Quantcast
Channel: Preclinical drugs – New Drug Approvals
Viewing all 87 articles
Browse latest View live

JNJ-54257099

$
0
0

STR1

 

 

 

Abstract Image

JNJ-54257099,

1-((2R,4aR,6R,7R,7aR)-2-Isopropoxy-2-oxidodihydro-4H,6H-spiro[furo[3,2-d][1,3,2]dioxaphosphinine-7,2′-oxetan]-6-yl)pyrimidine-2,4(1H,3H)-dione

MW 374.28, C14 H19 N2 O8 P

CAS 1491140-67-0

2,4(1H,3H)-Pyrimidinedione, 1-[(2R,2′R,4aR,6R,7aR)-dihydro-2-(1-methylethoxy)-2-oxidospiro[4H-furo[3,2-d]-1,3,2-dioxaphosphorin-7(6H),2′-oxetan]-6-yl]-

1-((2R,4aR,6R,7R,7aR)-2-Isopropoxy-2-oxidodihydro-4H,6H-spiro[furo[3,2-d][1,3,2]dioxaphos-phinine-7,2′-oxetan]-6-yl)pyrimidine-2,4(1H,3H)-dione

Janssen R&D Ireland INNOVATOR

Ioannis Nicolaos Houpis, Tim Hugo Maria Jonckers, Pierre Jean-Marie Bernard Raboisson, Abdellah Tahri

 

 

 

STR1

Tim Hugo Maria Jonckers

 

Tim Jonckers was born in Antwerp in 1974. He studied Chemistry at the University of Antwerp and obtained his Ph.D. in organic chemistry in 2002. His Ph.D. work covered the synthesis of new necryptolepine derivatives which have potential antimalarial activity. Currently he works as a Senior Scientist at Tibotec, a pharmaceutical research and development company based in Mechelen, Belgium, that focuses on viral diseases mainly AIDS and hepatitis. The company was acquired by Johnson & Johnson in April 2002 and recently gained FDA approval for its HIV-protease inhibitor PREZISTA™.

Abdellah TAHRI

Principal Scientist at Janssen, Pharmaceutical Companies of Johnson and Johnson

 

 

Pierre Raboisson

Pierre Raboisson

PhD, Pharm.D
Head of Medicinal Chemistry

DATA

Chiral SFC using the methods described(Method 1, Rt= 5.12 min, >99%; Method 2, Rt = 7.95 min, >99%).

1H NMR (400 MHz, chloroform-d) δ ppm 1.45 (dd, J = 7.53, 6.27 Hz, 6 H), 2.65–2.84 (m, 2 H), 3.98 (td, J = 10.29, 4.77 Hz, 1 H), 4.27 (t,J = 9.66 Hz, 1 H), 4.43 (ddd, J = 8.91, 5.77, 5.65 Hz, 1 H), 4.49–4.61 (m, 1 H), 4.65 (td, J = 7.78, 5.77 Hz, 1 H), 4.73 (d, J = 7.78 Hz, 1 H), 4.87 (dq, J = 12.74, 6.30 Hz, 1 H), 5.55 (br. s., 1 H), 5.82 (d, J = 8.03 Hz, 1 H), 7.20 (d, J = 8.03 Hz, 1 H), 8.78 (br. s., 1 H);

31P NMR (chloroform-d) δ ppm −7.13. LC-MS: 375 (M + H)+.

 

HCV is a single stranded, positive-sense R A virus belonging to the Flaviviridae family of viruses in the hepacivirus genus. The NS5B region of the RNA polygene encodes a RNA dependent RNA polymerase (RdRp), which is essential to viral replication. Following the initial acute infection, a majority of infected individuals develop chronic hepatitis because HCV replicates preferentially in hepatocytes but is not directly cytopathic. In particular, the lack of a vigorous T-lymphocyte response and the high propensity of the virus to mutate appear to promote a high rate of chronic infection. Chronic hepatitis can progress to liver fibrosis, leading to cirrhosis, end-stage liver disease, and HCC (hepatocellular carcinoma), making it the leading cause of liver transplantations. There are six major HCV genotypes and more than 50 subtypes, which are differently distributed geographically. HCV genotype 1 is the predominant genotype in Europe and in the US. The extensive genetic heterogeneity of HCV has important diagnostic and clinical implications, perhaps explaining difficulties in vaccine development and the lack of response to current therapy.

Transmission of HCV can occur through contact with contaminated blood or blood products, for example following blood transfusion or intravenous drug use. The introduction of diagnostic tests used in blood screening has led to a downward trend in post-transfusion HCV incidence. However, given the slow progression to the end-stage liver disease, the existing infections will continue to present a serious medical and economic burden for decades.

Therapy possibilities have extended towards the combination of a HCV protease inhibitor (e.g. Telaprevir or boceprevir) and (pegylated) interferon-alpha (IFN-a) / ribavirin. This combination therapy has significant side effects and is poorly tolerated in many patients. Major side effects include influenza-like symptoms, hematologic

abnormalities, and neuropsychiatric symptoms. Hence there is a need for more effective, convenient and better-tolerated treatments.

The NS5B RdRp is essential for replication of the single-stranded, positive sense, HCV RNA genome. This enzyme has elicited significant interest among medicinal chemists. Both nucleoside and non-nucleoside inhibitors of NS5B are known. Nucleoside inhibitors can act as a chain terminator or as a competitive inhibitor, or as both. In order to be active, nucleoside inhibitors have to be taken up by the cell and converted in vivo to a triphosphate. This conversion to the triphosphate is commonly mediated by cellular kinases, which imparts additional structural requirements on a potential nucleoside polymerase inhibitor. In addition this limits the direct evaluation of nucleosides as inhibitors of HCV replication to cell-based assays capable of in situ phosphorylation.

Several attempts have been made to develop nucleosides as inhibitors of HCV RdRp, but while a handful of compounds have progressed into clinical development, none have proceeded to registration. Amongst the problems which HCV-targeted

nucleosides have encountered to date are toxicity, mutagenicity, lack of selectivity, poor efficacy, poor bioavailability, sub-optimal dosage regimes and ensuing high pill burden and cost of goods.

Spirooxetane nucleosides, in particular l-(8-hydroxy-7-(hydroxy- methyl)- 1,6-dioxaspiro[3.4]octan-5-yl)pyrimidine-2,4-dione derivatives and their use as HCV inhibitors are known from WO2010/130726, and WO2012/062869, including

CAS-1375074-52-4.

There is a need for HCV inhibitors that may overcome at least one of the disadvantages of current HCV therapy such as side effects, limited efficacy, the emerging of resistance, and compliance failures, or improve the sustained viral response.

The present invention concerns HCV-inhibiting uracyl spirooxetane derivatives with useful properties regarding one or more of the following parameters: antiviral efficacy towards at least one of the following genotypes la, lb, 2a, 2b, 3,4 and 6, favorable

profile of resistance development, lack of toxicity and genotoxicity, favorable pharmacokinetics and pharmacodynamics and ease of formulation and administration.

Such an HCV-inhibiting uracyl spirooxetane derivative is a compound with formula I

including any pharmaceutically acceptable salt or solvate thereof.

PATENT

WO 2015077966

https://www.google.com/patents/WO2015077966A1?cl=en

Synthesis of compound (I)

(5) (6a)

Synthesis of compound (6a)

A solution of isopropyl alcohol (3.86 mL,0.05mol) and triethylamine (6.983 mL, 0.05mol) in dichloromethane (50 mL) was added to a stirred solution of POCI3 (5)

(5.0 mL, 0.055 lmol) in DCM (50 mL) dropwise over a period of 25 min at -5°C. After the mixture stirred for lh, the solvent was evaporated, and the residue was suspended in ether (100 mL). The triethylamine hydrochloride salt was filtered and washed with ether (20 mL). The filtrate was concentrated, and the residue was distilled to give the (6) as a colorless liquid (6.1g, 69 %yield).

Synthesis of compound (4):

CAS 1255860-33-3 is dissolved in pyridine and 1,3-dichloro-l, 1,3,3-tetraisopropyldisiloxane is added. The reaction is stirred at room temperature until complete. The solvent is removed and the product redissolved in CH2CI2 and washed with saturated NaHC03 solution. Drying on MgSC^ and removal of the solvent gives compound (2). Compound (3) is prepared by reacting compound (2) with p-methoxybenzylchloride in the presence of DBU as the base in CH3CN. Compound (4) is prepared by cleavage of the bis-silyl protecting group in compound (3) using TBAF as the fluoride source.

Synthesis of compound (7a)

To a stirred suspension of (4) (2.0 g, 5.13 mmol) in dichloromethane (50 mL) was added triethylamine (2.07 g, 20.46 mmol) at room temperature. The reaction mixture was cooled to -20°C, and then (6a) (1.2 g, 6.78mmol) was added dropwise over a period of lOmin. The mixture was stirred at this temperature for 15min and then NMI was added (0.84 g, 10.23 mmol), dropwise over a period of 15 min. The mixture was stirred at -15°C for lh and then slowly warmed to room temperature in 20 h. The solvent was evaporated, the mixture was concentrated and purified by column chromatography using petroleum ether/EtOAc (10: 1 to 5: 1 as a gradient) to give (7a) as white solid (0.8 g, 32 % yield).

