2H), 2.11 (s, 3H), 2.16 ( s, 3H), 2.22-2.27 (m, 3H), 2.77-2.85 (m, 4H), 3.41-3.49 (m, 4H), 4.35 (d, J = 5.31Hz, 2H), 4.56-4.68 (m, lH), 5.87 (s, 1H), 6.88 (d, J = 8.84Hz, 1H), 7.17 (d J = 1.52Hz, 1H), 7.26 (s, lH), 7.73 (d J = 1.26Hz, 1H) , 7.91 (dd, J = 8.84Hz, lH), 8.16 (t, J = 5.05Hz, lH), 8.50 (d, J = 2.53Hz, lH); 13C NMR (125MHz, DMSO- (I6) Sll .6 , 12.6,19.1, 19.9,21.7,30.4,35.9,46.3,46.9,52.4,107.6,108.2,108.5,110.6,116.9,122.6,123.8, 130.6,131.5,136.7,138.6,143.5,146.4,150.2,159.2,164.0 , 169.6.

 

PATENT

WO 2013067296

Examples 267 and 268

(S)-6-bromo-1 -(sec-butyl)-N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-3- methyl-1 H-indole-4-carboxamide (Ex 267) and (R)-6-Bromo-1 -(sec-butyl)-N-((4,6- dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-3-methyl-1 H-indole-4-carboxamide (Ex 268)

Figure imgf000120_0001

6-Bromo-1-(sec-butyl)-N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methy methyl-1 H-indole-4-carboxamide (racemic mixture, 1.9 g) was resolved by chiral HPLC (column : Chiralpak AD-H, 5 microns, 50 mm x 250 mm, UV detection :240 nM, flow rate: 100 mL/min, T = 20 deg C, eluent: 60:40:0.1 n-heptane:ethanol:isopropylamine

(isocratic)). For each run, 100 mg of the racemic compound was dissolved in 30 volumes (3.0 ml.) of warm ethanol with a few drops of isopropylamine added. A total of 19 runs were performed. Baseline resolution was observed for each run. The isomer that eluted at 8.3-10.1 min was collected (following concentration) as a white solid, which was dried at 50 °C (< 5 mm Hg) to afford 901 mg, and was determined to be the S isomer* (Ex. 267; chiral HPLC: >99.5% ee (no R isomer detected). The isomer that eluted at 10.8-13.0 min was collected as a white solid, which was dried at 50 °C (< 5 mm Hg) to afford 865 mg, and was determined to be the R isomer* (Ex. 268; chiral HPLC: 99.2% ee; 0.4% S isomer detected). 1H NMR and LCMS were consistent with the parent racemate.

* The absolute configuration was determined by an independent synthesis of each enantiomer from the corresponding commercially available homochiral alcohols via Mitsunobu reaction. The sterochemical assignments were also consistent by vibrational circular dichroism (VCD) analysis.

Example 269

1-(sec-butyl)-N-((4,6-dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-3-methyl-6-(6- (piperazin-1 -yl)pyridin-3-yl)-1 -indole-4-carboxamide

Figure imgf000120_0002

Added sequentially to a reaction vial were 6-bromo-1 -(sec-butyl)-N-((4,6-dimethyl- 2-OXO-1 , 2-dihydropyridin-3-yl)methyl)-3-methyl-1 H-indole-4-carboxamide (0.15 g, 0.338 mmol), 1-(5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazine (0.127 g, 0.439 mmol), and potassium phosphate (tribasic) (0.287 g, 1.350 mmol), followed by 1 ,4- Dioxane (3 mL) and water (0.75 mL). The suspension was stirred under N2 degassing for 10 min., and then added PdCI2(dppf)-CH2CI2adduct (0.028 g, 0.034 mmol). The reaction vial was sealed, placed into a heat block at 95 °C, and stirred for 1.5 h. The contents were removed from heating and allowed to cool to room temperature. The aq layer was removed from bottom of the reaction vial via pipette. The reaction mixture was diluted into EtOAc (20 mL) followed by addition of 0.2 g each of Thiol-3 silicycle resin and silica gel. The volatiles were removed in vacuo and the residue dried on hi-vac for 1 h. The contents were purified by silica gel chromatography (dry loaded, eluent : A:

Dichloromethane, B: 10% (2M Ammonia in Methanol) in Chloroform, Gradient B: 8- 95%). The obtained solid was concentrated from TBME and dried in vacuum oven at 45 °C for 18 h. The product was collected as 129 mg (70%). 1H NMR (400 MHz, DMSO-d6) δ ppm 0.73 (t, J=7.33 Hz, 3H), 1.40 (d, J=6.57 Hz, 3H), 1.80 (dq, J=10.07, 7.08 Hz, 2H), 2.1 1 (s, 3H), 2.14 – 2.19 (m, 3H), 2.24 (s, 3H), 2.76 – 2.85 (m, 4H), 3.41 – 3.49 (m, 4H), 4.35 (d, J=5.05 Hz, 2H), 4.54 – 4.67 (m, 1 H), 5.87 (s, 1 H), 6.88 (d, J=8.84 Hz, 1 H), 7.17 (d, J=1.26 Hz, 1 H), 7.26 (s, 1 H), 7.73 (d, J=1.26 Hz, 1 H), 7.91 (dd, J=8.84, 2.53 Hz, 1 H), 8.16 (t, J=5.05 Hz, 1 H), 8.50 (d, J=2.53 Hz, 1 H), 1 1.48 (br. s.,1 H) ; LCMS MH+ =527.3.

Example 270

A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -[(1 S)-1 -methylpropyl]-6- [6-(1-piperazinyl)-3-pyridinyl]-1 H-indole-4-carboxamide

Figure imgf000121_0001

To a 30 mL microwave vial were added (S)-6-bromo-1 -(sec-butyl)-N-((4,6- dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-3-methyl-1 H-indole-4-carboxamide (100 mg, 0.225 mmol), 1 -(5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazine (85 mg, 0.293 mmol), 1 ,2-Dimethoxyethane (DME) (3 mL), water (1.000 mL) and sodium carbonate (0.338 mL, 0.675 mmol), and the mixture was degassed for 5 min by bubbling nitrogen. PdCI2(dppf)-CH2CI2 adduct (14.70 mg, 0.018 mmol) was added and the tube was sealed. The mixture was irradiated (microwave) at 140 °C for 10 min. The mixture was concentrated and the residue was taken up into MeOH and filtered. The filtrate was purified using reverse-phase HPLC (eluent: 25%ACN/H20, 0.1 % NH4OH to 60%

ACN/H20, 0.1 % NH4OH ) to give 91 mg of product as off-white solid. 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.70 – 0.78 (m, 3H), 1.37 – 1.44 (m, 3H), 1 .75 – 1.87 (m, 2H), 2.1 1 (s, 3H), 2.16 (s, 3H), 2.22 – 2.27 (m, 3H), 2.77 – 2.85 (m, 4H), 3.41 – 3.49 (m, 4H), 4.35 (d, J=5.31 Hz, 2H), 4.56 – 4.68 (m, 1 H), 5.87 (s, 1 H), 6.88 (d, J=8.84 Hz, 1 H), 7.17 (d, J=1.52 Hz, 1 H), 7.26 (s, 1 H), 7.73 (d, J=1.26 Hz, 1 H), 7.91 (dd, J=8.84, 2.53 Hz, 1 H), 8.16 (t, J=5.05 Hz, 1 H), 8.50 (d, J=2.53 Hz, 1 H); LCMS: 527.8 (MH+).