Synthesis of compound (I)

To a solution of (7a) in CH3CN (30 mL) and H20 (7 mL) was add CAN portion wise below 20° C. The mixture was stirred at 15-20° C for 5h under N2. Na2S03 (370 mL) was added dropwise into the reaction mixture below 15°C, and then Na2C03 (370 mL) was added. The mixture was filtered and the filtrate was extracted with CH2C12

(100 mL*3). The organic layer was dried and concentrated to give the residue. The residue was purified by column chromatography to give the target compound (8a) as white solid. (Yield: 55%)

1H NMR (400 MHz, CHLOROFORM- ) δ ppm 1.45 (dd, J=7.53, 6.27 Hz, 6 H), 2.65 -2.84 (m, 2 H), 3.98 (td, J=10.29, 4.77 Hz, 1 H), 4.27 (t, J=9.66 Hz, 1 H), 4.43 (ddd, J=8.91, 5.77, 5.65 Hz, 1 H), 4.49 – 4.61 (m, 1 H), 4.65 (td, J=7.78, 5.77 Hz, 1 H), 4.73 (d, J=7.78 Hz, 1 H), 4.87 (dq, J=12.74, 6.30 Hz, 1 H), 5.55 (br. s., 1 H), 5.82 (d, J=8.03 Hz, 1 H), 7.20 (d, J=8.03 Hz, 1 H), 8.78 (br. s., 1 H); 31P NMR (CHLOROFORM-^) δ ppm -7.13; LC-MS: 375 (M+l)+

 

PATENT

https://www.google.co.in/patents/WO2013174962A1?cl=en

The starting material l-[(4R,5R,7R,8R)-8-hydroxy-7-(hydroxymethyl)-l,6-dioxa- spiro[3.4]octan-5-yl]pyrimidine-2,4(lH,3H)-dione (1) can be prepared as exemplified in WO2010/130726. Compound (1) is converted into compounds of the present invention via a p-methoxybenzyl protected derivative (4) as exemplified in the following Scheme 1. cheme 1

Figure imgf000011_0001

Examples

Scheme 2

Synthesis of compound (8a)

Figure imgf000015_0001

Synthesis of compound (2)

Compound (2) can be prepared by dissolving compound (1) in pyridine and adding l,3-dichloro-l,l,3,3-tetraisopropyldisiloxane. The reaction is stirred at room temperature until complete. The solvent is removed and the product redissolved in CH2CI2and washed with saturated NaHC03 solution. Drying on MgSC^ and removal of the solvent gives compound (2).

Synthesis of compound (3)

Compound (3) is prepared by reacting compound (2) with p-methoxybenzylchloride in the presence of DBU as the base in CH3CN.

Synthesis of compound (4)

Compound (4) is prepared by cleavage of the bis-silyl protecting group in compound (3) using TBAF as the fluoride source.

Synthesis of compound (6a)

A solution of isopropyl alcohol (3.86 mL,0.05mol) and triethylamine (6.983 mL, 0.05mol) in dichloromethane (50 mL) was added to a stirred solution of POCl3 (5) (5.0 mL, 0.055 lmol) in DCM (50 mL) dropwise over a period of 25 min at -5°C. After the mixture stirred for lh, the solvent was evaporated, and the residue was suspended in ether (100 mL). The triethylamine hydrochloride salt was filtered and washed with ether (20 mL). The filtrate was concentrated, and the residue was distilled to give the (6) as a colorless liquid (6.1g, 69 %yield).

Synthesis of compound (7a)

To a stirred suspension of (4) (2.0 g, 5.13 mmol) in dichloromethane (50 mL) was added triethylamine (2.07 g, 20.46 mmol) at room temperature. The reaction mixture was cooled to -20°C, and then (6a) (1.2 g, 6.78mmol) was added dropwise over a period of lOmin. The mixture was stirred at this temperature for 15min and then NMI was added (0.84 g, 10.23 mmol), dropwise over a period of 15 min. The mixture was stirred at -15°C for lh and then slowly warmed to room temperature in 20 h. The solvent was evaporated, the mixture was concentrated and purified by column chromatography using petroleum ether/EtOAc (10:1 to 5: 1 as a gradient) to give (7a) as white solid (0.8 g, 32 % yield).

Synthesis of compound (8a)

To a solution of (7a) in CH3CN (30 mL) and H20 (7 mL) was add CAN portion wise below 20°C. The mixture was stirred at 15-20°C for 5h under N2. Na2S03 (370 mL) was added dropwise into the reaction mixture below 15°C, and then Na2C03 (370 mL) was added. The mixture was filtered and the filtrate was extracted with CH2C12

(100 mL*3). The organic layer was dried and concentrated to give the residue. The residue was purified by column chromatography to give the target compound (8a) as white solid. (Yield: 55%)

1H NMR (400 MHz, CHLOROFORM- ) δ ppm 1.45 (dd, J=7.53, 6.27 Hz, 6 H), 2.65 – 2.84 (m, 2 H), 3.98 (td, J=10.29, 4.77 Hz, 1 H), 4.27 (t, J=9.66 Hz, 1 H), 4.43 (ddd, J=8.91, 5.77, 5.65 Hz, 1 H), 4.49 – 4.61 (m, 1 H), 4.65 (td, J=7.78, 5.77 Hz, 1 H), 4.73 (d, J=7.78 Hz, 1 H), 4.87 (dq, J=12.74, 6.30 Hz, 1 H), 5.55 (br. s., 1 H), 5.82 (d, J=8.03 Hz, 1 H), 7.20 (d, J=8.03 Hz, 1 H), 8.78 (br. s., 1 H); 31P NMR (CHLOROFORM-^) δ ppm -7.13; LC-MS: 375 (M+l)+ Scheme 3

Synthesis of compound (VI)

Figure imgf000017_0001

Step 1: Synthesis of compound (9)Compound (1), CAS 1255860-33-3 ( 1200 mg, 4.33 mmol ) and l,8-bis(dimethyl- amino)naphthalene (3707 mg, 17.3 mmol) were dissolved in 24.3 mL of

trimethylphosphate. The solution was cooled to 0°C. Compound (5) (1.21 mL, 12.98 mmol) was added, and the mixture was stirred well maintaining the temperature at 0°C for 5 hours. The reaction was quenched by addition of 120 mL of tetraethyl- ammonium bromide solution (1M) and extracted with CH2CI2 (2×80 mL). Purification was done by preparative HPLC (Stationary phase: RP XBridge Prep CI 8 ΟΒϋ-10μιη, 30x150mm, mobile phase: 0.25% NH4HCO3 solution in water, CH3CN) , yielding two fractions. The purest fraction was dissolved in water (15 mL) and passed through a manually packed Dowex (H+) column by elution with water. The end of the elution was determined by checking UV absorbance of eluting fractions. Combined fractions were frozen at -78°C and lyophilized. Compound (9) was obtained as a white fluffy solid (303 mg, (0.86 mmol, 20%> yield), which was used immediately in the following reaction. Step 2: Preparation of compound (VI)

Compound (9) (303 mg, 0.86 mmol) was dissolved in 8 mL water and to this solution was added N . N’- D ic y c ! he y !-4- mo rph line carboxamidine (253.8 mg, 0.86 mmol) dissolved in pyridine (8.4 mi.). The mixture was kept for 5 minutes and then

evaporated to dryness, dried overnight in vacuo overnight at 37°C. The residu was dissolved in pyridine (80 mL). This solution was added dropwise to vigorously stirred DCC (892.6 mg, 4.326 mmol) in pyridine (80 mL) at reflux temperature. The solution was kept refluxing for 1.5h during which some turbidity was observed in the solution. The reaction mixture was cooled and evaporated to dryness. Diethylether (50 mL) and water (50 mL) were added to the solid residu. N’N-dicyclohexylurea was filtered off, and the aqueous fraction was purified by preparative HPLC (Stationary phase: RP XBridge Prep C18 OBD-ΙΟμιη, 30x150mm, mobile phase: 0.25% NH4HCO3 solution in water, CH3CN) , yielding a white solid which was dried overnight in vacuo at 38°C. (185 mg, 0.56 mmol, 65% yield). LC-MS: (M+H)+: 333.

1H NMR (400 MHz, DMSO-d6) d ppm 2.44 – 2.59 (m, 2 H) signal falls under DMSO signal, 3.51 (td, J=9.90, 5.50 Hz, 1 H), 3.95 – 4.11 (m, 2 H), 4.16 (d, J=10.34 Hz, 1 H), 4.25 – 4.40 (m, 2 H), 5.65 (d, J=8.14 Hz, 1 H), 5.93 (br. s., 1 H), 7.46 (d, J=7.92 Hz, 1 H), 2H’s not observed

Paper

http://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.6b00382,

Discovery of 1-((2R,4aR,6R,7R,7aR)-2-Isopropoxy-2-oxidodihydro-4H,6H-spiro[furo[3,2-d][1,3,2]dioxaphosphinine-7,2′-oxetan]-6-yl)pyrimidine-2,4(1H,3H)-dione (JNJ-54257099), a 3′-5′-Cyclic Phosphate Ester Prodrug of 2′-Deoxy-2′-Spirooxetane Uridine Triphosphate Useful for HCV Inhibition

Janssen Infectious Diseases − Diagnostics BVBA, Turnhoutseweg 30, 2340 Beerse, Belgium
J. Med. Chem., Article ASAP
DOI: 10.1021/acs.jmedchem.6b00382
Publication Date (Web): May 14, 2016
Copyright © 2016 American Chemical Society
*Phone: +32 014601168. E-mail: tjoncker@its.jnj.com.