Example 271

A/-[(4,6-dimethyl-2-oxo-1 ,2-dihydro-3-pyridinyl)methyl]-3-methyl-1 -[(1 /?)-1-methylpropyl]- 6-[6-(1 -piperazinyl)-3-pyridinyl]-1 -indole-4-carboxamide

Figure imgf000122_0001

To a 30 mL microwave vial were added (R)-6-bromo-1-(sec-butyl)-N-((4,6- dimethyl-2-oxo-1 ,2-dihydropyridin-3-yl)methyl)-3-methyl-1 H-indole-4-carboxamide (100 mg, 0.225 mmol), 1 -(5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazine (85 mg, 0.293 mmol), 1 ,2-Dimethoxyethane (DME) (3 mL), water (1.000 mL) and sodium carbonate (0.338 mL, 0.675 mmol), and the mixture was degassed for 5 min by bubbling nitrogen. PdCI2(dppf)-CH2Cl2 adduct (14.70 mg, 0.018 mmol) was added and the tube was sealed. The mixture was irradiated (microwave) at 140 °C for 10 min. The mixture was concentrated and the residue was taken up into MeOH and filtered. The filtrate was purified using reverse-phase HPLC (eluent: 25%ACN/H20, 0.1 % NH4OH to 60%

ACN/H20, 0.1 % NH4OH ) to give 90 mg of product as off-white solid. 1 H NMR (400 MHz, DMSO-d6) δ ppm 0.73 (m, 3H), 1.41 (d, J=6.57 Hz, 3H), 1.81 (td, J=7.14, 2.91 Hz, 2H), 2.1 1 (s, 3H), 2.15 – 2.20 (m, 3H), 2.24 (s, 3H), 2.77 – 2.83 (m, 4H), 3.41 – 3.49 (m, 4H), 4.35 (d, J=5.05 Hz, 2H), 4.54 – 4.68 (m, 1 H), 5.87 (s, 1 H), 6.88 (d, J=8.84 Hz, 1 H), 7.17 (d, J=1.52 Hz, 1 H), 7.26 (s, 1 H), 7.73 (d, J=1.26 Hz, 1 H), 7.91 (dd, J=8.84, 2.53 Hz, 1 H), 8.16 (t, J=5.05 Hz, 1 H), 8.50 (d, J=2.27 Hz, 1 H); LCMS: 527.7 (MH+)

PATENT

WO 2011140324

Example 270

N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-3-methyl-l-[(15)-l-methylpropyl]-6-[6-(l-piperazinyl)-3-pyridinyl]-lH-indole-4-carboxamide

To a 30 niL microwave vial were added (S)-6-bromo-l-(sec-butyl)-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-3 -methyl- lH-indole-4-carboxamide (100 mg, 0.225 mmol), l-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazine (85 mg, 0.293 mmol), 1 ,2-Dimethoxyethane (DME) (3 mL), water (1.000 mL) and sodium carbonate (0.338 mL, 0.675 mmol), and the mixture was degassed for 5 min by bubbling nitrogen. PdCi2(dppf)-CH2Ci2 adduct (14.70 mg, 0.018 mmol) was added and the tube was sealed. The mixture was irradiated (microwave) at 140 °C for 10 min. The mixture was concentrated and the residue was taken up into MeOH and filtered. The filtrate was purified using reverse-phase HPLC (eluent: 25%ACN/H20, 0.1% NH4OH to 60% ACN/H20, 0.1% NH4OH ) to give 91 mg of product as off-white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.70 – 0.78 (m, 3H), 1.37 – 1.44 (m, 3H), 1.75 – 1.87 (m, 2H), 2.11 (s, 3H), 2.16 (s, 3H), 2.22 – 2.27 (m, 3H), 2.77 – 2.85 (m, 4H), 3.41 – 3.49 (m, 4H), 4.35 (d, J=5.31 Hz, 2H), 4.56 – 4.68 (m, IH), 5.87 (s, IH), 6.88 (d, J=8.84 Hz, IH), 7.17 (d, J=1.52 Hz, IH), 7.26 (s, IH), 7.73 (d, J=1.26 Hz, IH), 7.91 (dd, J=8.84, 2.53 Hz, IH), 8.16 (t, J=5.05 Hz, IH), 8.50 (d, J=2.53 Hz, IH); LCMS: 527.8 (MH+).

Example 271

N-[(4,6-dimethyl-2-oxo-l,2-dihydro-3-pyridinyl)methyl]-3-methyl-l-[(li?)-l-methylpropyl]-6-[6-(l-piperazinyl)-3-pyridinyl]-l -indole-4-carboxamide

To a 30 mL microwave vial were added (R)-6-bromo-l-(sec-butyl)-N-((4,6-dimethyl-2-oxo-l,2-dihydropyridin-3-yl)methyl)-3 -methyl- lH-indole-4-carboxamide (100 mg, 0.225 mmol), l-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridin-2-yl)piperazine (85 mg, 0.293 mmol), 1 ,2-Dimethoxyethane (DME) (3 mL), water (1.000 mL) and sodium carbonate (0.338 mL, 0.675 mmol), and the mixture was degassed for 5 min by bubbling nitrogen. PdCl2(dppf)-CH2Cl2 adduct (14.70 mg, 0.018 mmol) was added and the tube was sealed. The mixture was irradiated (microwave) at 140 °C for 10 min. The mixture was concentrated and the residue was taken up into MeOH and filtered. The filtrate was purified using reverse-phase HPLC (eluent: 25%ACN/H20, 0.1% NH4OH to 60% ACN/H20, 0.1% NH4OH ) to give 90 mg of product as off-white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.73 (m, 3H), 1.41 (d, J=6.57 Hz, 3H), 1.81 (td, J=7.14, 2.91 Hz, 2H), 2.11 (s, 3H), 2.15 – 2.20 (m, 3H), 2.24 (s, 3H), 2.77 – 2.83 (m, 4H), 3.41 – 3.49 (m, 4H), 4.35 (d, J=5.05 Hz, 2H), 4.54 -4.68 (m, 1H), 5.87 (s, 1H), 6.88 (d, J=8.84 Hz, 1H), 7.17 (d, J=1.52 Hz, 1H), 7.26 (s, 1H), 7.73 (d, J=1.26 Hz, 1H), 7.91 (dd, J=8.84, 2.53 Hz, 1H), 8.16 (t, J=5.05 Hz, 1H), 8.50 (d, J=2.27 Hz, 1H); LCMS: 527.7 (MH+).

REF

Tian X, Zhang S, Liu HM, et al. Histone lysine-specific methyltransferases and demethylases in carcinogenesis: new targets for cancer therapy and prevention. Curr Cancer Drug Targets. 2013 Jun 10;13(5):558-79. PMID: 23713993.

McCabe MT, Ott HM, Ganji G, et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature. 2012 Dec 6;492(7427):108-12. PMID: 23051747.