JNJ-54257099 (9) is a novel cyclic phosphate ester derivative that belongs to the class of 2′-deoxy-2′-spirooxetane uridine nucleotide prodrugs which are known as inhibitors of the HCV NS5B RNA-dependent RNA polymerase (RdRp). In the Huh-7 HCV genotype (GT) 1b replicon-containing cell line 9 is devoid of any anti-HCV activity, an observation attributable to inefficient prodrug metabolism which was found to be CYP3A4-dependent. In contrast, in vitro incubation of 9 in primary human hepatocytes as well as pharmacokinetic evaluation thereof in different preclinical species reveals the formation of substantial levels of 2′-deoxy-2′-spirooxetane uridine triphosphate (8), a potent inhibitor of the HCV NS5B polymerase. Overall, it was found that 9 displays a superior profile compared to its phosphoramidate prodrug analogues (e.g., 4) described previously. Of particular interest is the in vivo dose dependent reduction of HCV RNA observed in HCV infected (GT1a and GT3a) human hepatocyte chimeric mice after 7 days of oral administration of 9

////////////JNJ-54257099, 1491140-67-0, JNJ54257099, JNJ 54257099

O=C(C=C1)NC(N1[C@H]2[C@]3(OCC3)[C@H](O4)[C@@H](CO[P@@]4(OC(C)C)=O)O2)=O


Filed under: Preclinical drugs Tagged: 1491140-67-0, JNJ-54257099, JNJ54257099

PF-05388169

$
0
0

str1

PF-05388169

CAS 1604034-78-7,  MF C22 H21 N3 O4

MW 391.42

11H-Indolo[3,2-c]quinoline-9-carbonitrile, 2-methoxy-3-[2-(2-methoxyethoxy)ethoxy]-
IRAK4 inhibitor

Rheumatoid arthritis;
SLE

Preclinical

str1

PAPER

Bioorganic & Medicinal Chemistry Letters (2014), 24(9), 2066-2072.

http://www.sciencedirect.com/science/article/pii/S0960894X14002832

Identification and optimization of indolo[2,3-c]quinoline inhibitors of IRAK4

  • a Pfizer Global R&D, 445 Eastern Point Rd., Groton, CT 06340, USA
  • b Pfizer Global R&D, 200 Cambridge Park Dr., Cambridge, MA 02140, USA
  • c Pfizer Global R&D, 87 Cambridgepark Dr., Cambridge, MA 02140, USA
  • d Pfizer Global R&D, 1 Burtt Rd., Andover, MA 01810, USA

Image for unlabelled figure

IRAK4 is responsible for initiating signaling from Toll-like receptors (TLRs) and members of the IL-1/18 receptor family. Kinase-inactive knock-ins and targeted deletions of IRAK4 in mice cause reductions in TLR induced pro-inflammatory cytokines and these mice are resistant to various models of arthritis. Herein we report the identification and optimization of a series of potent IRAK4 inhibitors. Representative examples from this series showed excellent selectivity over a panel of kinases, including the kinases known to play a role in TLR-mediated signaling. The compounds exhibited low nM potency in LPS- and R848-induced cytokine assays indicating that they are blocking the TLR signaling pathway. A key compound (26) from this series was profiled in more detail and found to have an excellent pharmaceutical profile as measured by predictive assays such as microsomal stability, TPSA, solubility, and c log P. However, this compound was found to afford poor exposure in mouse upon IP or IV administration. We found that removal of the ionizable solubilizing group (32) led to increased exposure, presumably due to increased permeability. Compounds 26 and 32, when dosed to plasma levels corresponding to ex vivo whole blood potency, were shown to inhibit LPS-induced TNFα in an in vivo murine model. To our knowledge, this is the first published in vivo demonstration that inhibition of the IRAK4 pathway by a small molecule can recapitulate the phenotype of IRAK4 knockout mice.

SYNTHESIS

STR1

//////////PF-05388169, TLR signaling, Indoloquinoline, IRAK4, Kinase inhibitor, Inflammation, PRECLINICAL, 1604034-78-7

C(COC)OCCOc4c(cc3\C2=N\c1cc(ccc1/C2=C/Nc3c4)C#N)OC


Filed under: Preclinical drugs Tagged: 1604034-78-7, Indoloquinoline, inflammation, IRAK4, kinase inhibitor, PF-05388169, preclinical, TLR signaling

PF-05387252

$
0
0

str1

PF-05387252

CAS  1604034-71-0

C25H27N5O2
MW 429.51418 g/mol

2-methoxy-3-[3-(4-methylpiperazin-1-yl)propoxy]-11H-indolo[3,2-c]quinoline-9-carbonitrile

IRAK4 inhibitor

Rheumatoid arthritis;
SLE

Preclinical

In the past decade there has been considerable interest in targeting the innate immune system in the treatment of autoimmune diseases and sterile inflammation. Receptors of the innate immune system provide the first line of defense against bacterial and viral insults. These receptors recognize bacterial and viral products as well as pro-inflammatory cytokines and thereby initiate a signaling cascade that ultimately results in the up-regulation of inflammatory cytokines such as TNFα, IL6, and interferons. Recently it has become apparent that self-generated ligands such as nucleic acids and products of inflammation such as HMGB1 and Advanced Glycated End-products (AGE) are ligands for Toll-like receptors (TLRs) which are key receptors of the innate immune system.

This demonstrates the role of TLRs in the initiation and perpetuation of inflammation due to autoimmunity.

Interleukin-1 receptor associated kinase (IRAK4) is a ubiquitously expressed serine/threonine kinase involved in the regulation of innate immunity. IRAK4 is responsible for initiating signaling from TLRs and members of the IL-1/18 receptor family. Kinase-inactive knock-ins and targeted deletions of IRAK4 in mice lead to reductions in TLR and IL-1 induced pro-inflammatory cytokines. and 7 IRAK-4 kinase-dead knock-in mice have been shown to be resistant to induced joint inflammation in the antigen-induced-arthritis (AIA) and serum transfer-induced (K/BxN) arthritis models. Likewise, humans deficient in IRAK4 also display the inability to respond to challenge by TLR ligands and IL-1

 However, the immunodeficient phenotype of IRAK4-null individuals is narrowly restricted to challenge by gram positive bacteria, but not gram negative bacteria, viruses or fungi. This gram positive sensitivity also lessens with age implying redundant or compensatory mechanisms for innate immunity in the absence of IRAK4.These data suggest that inhibitors of IRAK4 kinase activity will have therapeutic value in treating cytokine driven autoimmune diseases while having minimal immunosuppressive side effects. Additional recent studies suggest that targeting IRAK4 may be a viable strategy for the treatment of other inflammatory pathologies such as atherosclerosis.

Indeed, the therapeutic potential of IRAK4 inhibitors has been recognized by others within the drug-discovery community as evidenced by the variety of IRAK4 inhibitors have been reported to-date.12, 13, 14, 15 and 16 However, limited data has been published about these compounds and they appear to suffer from a variety of issues such as poor kinase selectivity and poor whole-blood potency that preclude their advancement into the pre-clinical models. To the best of our knowledge, no in vivo studies of IRAK4 inhibitors have been reported to-date in the literature. Herein we report a new class of IRAK4 inhibitors that are shown to recapitulate the phenotype observed in IRAK4 knockout and kinase-dead mice.

PAPER

Bioorganic & Medicinal Chemistry Letters (2014), 24(9), 2066-2072.

doi:10.1016/j.bmcl.2014.03.056

http://www.sciencedirect.com/science/article/pii/S0960894X14002832

Identification and optimization of indolo[2,3-c]quinoline inhibitors of IRAK4

  • a Pfizer Global R&D, 445 Eastern Point Rd., Groton, CT 06340, USA
  • b Pfizer Global R&D, 200 Cambridge Park Dr., Cambridge, MA 02140, USA
  • c Pfizer Global R&D, 87 Cambridgepark Dr., Cambridge, MA 02140, USA
  • d Pfizer Global R&D, 1 Burtt Rd., Andover, MA 01810, USA

Image for unlabelled figure

Abstract

IRAK4 is responsible for initiating signaling from Toll-like receptors (TLRs) and members of the IL-1/18 receptor family. Kinase-inactive knock-ins and targeted deletions of IRAK4 in mice cause reductions in TLR induced pro-inflammatory cytokines and these mice are resistant to various models of arthritis. Herein we report the identification and optimization of a series of potent IRAK4 inhibitors. Representative examples from this series showed excellent selectivity over a panel of kinases, including the kinases known to play a role in TLR-mediated signaling. The compounds exhibited low nM potency in LPS- and R848-induced cytokine assays indicating that they are blocking the TLR signaling pathway. A key compound (26) from this series was profiled in more detail and found to have an excellent pharmaceutical profile as measured by predictive assays such as microsomal stability, TPSA, solubility, and c log P. However, this compound was found to afford poor exposure in mouse upon IP or IV administration. We found that removal of the ionizable solubilizing group (32) led to increased exposure, presumably due to increased permeability. Compounds 26 and 32, when dosed to plasma levels corresponding to ex vivo whole blood potency, were shown to inhibit LPS-induced TNFα in an in vivo murine model. To our knowledge, this is the first published in vivo demonstration that inhibition of the IRAK4 pathway by a small molecule can recapitulate the phenotype of IRAK4 knockout mice.