WO2005034845A2 * Jul 13, 2004 Apr 21, 2005 Supergen, Inc. Compositions and methods for treatment of cancer
WO2007053114A1 * Oct 30, 2006 May 10, 2007 S*Bio Pte Ltd Method of predicting a response to hdac inhibitors
WO2010090723A2 * Feb 2, 2010 Aug 12, 2010 University Of Georgia Research Foundation, Inc. Methods of inhibiting fibrogenesis and treating fibrotic disease
US20110035336 May 1, 2008 Feb 10, 2011 Yigang Cai Rating change for a prepaid session based on movement of a mobile device
US20110035340 Aug 7, 2009 Feb 10, 2011 Fibre-Craft Materials Corp. Decorating system and method of marketing and enhancing a surface area using a decorating system
US20110035344 Aug 6, 2009 Feb 10, 2011 International Business Machines Corporation Computing mixed-integer program solutions using multiple starting vectors
US20110064664 * Oct 8, 2008 Mar 17, 2011 The Board Of Regents Of The University Of Texas System Methods and compositions involving chitosan nanoparticles
WO2015077194A1 * Nov 18, 2014 May 28, 2015 Bristol-Myers Squibb Company Inhibitors of lysine methyl transferase
WO2015132765A1 * Mar 6, 2015 Sep 11, 2015 Glaxosmithkline Intellectual Property (No.2) Limited Enhancer of zeste homolog 2 inhibitors
WO2015141616A1 * Mar 16, 2015 Sep 24, 2015 第一三共株式会社 1,3-benzodioxole derivative
WO2016066697A1 * Oct 28, 2015 May 6, 2016 Glaxosmithkline Intellectual Property (No.2) Limited Enhancer of zeste homolog 2 inhibitors
US9051269 Nov 19, 2012 Jun 9, 2015 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9085583 Feb 11, 2013 Jul 21, 2015 Constellation—Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US20150344459 * Dec 20, 2013 Dec 3, 2015 Epizyme, Inc. 1,4-pyridone bicyclic heteroaryl compounds

/////////GSK-2816126,  GSK-126,  2816126, 1346574-57-9, GSK 126, GSK 126, GSK 2816126, GSK 2816126A

CC=5C=C(C)NC(=O)C=5CNC(=O)c1cc(cc2c1c(C)cn2[C@@H](C)CC)c3cnc(cc3)N4CCNCC4

Share

GSK-2838232

 phase 1, Uncategorized  Comments Off on GSK-2838232
Jun 132016
 

STR1

Figure imgf000135_0002

GSK-2838232

4-(((3aR,5aR,5bR,7aR,9S,11aR,11bR,13aS)-3a-((R)-2-((3-chlorobenzyl)(2-(dimethylamino)ethyl)amino)-1-hydroxyethyl)-1-isopropyl-5a,5b,8,8,11a-pentamethyl-2-oxo-3,3a,4,5,5a,5b,6,7,7a,8,9,10,11,11a,11b,12,13,13a-octadecahydro-2H-cyclopenta[a]chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid.

28-​Norlup-​18-​en-​21-​one, 3-​(3-​carboxy-​3-​methyl-​1-​oxobutoxy)​-​17-​[(1R)​-​2-​[[(4-​chlorophenyl)​methyl]​[2-​(dimethylamino)​ethyl]​amino]​-​1-​hydroxyethyl]​-​, (3β)​-

Phase I

Glaxosmithkline Llc INNOVATOR

Mark Andrew HATCHER, Brian Alvin Johns,Michael Tolar Martin, Elie Amine TABET, Jun Tang

A reverse transcriptase inhibitor potentially for the treatment of HIV infection.

GSK-2838232; GSK-8232; 2838232

CAS No. 1443460-91-0

C48H73ClN2O6,809.56

SYNTHESIS

PART 1

STR1

PART2

STR1

PART3

STR1

PART 4

STR1

AND UNWANTEDISOMER SHOWN BELOW

PART5

STR1

GSK2838232 is a novel human immune virus (HIV) maturation inhibitor being developed for the treatment of chronic HIV infection. GSK2838232 is a betulin derivative

Human immunodeficiency virus type 1 (HIV-1 ) leads to the contraction of acquired immune deficiency disease (AIDS). The number of cases of HIV continues to rise, and currently over twenty-five million individuals worldwide suffer from the virus. Presently, long-term suppression of viral replication with antiretroviral drugs is the only option for treating HIV-1 infection. Indeed, the U.S. Food and Drug Administration has approved twenty-five drugs over six different inhibitor classes, which have been shown to greatly increase patient survival and quality of life.

However, additional therapies are still required because of undesirable drug-drug interactions; drug-food interactions; non-adherence to therapy; and drug resistance due to mutation of the enzyme target.

Currently, almost all HIV positive patients are treated with therapeutic regimens of antiretroviral drug combinations termed, highly active antiretroviral therapy (“HAART”). However, HAART therapies are often complex because a combination of different drugs must be administered often daily to the patient to avoid the rapid emergence of drug-resistant HIV-1 variants. Despite the positive impact of HAART on patient survival, drug resistance can still occur. The emergence of multidrug-resistant HIV-1 isolates has serious clinical consequences and must be suppressed with a new drug regimen, known as salvage therapy.

Current guidelines recommend that salvage therapy includes at least two, and preferably three, fully active drugs. Typically, first-line therapies combine three to four drugs targeting the viral enzymes reverse transcriptase and protease. One option for salvage therapy is to administer different combinations of drugs from the same mechanistic class that remain active against the resistant isolates.

However, the options for this approach are often limited, as resistant mutations frequently confer broad cross-resistance to different drugs in the same class.

Alternative therapeutic strategies have recently become available with the development of fusion, entry, and integrase inhibitors. However, resistance to all three new drug classes has already been reported both in the lab and in patients. Sustained successful treatment of HIV-1 -infected patients with antiretroviral drugs will therefore require the continued development of new and improved drugs with new targets and mechanisms of action.

Presently, long-term suppression of viral replication with antiretroviral drugs is the only option for treating HIV-1 infection. To date, a number of approved drugs have been shown to greatly increase patient survival. However, therapeutic regimens known as highly active antiretroviral therapy (HAART) are often complex because a combination of different drugs must be administered to the patient to avoid the rapid emergence of drug-resistant HIV-1 variants. Despite the positive impact of HAART on patient survival, drug resistance can still occur.

The HIV Gag polyprotein precursor (Pr55Gag), which is composed of four protein domains – matrix (MA), capsid (CA), nucleocapsid (NC) and p6 – and two spacer peptides, SP1 and SP2, represents a new therapeutic target. Although the cleavage of the Gag polyprotein plays a central role in the progression of infectious virus particle production, to date, no antiretroviral drug has been approved for this mechanism.

In most cell types, assembly occurs at the plasma membrane, and the

MA domain of Gag mediates membrane binding. Assembly is completed by budding of the immature particle from the cell. Concomitant with particle release, the virally encoded PR cleaves Gag into the four mature protein domains, MA, CA, NC and p6, and the two spacer peptides, SP1 and SP2. Gag-Pol is also cleaved by PR, liberating the viral enzymes PR, RT and IN. Gag proteolytic processing induces a

morphological rearrangement within the particle, known as maturation. Maturation converts the immature, donut-shaped particle to the mature virion, which contains a condensed conical core composed of a CA shell surrounding the viral RNA genome in a complex with NC and the viral enzymes RT and IN. Maturation prepares the virus for infection of a new cell and is absolutely essential for particle infectivity.

Bevirimat (PA-457) is a maturation inhibitor that inhibits the final step in the processing of Gag, the conversion of capsid-SP1 (p25) to capsid, which is required for the formation of infectious viral particles. Bevirimat has activity against ART-resistant and wild-type HIV, and has shown synergy with antiretrovirals from all classes. Bevirimat reduced HIV viral load by a mean of 1.3 logi0/mL in patients who achieved trough levels of >= 20 μg/mL and who did not have any of the key baseline Gag polymorphisms at Q369, V370 or T371. However, Bevirimat users with Gag polymorphisms at Q369, V370 or T371 demonstrated significantly lower load reductions than patients without Gag polymorphisms at these sites.

Other examples of maturation inhibitors can be found in PCT Patent

Application No. WO201 1/100308, “Derivatives of Betulin”; PCT Patent Application No. PCT/US2012/024288, “Novel Anti-HIV Compounds and Methods of Use Thereof ; Chinese PCT Application No. PCT/CN201 1/001302, “Carbonyl Derivatives of Betulin”; Chinese PCT Application No. PCT/CN201 1/001303, “Methylene Derivatives of Betulin”; Chinese PCT Application Nos. PCT/CN201 1/002105 and PCT/CN201 1/002159, “Propenoate Derivatives of Betulin”. Maturation inhibitors in the prior art leave open gaps in the areas of polymorphism coverage whereby potency against a broad range of clinically relevant gag sequences is extremely important, along with overall potency including the clinically relevant protein adjusted antiviral activity that will be required for robust efficacy in long term durability trials. To date, no maturation inhibitor has achieved an optimal balance of these properties.

PATENT

WO 2013090664

https://www.google.com/patents/WO2013090664A1?cl=en

Example 17: Compound 50

4-(((3aR, 5aR, 5bR, 7aR, 9S, 11aR, 11bR, 13aS)-3a-((S)-1-Acetoxy-2-((4- chlorobenzyl)amino)ethyl)-1-isopropyl-5a, 5b, 8, 8, 11 a-pentamethyl-2-oxo- 3, 3a, 4, 5, 5a, 5b, 6, 7, 7a, 8,9, 10, 11, 11a, 11b, 12, 13, 13a-octadecahydro-2H- cyclopenta[a]chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid

Figure imgf000134_0001

[00241] The title compound was made in a similar manner to Example 16 and isolated as a TFA salt. 1H NMR (400MHz ,CHLOROFORM-d) δ = 7.49 – 7.30 (m, 4 H), 5.85 – 5.71 (m, 1 H), 4.59 – 4.40 (m, 1 H), 4.31 – 4.03 (m, 2 H), 3.41 – 2.79 (m, 4 H), 2.79 – 2.50 (m, 2 H), 2.37 (d, J = 18.1 Hz, 2 H), 2.02 – 0.63 (m, 49 H); LC/MS: m/z calculated 779.5, found 780.3 (M+1 )+.

Figure imgf000135_0001

Example 18: Compound 51

4-(((3aR, 5aR, 5bR, 7aR, 9S, 11aR, 11bR, 13aS)-3a-((R)-2-((4-Chlorobenzyl)(2- (dimethylamino)ethyl)amino)-1-hydroxyethyl)-1-isopropyl-5a,5b,8,8, 11a-pe

2-0X0-3, 3a, 4, 5, 5a, 5b, 6, 7, 7a, 8,9, 10, 11, 11a, 11b, 12, 13, 13a-octadecahydro-2H- cyclopenta[a]chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid

Figure imgf000135_0002

[00242] To a solution of 2-(dimethylamino)acetaldehyde, hydrochloride (6.75 g, 54.6 mmol) in methanol (20 ml_) was added 4-

(((3aR,5aR,5bR,7aR,9S, 1 1 aR, 1 1 bR, 13aS)-3a-((R)-2-((4-chlorobenzyl)amino)-1 – hydroxyethyl)-1 -isopropyl-5a,5b,8,8, 1 1 a-pentamethyl-2-oxo- 3,3a,4,5,5a,5b,6,7,7a,8,9,10,1 1 ,1 1 a,1 1 b,12,13,13a-octadecahydro-2H- cyclopenta[a]chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid , Trifluoroacetic acid salt (46) (9.5 g, 10.92 mmol). The pH was adjusted to 7-8 with Et3N. The reaction mixture was stirred at rt for 2 h. Sodium cyanoborohydride (0.686 g, 10.92 mmol) was then added and the mixture was stirred at rt overnight. After the reaction was complete, water (15 ml_) and EtOAc (15 ml_) were added, and then the organic phase was removed and concentrated under reduced presure. The product was extracted with EtOAc (80 ml_x3), the combined organic phase was washed with brine, dried, and concentrated. The product was purified by flash chromatography (DCM:EtOAc=2: 1 to 1 : 1 , then DCM:MeOH=100: 1 to 20: 1 ) to give 4- (((3aR,5aR,5bR,7aR,9S, 1 1 aR, 1 1 bR, 13aS)-3a-((R)-2-((4-chlorobenzyl)(2- (dimethylamino)ethyl)amino)-1 -hydroxyethyl)-1 -isopropyl-5a,5b,8,8, 1 1 a-pentamethyl- 2-0X0-3, 3a,4, 5, 5a, 5b, 6, 7, 7a, 8, 9, 10, 1 1 , 1 1 a, 1 1 b, 12, 13, 13a-octadecahydro-2H- cyclopenta[a]chrysen-9-yl)oxy)-2,2-dimethyl-4-oxobutanoic acid (51 ) (6 g, 7.41 mmol, 67.9 % yield) as white solid. Multiple batches of this material (were combined 95 g), dissolved in 600 mL of dichloromethane and washed with NaHC03 (400 ml_*3) and the organic phase was dried over Na2S04, filtered and concentrated. The solids were washed with a mixture of EtOAc: petroleum ether (600 mL), and filtered followed by lyophilization to provide the final title compound 62 g as a white solid. 1H NMR (400MHz ,METHANOL-d4) δ = 7.47 – 7.29 (m, 4 H), 4.48 (dd, J = 5.8, 10.3 Hz, 1 H), 4.15 – 4.04 (m, 1 H), 3.80 (d, J = 13.8 Hz, 1 H), 3.57 (d, J = 14.1 Hz, 1 H), 3.21 – 2.82 (m, 5 H), 2.72 – 2.41 (m, 9 H), 2.37 – 2.05 (m, 4 H), 2.05 – 0.74 (m, 45 H);

LC/MS: m/z calculated 808.5, found 809.5 (M+1 )+.

Figure imgf000137_0001

REFERENCES

Hatcher, Mark Andrew; Johns, Brian Alvin; Martin, Michael Tolar; Tabet, Elie Amine; Tang, Jun.  Preparation of betulin derivatives for the treatment of HIV, PCT Int. Appl. (2013), WO 2013090664 A1 20130620.

 

 

Mark Hatcher

Director, US R&D Policy and Scientific Affairs at GlaxoSmithKline

https://www.linkedin.com/in/mark-hatcher-232b904

 

Jun Tang

Chief Scientist at GlaxoSmithKline

https://www.linkedin.com/in/jun-tang-2a50629

Brian Johns

Chemistry Director, GlaxoSmithKline

https://www.linkedin.com/in/brian-johns-26a5953

////////GSK-2838232, 1443460-91-0, GSK 2838232,  GSK-8232,  2838232,  treatment of HIV, phase1

O=C(C1)C(C(C)C)=C2[C@@]1([C@@H](O)CN(CCN(C)C)CC3=CC=CC(Cl)=C3)CC[C@]4(C)[C@]2([H])CC[C@@]5([H])[C@@]4(C)CC[C@]6([H])[C@]5(C)CC[C@H](OC(CC(C)(C)C(O)=O)=O)C6(C)C

Karl Landsteiner’s 148th birthday

Share

TD 1607

 phase 1, Uncategorized  Comments Off on TD 1607
Jun 132016
 

STR1

STR1

TD-1607

Phase I

A glycopeptide-cephalosporin heterodimer potentially for the treatment of gram-positive bacterial infection.