CID 50992153.png

SYNTHESIS

STR1

////////PF-05387252,  1604034-71-0, PF 05387252, TLR signaling, Indoloquinoline, IRAK4, Kinase inhibitor, Inflammation, PRECLINICAL

N1(CCN(CC1)CCCOc3c(cc2c4nc5cc(ccc5c4cnc2c3)C#N)OC)C

OR

CN1CCN(CC1)CCCOC2=C(C=C3C(=C2)N=CC4=C3NC5=C4C=CC(=C5)C#N)OC


Filed under: Preclinical drugs Tagged: 1604034-71-0, Indoloquinoline, inflammation, IRAK4, kinase inhibitor, PF-05387252, preclinical, TLR signaling

Novel, isoform-selective inhibitor of histone deacetylase 8 (HDAC8)

$
0
0

str1

CAS 1620779-53-4
MF C22H20N4O2, MW 372.4

(S)-2-(5-(cyclopropylethynyl)-4-phenyl-1H-1,2,3-triazol-1-yl)-N-hydroxy-3-phenylpropanamide

1H-1,2,3-Triazole-1-acetamide, 5-(2-cyclopropylethynyl)-N-hydroxy-4-phenyl-α-(phenylmethyl)-, (αS)-

Applicants: TRUSTEES OF BOSTON UNIVERSITY
DANA-FARBER CANCER INSTITUTE, INC.
Inventors: Aaron Beaty BEELER
John A. PORCO, JR.
Oscar J. INGHAM
James E. BRADNER
As histone proteins bind DNA prior to transcription, their biochemical action plays a critical role in the regulation of gene expression and cellular differentiation. Histone deacetylases (HDACs) are an important family of proteins predominantly responsible for specific posttranslational modifications of histone proteins, the chief organizational component of chromatin. HDACs catalyze the removal of acetyl groups from histones and other cellular proteins. HDAC-mediated deacetylation of chromatin-bound histones regulates the expression of a variety of genes throughout the genome. Importantly, HDACs have been linked to cancer, as well as other health conditions. To date, eleven major HDAC isoforms have been described (HDACs 1-11). HDACs are categorized into two classes. Class I HDACs include HDAC1, HDAC2, HDAC3, HDAC8 and HDAC11. Class II HDACs include HDAC4, HDAC5, HDAC6, HDAC7, HDAC9 and HDAC10. HDAC’s are validated targets for a number of disease states, including cancer, neurodegenerative diseases, sickle-cell anemia, muscular dystrophy, and HIV. There are currently two HDAC inhibitors on the market, Vorniostat and Romidepsin. Both are approved for treatment of T-cell lymphoma. However, they are both pan active inhibitors showing very little specificity of binding to HDAC subclasses. Because of this lack of specificity they have a number of side effects.
      Non-selective HDAC inhibitors effect deacetylase activity of most, if not all, of the HDACs. The mechanisms of the anticancer effects of SAHA, a non-selective HDAC inhibitor, are not completely understood, and likely result from both altered gene expression and altered function of proteins regulating cell proliferation and cell death pathways. Non-selective HDAC inhibitors, such as SAHA, induce the accumulation of acetylated histone proteins and non histone proteins.
    Small molecule HDAC inhibitors that are isoform-selective are useful as therapeutic agents with reduced toxicity and as tools for probing the biology of the HDAC isoforms. The present disclosure is related, in part to small molecules that are selective HDAC inhibitors.

1H NMR (500 MHz, d4-MeOD) 0.80 (2H, m), 0.98 (2H, m), 1.47 (1H, m), 3.51 (1H, dd, J = 11.2, 14.2 Hz), 3.71 (1H, dd, J = 3.9, 14.2 Hz), 5.49 (1H, dd, J = 3.9, 11.2 Hz), 6.96 (2H, m), 7.17-7.20 (3H, m), 7.37 (1H, t, J = 7.3 Hz), 7.43 (2H, t, J = 7.3 Hz), 7.99 (2H, d, J = 8.8 Hz);

13C NMR (100 MHz, d4-MeOD) 0.02, 8.55, 37.07, 60.83, 62.59, 109.09, 118.98, 125.9, 127.16, 128.55, 128.65, 128.71, 129.16, 130.07, 136.09, 147.10, 165.20;

HRMS calculated for C22H21N4O2 + (M+H): 373.1659, found: 373.1665.

PATENT

WO2014116962

https://www.google.com/patents/WO2014116962A1?cl=en

SAR. libraries were synthesized to investigate substitution about the triazole core. In some examples, compounds were synthesized using the synthetic routes shown in Fig. 2.

In one study, compound
was synthesized as outline in Scheme I.

Scheme I

PATENT

US153441899

https://patentscope.wipo.int/search/en/detail.jsf?docId=US153441899&recNum=1&office=&queryString=FP%3A%28Aaron+Beeler%29&prevFilter=&sortOption=Pub+Date+Desc&maxRec=8

SAR libraries were synthesized to investigate substitution about the triazole core. In some examples, compounds were synthesized using the synthetic routes shown in FIG. 2. In one study, compound

 was synthesized as outline in Scheme I.

The HDAC assays were carried out as described in Bowers A, West N, Taunton J, Schreiber S L, Bradner J E, Williams R M Total Synthesis and Biological Mode of Action of Largazole: A Potent Class I Histone Deacetylase Inhibitor. J. Am. Chem. Soc. 2008, 130, 11219-11222. Assay results revealed that among the analogues tested a cyclopropane analog was the most active at 0.4 nM (>1000 fold selectivity). These results demonstrated that a small aliphatic group in the 5-position on the triazole can increase potency. Also, compounds with an L-phenylalanine moiety at the 3-position showed significant potency. To expand our understanding of how the molecule interacts with the binding pocket of HDAC 8 and to understand our preliminary SAR, molecular modeling was carried out. The phenyl group from the original amino methyl ester fits snuggly into the Zn binding site and the alkynyl phenyl group sits flat in a hydrophobic groove. In summary, the inventors have developed a potent and highly selective small molecule which inhibits HDAC-8 at approximately 500 pM with over 1000-fold selectivity over HDAC-6 and significantly greater selectivity for all other HDACs. To inventors’ knowledge, to date there are no compounds with this level of potency and selectivity.
All patents and other publications identified in the specification and examples are expressly incorporated herein by reference for all purposes. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents.
Although preferred embodiments have been depicted and described in detail herein, it will be apparent to those skilled in the relevant art that various modifications, additions, substitutions, and the like can be made without departing from the spirit of the invention and these are therefore considered to be within the scope of the invention as defined in the claims which follow. Further, to the extent not already indicated, it will be understood by those of ordinary skill in the art that any one of the various embodiments herein described and illustrated can be further modified to incorporate features shown in any of the other embodiments disclosed herein.

Paper

Abstract Image

A novel, isoform-selective inhibitor of histone deacetylase 8 (HDAC8) has been discovered by the repurposing of a diverse compound collection. Medicinal chemistry optimization led to the identification of a highly potent (0.8 nM) and selective inhibitor of HDAC8.

Development of a Potent and Selective HDAC8 Inhibitor

Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts 02215, United States
§ Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, United States
ACS Med. Chem. Lett., Article ASAP
DOI: 10.1021/acsmedchemlett.6b00239
*Tel: 617-358-3487. E-mail: beelera@bu.edu.

http://pubs.acs.org/doi/abs/10.1021/acsmedchemlett.6b00239

file:///C:/Users/Inspiron/Downloads/ml6b00239_si_001.pdf

 

Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States

Image result for Department of Chemistry and Center for Molecular Discovery (BY-CMD),

 

Image result for Department of Chemistry and Center for Molecular Discovery (BY-CMD),Center for Molecular Discovery (CMD) Director John Porco and members of the CMD lab team.

 

 

Image result for Aaron B. Beeler

Aaron Beeler

prof-beeler

Aaron Beeler received his Ph.D. in 2002 from Professor John Rimoldi’s laboratory in the Department of Medicinal Chemistry at the University of Mississippi. He then joined the Porco group as a postodoctoral fellow and subsequently the Center for Chemical Methodology and Library Development at Boston University, now the Center for Molecular Discovery. He was promoted to Assistant Director of the CMLD-BU in January 2005. In 2012 Aaron joined the Department of Chemistry as a tenure-track professor in medicinal chemistry.

Degrees and Positions

  • B.S. Belmont University, Biology,
  • Ph.D. University of Mississippi, Medicinal Chemistry

Research

The Beeler Research Group is truly multidisciplinary, combining organic chemistry, engineering, and biology to solve problems in medicinal chemistry. All of these elements are combined and directed toward significant problems in human health. The Beeler Group is addressing focused disease areas (e.g., schizophrenia, Parkinson’s, cystic fibrosis), as well as project areas with broader impact potential (e.g., new methods for discovery of small molecules with anti-cancer properties).

  • Medicinal Chemistry: The goals of medicinal chemistry projects are to optimize small molecules in order to: a) develop a probe that may be utilized as a tool in biological studies; b) develop a lead molecule to facilitate future therapeutics; and c) utilize small molecules to enhance understanding of biological targets that are important for human health. These projects provide students with training in organic chemistry, medicinal chemistry, and focused biology. Projects are selected based on their chemistry and/or biology significance and potential for addressing challenging questions.
  • Technology: One of the core components of the research in the Beeler Group is development of technologies and paradigms that facilitate rapid modification of complex scaffolds. These technologies enable optimization of biologically active lead compounds and identification of small molecule leads in biological systems. The projects focus on utilizing automation, miniaturization, and microfluidics to carry out chemical transformations. These projects are highly interdisciplinary with both chemistry and engineering components.
  • Photochemistry: This area focuses on photochemical transformations toward the synthesis of natural products, natural product scaffolds, and other complex chemotypes of interest to medicinal chemistry and chemical biology. The foundation of these projects is utilizing microfluidics to enable photochemical reaction development.

Techniques & Resources

Students in the Beeler Research Group will have opportunities to learn a number of exciting research disciplines. Organic synthesis will be at the heart of every project. This will include targeted synthesis, methodology development, and medicinal chemistry. Through collaborations with biological researchers and/or research projects carried out within the Beeler Group, students will learn methods for biological assays, pharmacology, and target identification. Many projects will also include aspects of engineering that will provide opportunities for learning techniques such as microfabrication and microfluidics.

Opportunities

It is becoming evident that successful and impactful science is realized in collaborative interdisciplinary environments. The Beeler Research Group’s multidisciplinary nature and collaborative projects provides opportunities to learn areas of research outside of traditional chemistry.

What’s Next for Graduates of the Beeler Group?

Members of the Beeler Research Group will be positioned for a wide range of future endeavors.

  • Undergraduates will be prepared to enter into graduate school for organic chemistry, chemical biology, or chemical engineering or to start careers in industry;
  • Graduate students will have the foundation required for postdoctoral studies in organic synthesis or chemical biology as well as an industrial career in biotech or pharma;
  • Postdoctoral associates will gain training and experience critical for both academic and industrial careers.

Assistant Professor
Office: SCI 484C
Laboratory: SCI 484A
Phone: 617.358.3487
Fax: 617-358-2847
beelera@bu.edu
Office Hours: by Appointment
Beeler Group Homepage
Google Scholar Page

Oscar J. Ingham below

Image result for Oscar J. Ingham

John A. PORCO, JR  below

Image result for John A. PORCO, JRImage result for James E. BRADNER

JAMES E. BRADNER, MD  above

Dana-Farber Cancer Institute

Image result for Dana-Farber Cancer Institute

 

Image result for Dana-Farber Cancer Institute

 

Ron ParanalRon Paranal

 

 

Image result for Randolph A. EscobarRandolph A. Escobar

 

Han YuehHan Yueh

 

US20090181943 * Apr 9, 2008 Jul 16, 2009 Methylgene Inc. Inhibitors of Histone Deacetylase
Reference
1 * GERARD, B ET AL.: ‘Synthesis of 1,4,5-trisubstituted-1,2,3-triazoles by copper-catalyzed cycloaddition-coupling of azides and terminal alkynes‘ TETRAHEDRON vol. 62, 12 May 2006, pages 6405 – 6411
2 * VANNINI, A ET AL.: ‘Crystal structure of a eukaryotic zinc-dependent histone deacetylase, human HDAC8, complexed with a hydroxamic acid inhibitor.‘ PNAS, [Online] vol. 101, no. 42, 19 October 2004, pages 15064 – 15069 Retrieved from the Internet: <URL:http://www.pnas.org/content/101/42/15064&gt;

///////////epigenetic,  HDACHDAC8,  Histone deacetylase,  histone deacetylase 8,  triazole, PRECLINICAL, Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States, Oscar J. InghamAaron Beeler

n1n(c(c(n1)c2ccccc2)C#CC3CC3)C(C(=O)NO)Cc4ccccc4


Filed under: Preclinical drugs Tagged: 590 Commonwealth Avenue, Aaron Beeler, Boston, Boston University, Department of Chemistry and Center for Molecular Discovery (BU-CMD), epigenetic, HDAC, HDAC8, histone deacetylase 8, histone deacetylase., Massachusetts 02215, Oscar J. Ingham, preclinical, triazole, united states

1, 2-Bis(4-(4-4-nitrophenyl)piperazin-1-yl)ethanone for androgen sensitive prostatic disorders

$
0
0

str1

 

str1

1, 2-Bis(4-(4-4-nitrophenyl)piperazin-1-yl)ethanone

Molecular Formula: C22H26N6O5
Molecular Weight: 454.47904 g/mol

str1

CAS 330633-91-5

CDRI-?

For treatment of androgen sensitive prostatic disorders

1,2-bis[4-(4-nitrophenyl)piperazin-1-yl]ethanone.png

1, 2-Bis(4-(4-4-nitrophenyl)piperazin-1-yl)ethanone

Graphical abstract: Design, synthesis and biological profiling of aryl piperazine based scaffolds for the management of androgen sensitive prostatic disorders

In the quest for novel scaffolds for the management of androgen sensitive prostatic disorders like prostate cancer and benign prostatic hyperplasia, a series of twenty-six aryl/heteroaryl piperazine derivatives have been described. Three compounds, 8a, 8c and 9a, exhibited good activity profiles against an androgen sensitive prostate cancer cell line (LNCaP) with EC50values of 9.8, 7.6 and 11.2 μM, respectively. These compounds caused a decrease in luciferase activity and a decline in PSA and Ca2+ levels, which are indicative of their anti-androgenic and α1A-adrenergic receptor blocking activities, respectively.

Compound 9a reduced the prostate weight of rats (47%) and in pharmacokinetic analysis at 10 mg kg−1 it demonstrated an MRT of ∼14 h post dose, exhibiting high levels in prostate. Compound 9a docked in a similar orientation to hydroxyflutamide on an androgen receptor and showed strong π–π interactions. These findings reveal that compound 9a is a promising candidate for management of prostatic disorders with anti-androgenic and α1A-blocking activities.

Design, synthesis and biological profiling of aryl piperazine based scaffolds for the management of androgen sensitive prostatic disorders

*Corresponding authors
aMedicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram ext., Lucknow-226031, India
E-mail: vl_sharma@cdri.res.in, vlscdri@gmail.com
Fax: +91 522 2771941
Tel: +91 522 2772450 Ext. 4671
bEndocrinology Division, CSIR-Central Drug Research Institute, Lucknow-226031, India
cPharmacokinetics and Metabolism Division, CSIR-Central Drug Research Institute, Lucknow-226031, India
dMolecular & Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow-226031, India
eAcademy of Scientific and Innovative Research (AcSIR), New Delhi-110001, India
Med. Chem. Commun., 2016, Advance Article

DOI: 10.1039/C6MD00426A, http://pubs.rsc.org/en/Content/ArticleLanding/2016/MD/C6MD00426A?utm_source=feedburner&utm_medium=feed&utm_campaign=Feed%3A+rss%2FMD+%28RSC+-+Med.+Chem.+Commun.+latest+articles%29#!divAbstract

1, 2-Bis(4-(4-4-nitrophenyl)piperazin-1-yl)ethanone (9a) To the mixture of 8a (0.3 g, 1.06 mmol) and Et3N (0.3 mL, 2.12 mmol) in CHCl3 (5 mL) was added 1-(4-nitrophenyl)piperazine (7a, 0.320 g, 1.59 mmol) in 5 mL CHCl3 dropwise within 1 h. After complete addition reaction mixture was further stirred in an oil bath at 80-85 °C for 15 h. The reaction mixture was cooled, washed with water (5 mL × 3) and the organic layer was separated. Combined organic layer was dried (anhyd. Na2SO4 and concentrated under reduced pressure in rotavapor. The solid obtained was purified by recrystallization using EtOAc/Hexane which furnished yellow crystals (yield 81%);

mp: 156-157 °C; IR (KBr)  (cm-1): 3019, 2399, 1640, 1597, 1506, 1423, 1330;

1H NMR (400 MHz, CDCl3):  8.14-8.09 (4H, m), 6.84-6.81 (4H, m), 3.84-3.83 (4H, m), 3.49-3.44 (8H, m), 3.33 (2H, s), 2.72 (4H, t, J = 5.0 Hz);

13C NMR (75.4 MHz, CDCl3):  167.7, 154.7, 154.3, 138.8, 138.4, 125.9, 125.8, 112.9, 112.7, 60.8, 52.5, 46.9, 46.7, 44.6;

HRMS (ESI positive) m/z calcd. for C22H26N6O5 [M+H]+ : 455.2043, found: 455.2034;

Anal calcd. for C22H26N6O5: C, 58.14; H, 5.77; N, 18.49, found: C, 58.31; H, 5.92; N, 18.66.

str1

str1

SONAL GUPTA

Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram ext., Lucknow-226031, India

Image result for Medicinal & Process Chemistry Division, CSIR-Central Drug Research InstituteImage result for Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute

str1

Dr. VISHNU LAL SHARMA

http://www.cdriindia.org/VL_Sharma.htm

Dr. VISHNU LAL SHARMA

Senior Principal Scientist (CSIR-CDRI ) / Professor (AcSIR)
Lab No. CSS-SF-201, Medicinal and Process Chemistry Division
Central Drug Research Institute,
B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road
Lucknow- 226031