CAS No. 827040-07-3

C95 H109 Cl3 N18 O31 S2, 
Molecular Weight, 2169.47
Vancomycin, 29-​[[[2-​[[6-​[[[1-​[[(6R,​7R)​-​7-​[[(2Z)​-​2-​(2-​amino-​5-​chloro-​4-​thiazolyl)​-​2-​(methoxyimino)​acetyl]​amino]​-​2-​carboxy-​8-​oxo-​5-​thia-​1-​azabicyclo[4.2.0]​oct-​2-​en-​3-​yl]​methyl]​pyridinium-​4-​yl]​methyl]​amino]​-​1,​6-​dioxohexyl]​amino]​ethyl]​amino]​methyl]​-​, inner salt
Vancomycin, 29-[[[2-[[6-[[[1-[[(6R,7R)-7-[[(2Z)-(2-amino-5-chloro-4-thiazolyl)(methoxyimino)acetyl]amino]-2-carboxy-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-en-3-yl]methyl]pyridinium-4-yl]methyl]amino]-1,6-dioxohexyl]amino]ethyl]amino]methyl]-, inner salt
  • Originator Theravance
  • Developer Theravance Biopharma
  • Class Antibacterials; Cephalosporins; Glycopeptides
  • Mechanism of Action Cell wall inhibitors
    • Phase I Gram-positive infections

    Most Recent Events

    • 21 Apr 2016 Phase I development is ongoing in USA
    • 01 Jul 2014 Theravance completes a phase I trial in Healthy volunteers in in USA (NCT01949103)
    • 02 Jun 2014 Theravance Biopharma is formed as a spin-off of Theravance
    • Company Theravance Biopharma Inc.
      Description Glycopeptide cephalosporin heterodimer antibiotic
      Molecular Target
      Mechanism of Action
      Therapeutic Modality Small molecule: Combination
      Latest Stage of Development Phase I
      Standard Indication Gram-negative bacterial infection
      Indication Details Treat Gram-positive bacterial infections

PATENT
WO 2005005436

The present invention provides novel cross-linked glycopeptide – cephalosporin compounds that are useful as antibiotics. The compounds of this invention have a unique chemical structure in which a glycopeptide group is covalently linked to a pyridinium moiety of a cephalosporin group. Among other properties, compounds of this invention have been found to possess surprising and unexpected potency against Gram-positive bacteria including methicillin-resistant Staphylococci aureus (MRSA). Accordingly, in one aspect, the invention provides a compound of formula I:

Figure imgf000004_0001
////////Theravance Biopharma, TD 1607, phase 1, glycopeptide-cephalosporin heterodimer ,  gram-positive bacterial infection
Share

DS 2330 by Daiichi Sankyo

 phase 1  Comments Off on DS 2330 by Daiichi Sankyo
Apr 072016
 

 

str1

DS 2330

 

 

4-[2-(4-{[2-({3-[(trans-4-carboxy-cyclohexyl)(ethyl)sulfocarbamoyl]benzoyl}amino)-5-(piperidin-1-yl)benzoyl]amino}phenyl)ethyl]benzoic acid,

4- [2- (4 – {[2 – ({3 – [(trans-4-carboxy-cyclohexyl) (ethyl) sulfur carbamoyl] benzoyl} amino) -5- (piperidin-1-yl) benzoyl] amino} phenyl) ethyl] benzoate

CAS 1634680-81-1
C43 H48 N4 O8 S, 780.9
Benzoic acid, 4-​[2-​[4-​[[2-​[[3-​[[(trans-​4-​carboxycyclohexyl)​ethylamino]​sulfonyl]​benzoyl]​amino]​-​5-​(1-​piperidinyl)​benzoyl]​amino]​phenyl]​ethyl]​-
CIS CAS 1634681-85-8
DISODIUM SALT 1634681-00-7
  • OriginatorDaiichi Sankyo Inc
  • ClassHyperphosphataemia therapies

useful for treating hyperphosphatemia, DS-2330, a phosphorous lowering agent, being developed by Daiichi Sankyo, for treating hyperphosphatemia in chronic kidney disease. In April 2016, DS-2330 was reported to be in phase 1 clinical development.

  • Phase IHyperphosphataemia
  • 31 Oct 2015Phase-I clinical trials in Hyperphosphataemia in USA (unspecified route)

 

str1

 

SEE  WO2015108038,

PATENT

WO2014175317

http://www.google.com/patents/EP2990400A1?cl=en

 

str1

PATENT

WO-2016047613

he problem is to provide a pharmaceutical for the prevention or treatment of hyperphosphatemia. The solution is a salt of a compound including formula (I), or a crystal of a hydrate thereof.

 

 

 

 

 

 

(Example 1)
disodium 4- [2- (4 – {[2 – ({3 – [(trans-4-carboxy-cyclohexyl) (ethyl) sulfur carbamoyl] benzoyl} amino) -5- (piperidin-1-yl ) benzoyl] amino} phenyl) ethyl] benzoic acid trihydrate
Disodium 4- [2- (4 – { [2 – ({3 – [(trans-4-carboxylatocyclohexyl) (ethyl) sulfamoyl] benzoyl} amino) – 5- (piperidin-1-yl) benzoyl] amino} phenyl) ethyl] benzoate trihydrate
of α crystal

 

[Formula 7] crystal of disodium salt trihydrate of (α crystal)

 

(1)
4- [2- (4 – {[2 – ({3 – [(trans-4-carboxy-cyclohexyl) (ethyl) sulfur carbamoyl] benzoyl} amino) -5- (piperidin-1-yl) benzoyl] amino} phenyl) ethyl] 1 mol / L NaOH aqueous solution to benzoic acid (1.2 g) (3.1 mL) was added and dissolved completely. After stirring at room temperature for 1 day was added acetonitrile (60 mL), at 40 ° C.
and stirred for further 1 day. The precipitated solid was collected by filtration, and 3 hours drying under reduced pressure at room temperature to give the title compound 1.1 g (85%).
(2)
 4- [2- (4 – {[2 – ({3 – [(trans-4-carboxy-cyclohexyl) (ethyl) sulfur carbamoyl] benzoyl} amino) -5- (piperidin-1-yl) benzoyl] amino} phenyl) ethyl] benzoate (40.0 g)
in water (46.4 mL), 1-PrOH (72 mL), 4 mol / L NaOH aqueous solution (25.54 mL) was added, then filtered after stirring insolubles at room temperature, water / 1-PrOH: was washed with (3 7, 80 mL). The filtrate was heated up to 40 ℃, 1-PrOH the (160 mL) was added, and further seed crystal (α crystals, 0.2g) was added. Then the temperature was raised to 50 ℃, 1-PrOH (96 ml) was added, and the mixture was stirred overnight.Thereafter, 1-PrOH (480 ml) was added and after overnight stirring, was collected by filtration the precipitated solid was cooled to room temperature.Thereafter, and vacuum dried overnight at 40 ° C., to give the title compound 39.4 g (96%).

 

REFERENCES

http://www.daiichisankyo.com/media_investors/investor_relations/ir_calendar/files/005280/Presentation%20Material.pdf

////////////DS 2330, DS-2330, DAIICHI SANKYO, phase 1

O=C(O)[C@@H]1CC[C@H](CC1)N(CC)S(=O)(=O)c2cccc(c2)C(=O)Nc5ccc(cc5C(=O)Nc4ccc(CCc3ccc(cc3)C(=O)O)cc4)N6CCCCC6

OR

O=C(O)[C@@H]1CC[C@H](CC1)N(CC)S(=O)(=O)c2cccc(c2)C(=O)Nc5ccc(cc5C(=O)Nc4ccc(CCc3ccc(cc3)C(=O)O)cc4)N6CCCCC6

 

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

Share

GDC-0084

 phase 1, Uncategorized  Comments Off on GDC-0084
Apr 062016
 

 

GDC-0084
CAS#: 1382979-44-3
Chemical Formula: C18H22N8O2
Exact Mass: 382.1866

Synonym: RG7666; RG-7666; RG 7666; GDC-0084; GDC0084; GDC 0084.