Educational Qualifications M.Sc (Organic Chemistry, Lucknow University, Lucknow, Uttar Pradesh, 1978)
Ph.D. (Chemistry, Lucknow University, Lucknow, Uttar Pradesh, 1985)
Date of Birth February 7th, 1958
E- Mail vl_sharma@cdri.res.in, vlscdri@gmail.com
Phone No. +91-0522-2772450/550, Ext. 4671.
Mobile No. +91-9415074195
Fax No. +91-522-2771941
Research Experience (Area) Medicinal chemistry, Organic chemistry.
Google Scholar https://scholar.google.co.in/citations?user=cAsQaiYAAAAJ&hl=en
Research gate https://www.researchgate.net/profile/Vishnu_Sharma13
 POST-DOCTORAL RESEARCH (ABROAD)
•
University of Dusseldorf, Dusseldorf, Germany, Oct., 1994 to Dec., 1994
CURRENT AREAS OF INTEREST
•
Medicinal Chemistry, Synthetic organic chemistry and Process chemistry.
•
The research focused in my group is related to design and synthesis of small molecule libraries of biomedical importance and development of new methodologies and process developments of candidate drugs.
no image
From left to right upper row: Dr. S.T.V.S. Kiran Kumar, Dr. Lalit Kumar, Dr. V.L. Sharma, Dr. Nand Lal, Dr. Amit Sarswat
Lower row: Dhanaraju Mandalapu, Sonal Gupta, Mrs. Tara Rawat (S.T.O.), Dr. Veenu bala, Dr. Santosh Jangir
THESIS SUPERVISED
•
Seven (7) students for their Ph.D.
•
Twenty two (22) students for their Post Graduation degrees
FORMER Ph.D. STUDENTS
•
Dr. S.T.V.S. Kiran Kumar, 2006,Research Scientist at University of Virginia Charlottesville, Virginia.
•
Dr. Lalit Kumar, 2011, KIMIA Biosciences Pvt.Ltd., Rajasthan, India .
•
Dr. Amit Sarswat, 2011, Postdoctoral Fellow, University Health Network, Toronto, Ontario, Canada.
•
Dr. Nand Lal, 2012, Scientist E1 at HLL-Lifecare Limited, Thiruvananthapuram, Kerala, India.
•
Dr. Santosh Jangir, 2014.
•
Dr. Veenu bala, 2014, Assistant Professor at Mohan Lal Sukhdia University, Rajasthan, India.
•
Ms. Sonal Gupta, 2015.
FORMER PROJECT ASSISTANTS
•
Ms. Mala Singh (2014-2016)
PRESENT Ph.D. STUDENTS
•
Mr. Dhanaraju Mandalapu (CSIR-SRF; 2012-present)
FORMER POSTGRADUATE STUDENTS
•
M. Jay Kothari (1997)
•
A.N. Misra (1997)
•
Ritu Chadda (1998)
•
Arun Kumar Misra (2000)
•
S.Nitya (2003)
•
Vishwanath Pratap Gupta (2004)
•
Divya (2006)
•
Charu Mahawar (2007)
•
Desh Deepak Pandey (2008)
•
Priyanka Pandey (2010)
•
Sumit Kumar (2010)
•
Sourabh Maheswari (2011)
•
Kartheek Nandikonda (2012)
•
Naveen Gupta (2012)
•
Pallavi Nayak (2012)
•
Neetika (2013)
•
Vikas Kumar (2013)
•
Neha Yadav (2013)
•
Subhadra Thakur (2014)
•
Jitendra Kumar (2015)
•
Suyash Tewari (2015)
•
Anjali Misra (2015)
MEMBERSHIP OF SOCIETES :
1. The Uttar Pradesh Association for Advancement of Science, Lucknow (India)
2. Indian Chemical Society (Calcutta)
3. Chemical Research Society of India, (Bangalore)
PROJECTS:
Reproductive Health Research: Male Reproductive Health and Contraception
1 Co – Principal Investigator: “Designed synthesis, evaluation and identification of novel, dually-effective spermicidal agents with anti-Trichomonal activity for ‘prophylactic’ contraception” (July 2014 – ongoing ), Funded by DHR, Indian council of Medical Research (ICMR), New Delhi.
2 Co-Principal Investigator: “Preclinical development of S,S’-Disulfanediylbis(pyrrolidinopropane-2,1-diyl) bis (piperidinothiocarbamate) as a vaginal contraceptive” (July 2011 – June 2013), Funded by Indian council of Medical Research (ICMR), New Delhi.
3 Principal Investigator: “Designed synthesis and biological evaluation of novel agents for management of benign prostatic hyperplasia” (November 2012 – October 2015), Funded by Indian council of Medical Research (ICMR), New Delhi.
PUBLICATIONS & PATENTS-
Total number of peer reviewed publications- 69 (Sixty Nine )
Total number of patents: (1 World patent and 4 National patents) – 5 (Five)
Citations to all publications: -Sum of times cited – 486, h-index- 12
SELECTED PUBLICATIONS
Dhanaraju Mandalapu, Deependra Kumar Singh, Sonal Gupta, Vishal M. Balaramnavar, Mohammad Shafiq, Dibyendu Banerjee, Vishnu Lal Sharma. Discovery of monocarbonyl curcumin hybrids as a novel class of human DNA ligase I inhibitors: in silico design, synthesis and biology. RSC Advances, 2016, 6, 26003.
Subhashis Pal, Kainat Khan, Shyamsundar Pal China, MonikaMittal, Konica porwal, Richa Shrivastava, Isha Taneja, Zakir Hossain, Dhanaraju Mandalapu, Jiaur R. Gayen, Muhammad Wahajuddin, Vishnu Lal Sharma, Arun K. Trivedi, Sabyasachi Sanyal, Smrati Bhadauria, Madan M. Godbole , Sushil K. Gupta, Naibedya Chattopadhyay. Theophylline, a methylxanthine drug induces osteopenia and alters calciotropic hormones and prophylactic vitamin D treatment protects against these changes in rats. Toxicology and Applied Pharmacology, 2016, 295, 12-25.
Bhavana Kushwaha, Dhanaraju Mandalapu, Veenu Bala, Lokesh Kumar, Aastha Pandey, Deepti Pandey, Santosh Kumar Yadav, Pratiksha Singh, P.K. Shukla, Jagdamba P. Maikhuri, Satya N. Sankhwar, Vishnu L. Sharma, Gopal Gupta. Ammonium salts of carbamodithioic acid as potent vaginal trichomonacides and fungicides. International Journal of Antimicrobial Agents, 2016, 47, 36-47.
Dhanaraju Mandalapu, Nand Lal, Lokesh Kumar, Bhavana Kushwaha, Sonal Gupta, Lalit Kumar, Veenu Bala, Santosh K. Yadav, Pratiksha Singh, Nidhi Singh, Jagdamba P. Maikhuri, Satya N. Sankhwar, Praveen K. Shukla, Imran Siddiqi, Gopal Gupta, Vishnu L. Sharma. Innovative Disulphide Esters of Dithiocarbamic acid as Women Controlled Contraceptive Microbicides: A Bioisosterism Approach. ChemMedChem, 2015, 10, 1739-1753.
Rachumallu Ramakrishna, Santosh kumar Puttrevu, Manisha Bhateria,Veenu Bala,Vishnu L. Sharma, Rabi Sankar Bhatta. Simultaneous determination of azilsartan and chlorthalidone in rat and human plasma by liquid chromatography-electrospray tandemmass spectrometry. Journal of Chromatography B, 2015,990, 185–197.
Hardik Chandasana, Yashpal S. Chhonkera, Veenu Bala, Yarra D. Prasad ,Telaprolu K. Chaitanya, Vishnu L. Sharma, Rabi S. Bhatta. Pharmacokinetic bioavailability, metabolism and plasma proteinbinding evaluation of NADPH-oxidase inhibitor apocynin using LC–MS/MS. Journal of Chromatography B, 2015, 985, 180–188.
Rajeev Kumar, Vikas Verma, Vikas Sharma, Ashish Jain, Vishal Singh, Amit Sarswat , Jagdamba P. Maikhuri, Vishnu L. Sharma, Gopal Gupta. A precisely substituted benzopyran targets androgen refractory prostate cancer cells through selective modulation of estrogen receptors. Toxicology and Applied Pharmacology, 2015, 283, 187-197.
Nand Lal, Amit Sarswat, Lalit Kumar, Karthik Nandikonda, Santosh Jangir, Veenu Bala, Vishnu Lal Sharma. Synthesis of Dithiocarbamates Containing Disulfide Linkage Using Cyclic Trithiocarbonate and Amines under Solvent–Catalyst Free Condition. Journal of Heterocyclic Chemistry, 2015, 52, 156-162.
Veenu Bala, Santosh Jangir, Dhanaraju Mandalapu, Sonal Gupta, Yashpal S. Chhonker, Nand Lal, Bhavana Kushwaha, Hardik Chandasana, Shagun Krishna, Kavita Rawat, Jagdamba P. Maikhuri, Rabi S. Bhatta, Mohammad I. Siddiqi,Rajkamal Tripathi, Gopal Gupta, Vishnu L. Sharma. Dithiocarbamate- Thiourea Hybrids Useful as Vaginal Microbicides Also Show Reverse Transcriptase Inhibition: Design, Synthesis, Docking and Pharmacokinetic studies. Bioorganic & Medicinal Chemistry Letters, 2015, 25, 881-886.
Gopal Gupta, Santosh Jangir and Vishnu Lal Sharma. Targeting post-ejaculation sperm for value-added contraception. Current Molecular Pharmacology, 2014, 7, 167-174.
Veenu Bala, Santosh Jangir, Vikas Kumar, Dhanaraju Mandalapu, Sonal Gupta, Lalit Kumar, Bhavana Kushwaha, Yashpal S. Chhonker, Atul Krishna, Jagdamba P. Maikhuri, Praveen K. Shukla, Rabi S. Bhatta, Gopal Gupta, Vishnu L. Sharma. Design and synthesis of substituted morpholin/piperidin-1-yl-carbamodithioates as promising vaginal microbicides with spermicidal potential. Bioorganic & Medicinal Chemistry Letters, 2014, 24, 5782-5786.
Veenu Bala, Gopal Gupta, Vishnu Lal Sharma. Chemical and Medicinal Versatility of Dithiocarbamates: An Overview. Mini Review Medicinal Chemistry, 2014, 14, 1021–1032.
Rakesh Kumar Asthana, Rasna Gupta, Nidhi Agrawal, Atul Srivastava, Upma Chaturvedi, Sanjeev Kanojiya, Ashok Kumar Khanna, Gitika Bhatia, Vishnu Lal Sharma. Evaluation of antidyslipidemic effect of mangiferin and amarogentin from swertia chirayita extract in hfd induced charles foster rat model and in vitroantioxidant activity and their docking studies. International Journal of Pharmaceutical Sciences and Research, 2014, 5(9), 3734-3740.
Santosh Jangir, Veenu Bala, Nand Lal, Lalit Kumar, Amit Sarswat, Amit Kumar, Hamidullah, Karan S. Saini, Vikas Sharma, Vikas Verma, Jagdamba P. Maikhuri, Rituraj Konwar, Gopal Gupta, Vishnu L. Sharma. Novel alkylphospholipid-DTC hybrids as promising agents against endocrine related cancers acting via modulation of Akt-pathway. European Journal of Medicinal Chemistry, 2014,85, 638-647.
Hardik Chandasana, Yashpal S. Chhonker, Veenu Bala, Yarra Durga Prasad,Vishnu L. Sharma, Rabi S. Bhatta. A rapid and sensitive LC-MS/MS analysis of diapocynin in rat plasma to investigate in vitro and in vivo pharmacokinetics.Analytical Methods 2014, 6, 7075-82.
Yashpal S. Chhonker, Hardik Chandasanaa, Veenu Bala, Lokesh Kumar,Vishnu Lal Sharma, Gopal Gupta, Rabi S. Bhatta. Quantitative determination of microbicidal spermicide ‘nonoxynol-9’ in rabbit plasma and vaginal fluid using LC–ESI–MS/MS: Application to pharmacokinetic. Journal of Chromatography B, 2014, 965, 127–132.
Mittal M, Khan K, Pal S, Porwal K, China SP, Barbhuyan TK, Bhagel KS, Rawat T, Sanyal S, Bhaduria S, Sharma VL, Chattopadhyay N. The Thiocarbamate Disulphide Drug, Disulfiram Induces Osteopenia in Rats by Inhibition of Osteoblast Function Due to Suppression of Acetaldehyde Dehydrogenase Activity.Toxicological Sciences, 2014, 239, 257-270.
Santosh Jangir, Veenu Bala, Nand Lal, Lalit Kumar, Amit Sarswat, Lokesh Kumar, Bhavana Kushwaha, Pratiksha Singh, Praveen K. Shukla, Jagdamba P. Maikhuri, Gopal Gupta, Vishnu L. Sharma. A unique dithiocarbamate chemistry during design & synthesis of novel sperm-immobilizing agents. Organic & Biomolecular Chemistry, 2014, 12 , 3090-3099.
Amit Anthwal, U. Chinna Rajesh, M.S.M. Rawat, Bhavana Kushwaha, Jagdamba P. Maikhuri, Vishnu L. Sharma, Gopal Gupta, Diwan S. Rawat. Novel metronidazole-chalcone cojugates with potential to counter drug resistance inTrichomona vaginalis. European Journal of Medicinal Chemistry, 2014, 79, 89-94.
Ashish Jain, Lokesh Kumar, Bhavana Kushwaha, Monika Sharma, Aastha Pandey, Vikas Verma, Vikas Sharma, Vishal Singh, Tara Rawat, Vishnu L. Sharma, Jagdamba P. Maikhuri, Gopal Gupta. Combining a synthetic spermicide with a natural trichomonacide for safe, prophylactic contraception. Human Reproduction, 2014, 29, 242-252.
Lalit Kumar, Nand Lal, Vikash Kumar, Amit Sarswat, Santosh Jangir, Veenu Bala, Lokesh Kumar, Bhavana Kushwaha, Atindra K. Pandey, Mohammad I. Siddiqi, Praveen K. Shukla, Jagdamba P. Maikhuri, Gopal Gupta, Vishnu L. Sharma. Azole-carbodithioate hybrids as vaginal anti-Candida contraceptive agents: design, synthesis and docking studies. European Journal of Medicinal Chemistry, 2013,70, 68-77.
Monika Sharma, Lokesh Kumar, Ashish Jain, Vikas Verma, Vikas Sharma, Bhavna Kushwaha, Nand Lal, Lalit Kumar, Tara Rawat, AK Dwivedi, JP Maikhuri, VL Sharma, Gopal Gupta. Designed chemical intervention with thiols for prophylactic contraception. PLOS-One, 2013, 8 (6), page 67365.
Lalit Kumar, Ashish Jain, Nand Lal, Amit Sarswat, Santosh Jangir, Lokesh Kumar, Priyanka Shah, Swatantra K. Jain, Jagdamba P. Maikhuri, Mohammad I. Siddiqi, Gopal Gupta, Vishnu L. Sharma. Potentiating metronidazole scaffold against resistant trichomonas: Design, synthesis, biology and 3D–QSAR analysis. ACS Medicinal Chemistry Letters, 2012, 3 (2), 83-87.
Kumar R, Verma V, Sarswat A, Maikhuri JP, Jain A, Jain RK, Sharma VL, Dalela D, Gupta G. Selective estrogen receptor modulators regulate stromal proliferation in human benign prostatic hyperplasia by multiple beneficial mechanisms-action of two new agents. Investigational New Drugs, 2012, 30, 582-593.
Ashish Jain, Nand Lal, Lokesh Kumar, Vikas Verma, Rajiv Kumar, Lalit Kumar, Vishal Singh, Raghav K. Mishra, Amit Sarswat, S. K. Jain, J. P. Maikhuri, V. L. Sharma, Gopal Gupta. Novel trichomonacidal spermicides. Antimicrobial Agents and Chemotherapy, 2011, 55 (9), 4343-4351.
Nand Lal, Lalit Kumar, Amit Sarswat, Santosh Jangir, Vishnu Lal Sharma. Synthesis of S-(2-thioxo-1,3-dithiolan-4-yl)methyl-dialkylcarbamothioate and S-thiiran-2-ylmethyl-dialkylcarbamothioate via Intermolecular O−S Rearrangement in Water. Organic Letters, 2011, 13 (9), 2330-2333.
Amit Sarswat, Rajeev Kumar, Lalit Kumar, Nand Lal, Smiriti Sharma, Yenamandra S. Prabhakar, Shailendra K. Pandey, Jawahar Lal, Vikas Verma, Ashish Jain, Jagdamba P. Maikhuri, Diwakar Dalela, Kirti, Gopal Gupta, Vishnu L. Sharma. Arylpiperazines for Management of Benign Prostatic Hyperplasia: Design, Synthesis, Quantative Structure – Activity Relationships, and Parmacokinetic Studies. Journal of Medicinal Chemistry, 2011, 54 (1), 302-311.
Lalit Kumar, Amit Sarswat, Nand Lal, Ashish Jain, Sumit Kumar, S.T.V.S. Kiran Kumar, Jagdamba P. Maikhuri, Atindra K. Pandey, Praveen K. Shukla, Gopal Gupta, Vishnu L. Sharma. Design and Synthesis of 3-(azol-1-yl)phenylprapanes as spermicide for prophylactic contraception . Bioorganic & Medicinal Chemistry Letters, 2011, 21(1), 176-181.
LIST OF PATENTS
1 Kalpana Bhandari, V.L. Sharma and S. Ray. “An improved process for the synthesis of 3,4-disubstituted-1,5-dihydro-2H-3-pyrrolin-2-one” Indian PatentAppl. 323/Del/01 dt 23.3.2001.
2 A.K.Dwivedi, V.L.Sharma, N.Kumaria, Kiran Kumar, G.Gupta, J.P.Maikhuri, J.D.Dhar, Pradeep Kumar, A.H.Ansari, P.K.Shukla, M.Kumar, Raja Roy , K.P.Madhusudanan, R.C.Gupta, Pratima Srivastava, R.Pal, and S.Singh. “Novel spermicidal and antifungal agents” Indian Patent 245815 dt 25.01.2011 ; Appl. No.1792/Del/04 dt 22.09.2004.
3 Vishanu Lal Sharma, Nand Lal, Amit Sarswat, Santosh Jangir, Veenu Bala, Lalit Kumar, Tara Rawat, Ashish Jain, Lokesh Kumar, Jagdamba Prasad Maikhuri, Gopal Gupta. “ Carbodithioates and process for preparation thereof ” NF No. 0030/NF2013/IN, Indian Patent Appl. no.0373/DEL/2013 dated 08.02.2013.
4 Vishanu Lal Sharma, Nand Lal, Amit Sarswat, Santosh Jangir, Veenu Bala, Lalit Kumar, Tara Rawat, Ashish Jain, Lokesh Kumar, Jagdamba Prasad Maikhuri, Gopal Gupta, “ Carbodithioates with spermicidal activity and process for preparation thereof ” PCT Patent no. WO 2014122670 August 14, 2014.
5 Dhanaraju Mandalapu, Rajesh K. Arigela, Tara Rawat, and Vishnu L. Sharma, “An Improved Process For Preparation Of 4-Substituted amino-2,3-polymethylenequinoline hydrochloride ” Indian Patent IN 201611003055 dated: 28.01.2016.