IUPAC/Chemical Name: 5-(6,6-dimethyl-4-morpholino-8,9-dihydro-6H-[1,4]oxazino[4,3-e]purin-2-yl)pyrimidin-2-amine

Company Roche
Description Phosphoinositide 3-kinase (PI3K) inhibitor
Molecular Target Phosphoinositide 3-kinase (PI3K)
Mechanism of Action Phosphoinositide 3-kinase (PI3K) inhibitor
Therapeutic Modality Small molecule
Latest Stage of Development Phase I
Standard Indication Brain cancer
Indication Details Treat progressive or recurrent high-grade glioma
Regulatory Designation
Partner Genentech Inc.
  • Originator Genentech
  • Class Antineoplastics; Small molecules
  • Mechanism of Action 1 Phosphatidylinositol 3 kinase inhibitors
  • 28 Jan 2015 Discontinued – Phase-I for Glioma in Spain (unspecified route)
  • 28 Jan 2015 Discontinued – Phase-I for Glioma in USA (unspecified route)
  • 01 Jan 2015 Genentech completes a phase I trial in Glioma in USA and Spain (NCT01547546)

GDC-0084, also known as RG7666, is a phosphatidylinositol 3-kinase (PI3K) inhibitor with potential antineoplastic activity. PI3K inhibitor GDC-0084 specifically inhibits PI3K in the PI3K/AKT kinase (or protein kinase B) signaling pathway, thereby inhibiting the activation of the PI3K signaling pathway. This may result in the inhibition of both cell growth and survival in susceptible tumor cell populations. Activation of the PI3K signaling pathway is frequently associated with tumorigenesis.

 

str1

http://pubs.acs.org/doi/pdf/10.1021/acsmedchemlett.6b00005

 

 

Abstract Image

An improved, efficient process with a significantly reduced process mass intensity (PMI) led to the multikilogram synthesis of a brain penetrant PI3K inhibitor GDC-0084. Highlights of the synthesis include a phase transfer catalyzed annulation in water, an efficient Suzuki-Miyaura cross-coupling of a chloropyrimidine with an arylboronic acid using a low palladium catalyst loading, and the development of a controlled crystallization to provide the API. The process delivered GDC-0084 with low levels of both impurities and residual metals.

Development of an Efficient, Safe, and Environmentally Friendly Process for the Manufacture of GDC-0084

Small Molecule Process Chemistry, Small Molecule Analytical Chemistry, Genentech, Inc., A Member of the Roche Group, 1 DNA Way, South San Francisco, California 94080, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00011
Publication Date (Web): March 11, 2016
Copyright © 2016 American Chemical Society

//////GDC-0084

NC1=NC=C(C2=NC(N3CCOCC3)=C4N=C(C(C)(C)OCC5)N5C4=N2)C=N1

str1

str1

5-(6,6-Dimethyl-4-morpholino-8,9-dihydro-6H-[1,4]oxazino[4,3-e]purin-2-yl)pyrimidin-2-amine GDC-0084 

mp 211 °C; 1H NMR (500 MHz, DMSO-d6) δ 9.09 (s, 2H), 7.03 (s, 2H), 4.32–4.17 (m, 4H), 4.17–4.04 (m, 4H), 3.84–3.65 (m, 4H), 1.58 (s, 6H); 13C NMR (125 MHz, DMSO-d6) δ 163.8, 157.6, 154.2, 152.5, 151.3, 151.0, 120.3, 117.3, 73.7, 66.2, 57.8, 45.2, 41.5, 27.3. HRMS [M + H]+calcd for C18H22N8O2 383.1938; found 383.1945.

  1. The Discovery of Clinical Development Candidate GDC-0084, a Brain Penetrant Inhibitor of Class I Phosphoinositide 3-Kinases (PI3K) and mTOR.

    HeffronT.NdubakuC.SalphatiL.AlickeB.CheongJ.;DrobnickJ.EdgarK.GouldS.LeeL.LesnickJ.LewisC.NonomiyaJ.Pangj.PliseE.Sideris,S.WallinJ.WangL.ZhangX.OliveroA. ACS Med. Chem. Lett. 2016, , DOI: 10.1021/acsmedchemlett.6b00005

  2. 3.

    (a) Purine Derivatives Useful as PI3 Kinase Inhibitors. GoldsmithP.HancoxT. C.HudsonA.PeggN. A.KulagowskiJ. J.NadinA. J.PriceS. PCT Int. Appl. WO 2009053716 A1 Apr 30, 2009.

    (b) Preparation of Purine Derivatives with PI3K Inhibitory Activity and Methods of Use Thereof. CastanedoG.Chuckowree,I.FolkesA.SutherlinD. P.WanN. C. PCT Int. Appl. WO 2009146406 A1 Dec 3, 2009

Share

P7435 from Piramal Enterprises Mumbai, India

 phase 1  Comments Off on P7435 from Piramal Enterprises Mumbai, India
Apr 052016
 

str1

str1

P7435

Piramal Enterprises Mumbai, India

P-7435; P7435-DGAT1, P7435, P 7435

CAS 1210756-48-1,
C22 H19 F N4 O4 S
L-​Valine, N-​[[3-​[4-​[(6-​fluoro-​2-​benzothiazolyl)​amino]​phenyl]​-​5-​isoxazolyl]​carbonyl]​-
Molecular Weight, 454.47

GDAT1 inhibitor

  • Phase IDiabetes mellitus; Lipid metabolism disorders
  • ClassAntihyperglycaemics; Antihyperlipidaemics; Small molecules
  • Mechanism of ActionDiacylglycerol O acyltransferase inhibitors
Company Piramal Enterprises Ltd.
Description Diacylglycerol O-acyltransferase-1 (DGAT1) inhibitor
Molecular Target Diacylglycerol O-acyltransferase-1 (DGAT1)
Mechanism of Action Diacylglycerol O-acyltransferase-1 (DGAT1) inhibitor
Therapeutic Modality
Latest Stage of Development Phase I
Standard Indication Metabolic (unspecified)
Indication Details Treat metabolic disorders

https://clinicaltrials.gov/ct2/show/NCT01910571

https://clinicaltrials.gov/ct2/show/NCT01764425

  • 24 Nov 2014Piramal Enterprises completes a phase I trial in healthy, overweight or obese subjects in USA (NCT01910571)
  • 17 Jun 2014Adverse events and pharmacokinetics data from a phase I trial in healthy male volunteers presented at the 74th Annual Scientific Sessions of the American Diabetes Association (ADA-2014)
  • 17 Jun 2014Pharmacodynamics data from preclinical studies in Dyslipidaemia and obesity presented at the 74th Annual Scientific Sessions of the American Diabetes Association (ADA-2014)

Chairman Ajay Piramal

Swati Piramal-The Vice Chairperson of Piramal Enterprises Ltd

Nandini Piramal, Executive Director, Piramal Enterprises

Piramal Enterprises gets US FDA approval for P7435 IND

http://www.pharmabiz.com/NewsDetails.aspx?aid=76992&sid=2

Our Bureau, Mumbai
Tuesday, August 06, 2013, 12:25 Hrs  [IST]

Piramal Enterprises Ltd has received US Food and Drug Administration (FDA) approval for its Investigational New Drug (IND) P7435. This is a novel, potent and highly selective, oral diacylglycerolacyltransferase 1 (DGAT1) inhibitor.