 

Image result for Medicinal & Process Chemistry Division, CSIR-Central Drug Research Institute

From left to right upper row: Dr. S.T.V.S. Kiran Kumar, Dr. Lalit Kumar, Dr. V.L. Sharma, Dr. Nand Lal, Dr. Amit Sarswat
Lower row: Dhanaraju Mandalapu, Sonal Gupta, Mrs. Tara Rawat (S.T.O.), Dr. Veenu bala, Dr. Santosh Jangir

///////////aryl piperazine, androgen sensitive prostatic disorders, 330633-91-5, CDRI-?

c1(ccc(cc1)[N+]([O-])=O)N2CCN(CC2)C(=O)CN3CCN(CC3)c4ccc(cc4)[N+]([O-])=O

 


Filed under: Preclinical drugs, Uncategorized Tagged: 330633-91-5, androgen sensitive prostatic disorders, aryl piperazine, cdri

DDD 107498

$
0
0

str1

 

DDD 107498, DDD 498

PATENT WO 2013153357,  US2015045354

6-Fluoro-2-[4-(morpholinomethyl)phenyl]-N-(2-pyrrolidin-1-ylethyl)quinoline-4-carboxamide

6-Fluoro-2-[4-(4-morpholinylmethyl)phenyl]-N-[2-(1-pyrrolidinyl)ethyl]-4-quinolinecarboxamide

4-Quinolinecarboxamide, 6-fluoro-2-[4-(4-morpholinylmethyl)phenyl]-N-[2-(1-pyrrolidinyl)ethyl]-

CAS 1469439-69-7

CAS 1469439-71-1 SUCCINATE

MF C27H31FN4O2
MW 462.559043 g/mol
      6-fluoro-2-[4-(morpholin-4-ylmethyl)phenyl]-N-(2-pyrrolidin-1-ylethyl)quinoline-4-carboxamide
  • Originator Medicines for Malaria Venture; University of Dundee
  • Class Small molecules
  • Mechanism of Action Protein synthesis inhibitors

Highest Development Phases

  • No development reported Malaria

Most Recent Events

  • 16 Jul 2016 No recent reports of development identified for preclinical development in Malaria in United Kingdom
  • 01 Apr 2015 DDD 498 licensed to Merck Serono worldwide for the treatment of Malaria
Inventors Ian Hugh Gilbert, Neil Norcross, Beatriz Baragana Ruibal, Achim Porzelle
Original Assignee University Of Dundee

str1Image result for School of Life Sciences University of Dundee

Prof Ian Gilbert:

Head of Biological Chemistry and Drug Discovery

BCDD, College of Life Sciences, University of Dundee, DD1 5EH, UK
Tel: +44 (0) 1382-386240

 

University of Dundee

Image result for School of Life Sciences University of Dundee

 

Image result for School of Life Sciences University of Dundee

SCHEMBL15322600.pngDDD498

 

str1

 

Merck Serono and MMV sign agreement to develop potential antimalarial therapy

Agreement further diversifies MMV’s partner base, strengthening our antimalarial research and development portfolio

01 April 2015

Photo © Merck Serono

Merck Serono, the biopharmaceutical business of Merck, and MMV announced today that an agreement has been signed for Merck Serono to obtain the rights to the investigational antimalarial compound DDD107498 from MMV. This agreement underscores the commitment of Merck Serono to provide antimalarials for the most vulnerable populations in need.

“This agreement strengthens our Global Health research program and our ongoing collaboration with Medicines for Malaria Venture,” said Luciano Rossetti, Executive Vice President, Global Head of Research & Development at Merck Serono. “MMV is known worldwide for its major contribution to delivering innovative antimalarial treatments to the most vulnerable populations suffering from this disease, and at Merck Serono we share this goal.”

DDD107498 originated from a collaboration between MMV and the University of Dundee Drug Discovery Unit, led by Prof. Ian Gilbert and Dr. Kevin Read. The objective of the clinical program is to demonstrate whether the investigational compound exerts activity on a number of malaria parasite lifecycle stages, and remains active in the body long enough to offer potential as a single-dose treatment against the most severe strains of malaria.

While development and commercialization of the compound is under Merck Serono’s responsibility, MMV will provide expertise in the field of malaria drug development, including its clinical and delivery expertise, and provide access to its public and private sector networks in malaria-endemic countries.

Merck Serono has a dedicated Global Health R&D group working to address key unmet medical needs related to neglected diseases, such as schistosomiasis and malaria, with a focus on pediatric populations in developing countries. Its approach is based on public-private partnerships and collaborations with leading global health institutions and organizations in both developed and developing countries.

“Working with partners like Merck Serono is critical to the progress of potential antimalarial compounds, like DDD107498, through the malaria drug pipeline,” said Dr. Timothy Wells, Chief Scientific Officer at MMV. “Their Global Health Program is gaining momentum and we need more compounds to tackle malaria, a disease that places a huge burden on the world’s most vulnerable populations. DDD107498 holds great promise and we look forward to working with the Merck Serono team through the development phase.”

According to the World Health Organization, there were an estimated 198 million cases of malaria worldwide in 2013, and an estimated 584,000 deaths, primarily in young children from the developing world. The launch of the not-for-profit research foundation, MMV, in 1999 and a number of collaborations and partnerships, including those with Merck Serono, has contributed to reducing the major gap in malaria R&D investment and subsequent dearth of new medicines.

“It’s hugely encouraging to see the German pharmaceutical industry increasing their engagement in the development of novel antimalarials,” said global malaria expert Prof. Dr. Peter Kremsner, Director of the Institute for Tropical Medicine at the University of Tübingen, Germany. “The Merck Serono and MMV collaboration to develop DDD107498 is a great step. It’s a compound that offers lots of promise so I’m excited to see how it progresses.

str1str2

Scots scientists in ‘single dose’ malaria treatment breakthrough

An antimalarial drug that could treat patients was discovered by Dundee university scientists

Scientists have discovered an antimalarial compound that could treat malaria patients in a single dose and help prevent the spread of the disease from infected people.

The compound DDD107498 also has the potential to treat patients with malaria parasites resistant to current medications, researchers say.

Scientists hope it could lead to treatments and protection against the disease, which claimed almost 600,000 lives amid 200 million reported cases in 2013.

The compound was identified through a collaboration between the University of Dundee’s drug discovery unit (DDU) and the Medicines for Malaria Venture (MMV), a separate organisation.

The compound is now undergoing further safety testing with a view to entering human clinical trials within the next year.

Details of the discovery have been published in the journal Nature.

Professor Ian Gilbert, head of chemistry at the DDU, who led the team that discovered the compound, said: “The publication describes the discovery and profiling of this exciting new compound.

“It reveals that DDD107498 has the potential to treat malaria with a single dose, prevent the spread of malaria from infected people and protect a person from developing the disease in the first place.

“There is still some way to go before the compound can be given to patients. However, we are very excited by the progress that we have made.”

The World Health Organisation reports that there were 200 million clinical cases of malaria in 2013, with 584,000 people dying from the disease. Most of these deaths were children under the age of five and pregnant women.

MMV chief executive officer Dr David Reddy said: “Malaria continues to threaten almost half of the world’s population – the half that can least afford it.

“DDD107498 is an exciting compound since it holds the promise to not only treat but also protect these vulnerable populations.

“The collaboration to identify and progress the compound, led by the drug discovery unit at the University of Dundee, drew on MMV’s network of scientists from Melbourne to San Diego.”The publication of the research is an important step and a clear testament to the power of partnership.”

MMV selected DDD107498 to enter preclinical development in October 2013 following the recommendation of its expert scientific advisory committee.

Since then, with MMV’s leadership, large quantities of the compound have been produced and it is undergoing further safety testing with a view to entering human clinical trials within the next year.

Merck Serono has recently obtained the right to develop and, if successful, commercialise the compound, with the input of MMV’s expertise in the field of malaria drug development and access and delivery in malaria-endemic countries.

Dr Michael Chew from the Wellcome Trust, which provides funding for the DDU and MMV, said: “The need for new antimalarial drugs is more urgent than ever before, with emerging strains of the parasite now showing resistance against the best available drugs.

“These strains are already present at the Myanmar-Indian border and it’s a race against time to stop resistance spreading to the most vulnerable populations in Africa.

“The discovery of this new antimalarial agent, which has shown remarkable potency against multiple stages of the malaria lifecycle, is an exciting prospect in the hunt for viable new treatments.”

PAPER

 

Abstract Image

Figure

Discovery of a Quinoline-4-carboxamide Derivative with a Novel Mechanism of Action, Multistage Antimalarial Activity, and Potent in Vivo Efficacy

Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, U.K.
Cell and Molecular Biology, Department of Life Sciences, Imperial College, London, SW7 2AZ, U.K.
§ School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
Eskitis Institute, Griffith University, Brisbane Innovation Park, Nathan Campus, Brisbane, QLD 4111, Australia
Swiss Tropical and Public Health Institute, Swiss TPH, Socinstrasse 57, 4051 Basel, Switzerland
#University of Basel, CH-4003 Basel, Switzerland
Medicines for Malaria Venture, International Centre Cointrin, Entrance G, 3rd Floor, Route de Pré-Bois 20, P.O. Box 1826, CH-1215, Geneva 15, Switzerland
J. Med. Chem., Article ASAP
DOI: 10.1021/acs.jmedchem.6b00723
*K.D.R.: phone, +44 1382 388 688; e-mail, k.read@dundee.ac.uk., *I.H.G.: phone, +44 1382 386 240; e-mail,i.h.gilbert@dundee.ac.uk.
Figure
Conditions: (a) morpholine, Et3N, DCM, 16 h, 72% yield; (b) MeMgBr, toluene, reflux, 4 h and then a 10% aqueous HCl, reflux, 1 h, 70% yield; (c) NBS, benzoyl peroxide, dichlorobenzene, 140 °C, 16 h, 70% yield; (d) morpholine, K2CO3, acetonitrile, 40 °C, 16 h, 64% yield; (e) 5-fluoroisatin, KOH, EtOH, 120 °C, microwave, 20 min, 30–76% yield; (f) amine, CDMT, N-methylmorpholine, DCM, 20–61% yield.

// https://tpc.googlesyndication.com/pagead/js/r20160906/r20110914/abg.js//

 

A single-dose treatment against malaria worked in mice to cure them of the disease. The drug also worked to block infection in healthy mice and to stop transmission, according to a study published in Nature today. The fact that the drug can act against so many stages of malaria is pretty new, but what’s even more exciting is the compound’s mode of action: it kills malaria in a completely new way, researchers say. The feature would make it a welcome addition to our roster of antimalarials — a roster that’s threatened by drug resistance.

RESEARCHERS SIFTED THROUGH A LIBRARY OF ABOUT 4,700 COMPOUNDS TO FIND THIS ONE

Malaria is an infectious disease that’s transmitted through mosquito bites; it’s also a leading cause of death in a number of developing countries. Approximately 3.4 billion people live in areas where malaria poses a real threat. As a result, there were 207 million cases of malaria in 2012 — and 627,000 deaths. There are drugs that can be used to prevent malaria, and even treat it, but drug resistance is halting the use of certain treatments in some areas.

A long search

Searching for a new drug is all about trial and error. To find this particular compound, researchers sifted through a library of about 4,700 compounds, testing them to see if they were capable of killing the malaria parasite in a lab setting. When they found something that worked, they tweaked the drug candidate to see if it could perform more effectively. “We went through a lot of these cycles of testing and designing new compounds,” says Ian Gilbert, a medicinal chemist at the University of Dundee in the UK, and a co-author of the study. “Eventually we optimized to the compound which is the subject of the paper.” For now, that compound’s unwieldy name is DDD107498.

To make sure DDD107498 really had potential, the researchers tested it on mice that had already been infected with malaria. A single dose was enough to provoke a 90 percent reduction in the number of parasites in their blood. The scientists also gave the compound to healthy mice that were subsequently exposed to malaria. DDD107498 helped the mice evade infection with a single dose, but it’s unclear how long that effect would last in humans. Finally, the researchers looked at whether the compound could prevent the transmission from an infected mouse to a mosquito. A day after receiving the treatment, mice were put in contact with mosquitoes. The scientists noted a 91 percent reduction in infected mosquitoes.

“IT HAS THE ABILITY TO