P7435 has been developed by the NCE Research Division of PEL for the management of metabolic disorders such as lipid abnormalities and diabetes. It is well-established that increased lipid levels’ (including triglycerides) is one of the major risk factors for cardiovascular disease (CVD). It has been reported by the World Health Organisation, that CVD, is the number one cause of deaths globally, representing approximately 30 per cent of all deaths. Currently, there is a significant medical need for effective and safe drugs for the management of lipid abnormalities and metabolic disorders.

P7435 has demonstrated its lipid lowering potential in various preclinical studies by showing significant reduction in triglyceride levels, glucose and insulin levels,and decrease in food intake and body weight gain -factors which are associated with lipid abnormalities and metabolic disorders.

PEL has established the safety and tolerability of P7435 in a phase I trial recently completed in India. This extension trial in the US will further evaluate the safety and efficacy of P7435 in a larger population.

Dr Swati Piramal, vice chairperson, Piramal Enterprises, said, “The NCE Research division of PEL continues its ambitious diabetes/metabolic disorders programme to discover and develop NCEs to fight against diseases like diabetes and lipid disorders. With P7435 we are looking at addressing a serious need for effective and well-tolerated drugs that treat lipid disorders, which are commonly associated with diabetes and CVDs. Expansion of this trial will allow testing this NCE in a wider population,which is critical to the development of this drug and will provide therapeutic solutions not just to India but also to the rest of the world.”

The NCE Research division of Piramal Enterprises focuses on the discovery and development of innovative small molecule medicines to improve the lives of patients suffering from cancer, metabolic disorders and inflammatory conditions. The key elements of its strategy include capitalizing on Piramal’s strengths, in particular the India advantage, and leveraging external partnerships to achieve high levels of R&D productivity. Piramal’s state-of-the-art Research Centre in Mumbai has comprehensive capabilities spanning target identification all the way through clinical development. Its robust pipeline, including 8 compounds in clinical development, bears testimony to its innovative and rigorous drug discovery process.

PAPER

European Journal of Medicinal Chemistry (2012), 54, 324-342

http://www.sciencedirect.com/science/article/pii/S0223523412003133

PATENT

WO 2010023609

http://www.google.co.in/patents/WO2010023609A1?cl=en

/////////Piramal Enterprises,  Mumbai, India, P-7435, P7435-DGAT1, P7435, P 7435, GDAT1 inhibitor

O=C(O)[C@@H](NC(=O)c1cc(no1)c2ccc(cc2)Nc3nc4ccc(F)cc4s3)C(C)C

Share

LY 2922470

 phase 1, Uncategorized  Comments Off on LY 2922470
Mar 292016
 

str1

LY 2922470

as per WO2013025424A1

Figure imgf000004_0001

 
LY 2922470

Picture credit….

SCHEMBL14695980.png

(3S)-3-[4-[[5-[(8-methoxy-3,4-dihydro-2H-quinolin-1-yl)methyl]thiophen-2-yl]methoxy]phenyl]hex-4-ynoic acid

Benzenepropanoic acid, 4-​[[5-​[(3,​4-​dihydro-​8-​methoxy-​1(2H)​-​quinolinyl)​methyl]​-​2-​thienyl]​methoxy]​-​β-​1-​propyn-​1-​yl-​, (βS)​-

Glucose Lowering Agents, Signal Transduction Modulators

CAS 1423018-12-5
Molecular Formula: C28H29NO4S
Molecular Weight: 475.59916 g/mol

https://clinicaltrials.gov/ct2/show/NCT01867216

  • Phase I Type 2 diabetes mellitus

Eli Lilly

Eli Lilly And Company

Antihyperglycaemics

  • 28 Jan 2014 Eli Lilly completes a phase I trial in Type-2 diabetes mellitus in USA (NCT01867216)
  • 30 Jun 2013 Phase-I clinical trials in Type-2 diabetes mellitus in USA (PO)
  • 14 Jun 2013 Eli Lilly plans a phase I trial for Type-2 diabetes mellitus in USA (NCT01867216)

 

PATENT

WO 2013025424

https://www.google.com/patents/US20130045990?cl=de

Also published as CA2843474A1, CA2843474C, CN103687856A, CN103687856B, EP2744806A1, US8431706, WO2013025424A1, Less «
Inventors Chafiq Hamdouchi
Original Assignee Eli Lilly And Company

 

 

Figure US20130045990A1-20130221-C00001

 

Figure US20130045990A1-20130221-C00004

Figure US20130045990A1-20130221-C00005

Preparation 18-Methoxyquinoline

Add potassium hydroxide (435 g, 7.76 mol) to a solution of 8-hydroxy quinoline (250 g, 1.724 mol) in THF (10 L) at ambient temperature and stir. Add methyl iodide (435 g, 2.58 mol) dropwise and stir overnight. Filter the reaction mixture and wash the solid with THF (2 L). Concentrate the solution to dryness; add water; extract with dichloromethane (2×3 L); combine the organic layers; and wash with brine. Collect the organic layers and dry over sodium sulfate. Remove the solids by filtration. Collect the filtrate and concentrate under reduced pressure to give a red oil, which solidifies on standing, to give the title compound (281 g, 102%), which can be used without further purification. ESI (m/z) 160(M+H).

Preparation 2

8-Methoxy-1,2,3,4-tetrahydroquinoline

Add sodium cyanoborohydride (505 g, 8.11 mol) in EtOH (1 L) to a solution of 8-methoxy quinoline (425 g, 2.673 mol) in EtOH (9 L), and stir. Cool the reaction mixture to an internal temperature of 0° C. and add HCl (35%, 1.12 L, 10.962 mol) dropwise over 60 min so that the internal temperature did not rise above 20° C. Allow the reaction mixture to warm to ambient temperature and then heat to reflux for 2.5 hours. Cool to ambient temperature and stir overnight. Add ammonium hydroxide (25%, 1 L); dilute with water (15 L); and extract the mixture with dichloromethane (3×10 L). Combine the organic layers and dry over sodium sulfate. Remove the solids by filtration. Collect the filtrate and concentrate under reduced pressure to give a residue. Purify the residue by silica gel flash chromatography, eluting with ethyl acetate: hexane (1:10) to give the title compound (357 g, 82%). ESI (m/z) 164(M+H).

Preparation 3

Methyl-5-methylthiophene-2-carboxylate

Add thionyl chloride (153 ml, 2.1 mol) dropwise over 20 min to a solution of 5-methylthiophene-2-carboxylic acid (100 g, 0.703 mol) in MeOH (1 L) at 0° C. and stir. After the addition is complete, heat the reaction mixture to reflux for 3.5 hours. Cool and concentrate in vacuo to give a thick oil. Dilute the oil with EtOAc (500 ml) and sequentially wash with water (300 ml) then brine (300 ml). Dry the organic layer over sodium sulfate. Remove the solids by filtration. Collect the filtrate and concentrate under reduced pressure to give the title compound (106 g, 97%), which is used without further purification. ESI (m/z) 156(M+H).

Preparation 4

Methyl 5-(bromomethyl)thiophene-2-carboxylate

Add freshly recrystallised NBS (323.8 g, 1.81 mol) to a solution of methyl-5-methylthiophene-2-carboxylate (258 g, 1.65 mol) in chloroform (2.6 L) at room temperature, and stir. Add benzoyl peroxide (3.99 g, 0.016 mol) and heat the reaction mixture to reflux for 7 hours. Cool the reaction mixture to ambient temperature and filter through diatomaceous earth. Wash the filter cake with chloroform (250 ml). Collect the organic layers and remove the solvent to give the title compound (388 g, 100%), which is used without further purification. ESI (m/z) 236(M+H).

Preparation 5

Methyl-5-[8-methoxy-3,4-dihydro-2H-quinolin-1-yl)methyl]thiophene-2-carboxylate

Add methyl-5-(bromoethyl)thiophene-2-carboxylate (432.5 g, 1.84 mol) in EtOH (500 ml) to a solution of 8-methoxy-1,2,3,4-tetrahydroquinoline (300 g 1.84 mol) in EtOH (1 L) and stir. Add DIPEA (641 ml, 3.67 mol) dropwise and stir at room temperature overnight. After completion of the reaction, remove the EtOH in vacuo, and add water (5 L). Extract the aqueous with EtOAc (3×3 L); combine the organic layers; and dry over sodium sulfate. Filter the solution and concentrate under reduced pressure to give a residue. Purify the residue by silica gel flash chromatography eluting with ethyl acetate: hexane (6:94) to give the title compound (325 g, 56%). ESI (m/z) 318(M+H).

Preparation 6

[5-[(8-Methoxy-3,4-dihydro-2H-quinolin-1-yl)methyl]-2-thienyl]methanol

Add DIBAL-H (1 M in toluene 2.7 L, 2.66 mol) slowly via a cannula over a period of 1.5 h to a stirred solution of methyl-5-(8-methoxy-3,4-dihydroquinolin-1(2H)-yl)methyl)thiophene-2-carboxylate (281 g, 0.886 mol) in THF (4 L) at −70° C. Monitor the reaction via thin layer chromatography (TLC) for completion. After completion of the reaction, allow the reaction mixture to warm to 20° C. and add a saturated solution of ammonium chloride. Add a solution of sodium potassium tartrate (1.3 Kg in 5 L of water), and stir overnight. Separate the organic layer; extract the aqueous phase with EtOAc (2×5 L); then combine the organic layers; and dry the combined organic layers over sodium sulfate. Remove the solids by filtration. Remove the solvent from the filtrate under reduced pressure to give the title compound as a white solid (252 g, 98%). ESI (m/z) 290(M+H).

Preparation 7

Ethyl(3S)-3-[4-[[5-[(8-methoxy-3,4-dihydro-2H-quinolin-1-yl)methyl]-2-thienyl]methoxy]phenyl]hex-4-ynoate

Add tributylphosphine (50% solution in EtOAc, 543 ml, 1.34 mol) to a solution of ADDP (282.5 g, 1.5 eq) in THF (3 L) and cool the mixture to an internal temperature of 0° C., then stir for 15 minutes. Add (S)-ethyl 3-(4-hydroxyphenyl)hex-4-ynoate (173.5 g, 0.747 mol) in THF (3 L) dropwise over 15 min; then add 5-((8-methoxy-3,4-dihydroquinolin-1(2H)-yl)methyl)thiophene-2-yl)methanol (216 g, 0747 mol) in THF (5 L) dropwise. Allow the reaction mixture to warm to ambient temperature and stir overnight. Filter the reaction mixture through diatomaceous earth and wash the filter cake with ethyl acetate (2 L). Concentrate the organic filtrate to dryness. Add water (4 L); extract with ethyl acetate (3×5 L); combine the organic layers; and dry the combined organic layers over sodium sulfate. Remove the solids by filtration and concentrate under reduced pressure to give an oil. Purify the residue by silica gel flash chromatography by eluting with ethyl acetate: hexane (6:94) to give the title compound (167 g, 44%). ESI (m/z) 504(M+H).

Example 1

(3S)-3-[4-[[5-[(8-Methoxy-3,4-dihydro-2H-quinolin-1-yl)methyl]-2-thienyl]methoxy]phenyl]hex-4-ynoic acid

Figure US20130045990A1-20130221-C00006

Add a solution of potassium hydroxide (49.76 g, 0.88 mol) in water (372 ml) to a solution of (S)-ethyl-3-(4-((5-8-methoxy-3,4-dihydroquinolin-1(2H)-yl)methyl)thiophen-2-yl)methoxy) phenyl)hex-4-ynoate (149 g, 0.296 mol) in EtOH (1.49 L) at room temperature and stir overnight. Concentrate the reaction mixture to dryness and add water (1.3 L). Extract the resulting solution with EtOAc (2×300 ml) and separate. Adjust the pH of the aqueous layer to pH=6 with 2 N HCl. Collect the resulting solids. Recrystallise the solids from hot MeOH (298 ml, 2 vol) to give the title compound (91 g, 65%). ESI (m/z) 476(M+H).

 

Abstract

GPR40 agonists for the treatment of type 2 diabetes: From the laboratory to the patient
251st Am Chem Soc (ACS) Natl Meet (March 13-17, San Diego) 2016, Abst MEDI 260

str1

str1

Presenter

Chafiq Hamdouchi

Chafiq Hamdouchi

Senior Research Advisor at Eli Lilly and Company

https://www.linkedin.com/in/chafiq-hamdouchi-4988126

Summary

Dr. Hamdouchi earned his bachelor’s degree and doctorate in organic chemistry from Louis Pasteur University, Strasbourg-France.
Following two postdoctoral fellowships, sponsored by the National Science Foundation-USA and Ministerio de Educación y Ciencia-Spain, he joined Eli Lilly and Company in 1995.
Throughout his 20 years of career at Lilly, he has contributed to a sustainable drug discovery portfolio from preclinical hypothesis to clinical proof-of-concept that spans the oncology, neuroscience and endocrinology therapeutic areas. He has led multidisciplinary (chemistry, pharmacology, ADMET, PK, medical) scientific teams in USA, Europe and Asia to deliver a number of compounds that achieved first human dose.
He is a co-inventor of six innovative molecules being pursued in clinical development for the treatment of Diabetes, Cancer and Neurodegenerative Diseases.
He has an extensive patent and publication record and deep experience in conducting drug discovery and development in Asia through effective partnership and mentorship.

SEE AT…………ONE ORGANIC CHEMIST ONE DAY BLOG

LINK……http://oneorganichemistoneday.blogspot.in/2016/03/chafiq-hamdouchi-senior-research.html

Patent ID Date Patent Title
US8431706 2013-04-30 1,2,3,4-tetrahydroqinoline derivative useful for the treatment of diabetes

References

GPR40 agonists for the treatment of type 2 diabetes: From the laboratory to the patient
251st Am Chem Soc (ACS) Natl Meet (March 13-17, San Diego) 2016, Abst MEDI 260

//////Phase 1, LY2922470, LY 2922470, Eli Lilly, Type 2 diabetes mellitus, 1423018-12-5, Chafiq Hamdouchi

 

CC#CC(CC(=O)O)C1=CC=C(C=C1)OCC2=CC=C(S2)CN3CCCC4=C3C(=CC=C4)OC

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

 

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, amcrasto@gmail.com, +91 9323115463 India.

I , Dr A.M.Crasto is writing this blog to share the knowledge/views, after reading Scientific Journals/Articles/News Articles/Wikipedia. My views/comments are based on the results /conclusions by the authors(researchers). I do mention either the link or reference of the article(s) in my blog and hope those interested can read for details. I am briefly summarising the remarks or conclusions of the authors (researchers). If one believe that their intellectual property right /copyright is infringed by any content on this blog, please contact or leave message at below email address amcrasto@gmail.com. It will be removed ASAP

Share
Follow

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

Join other followers: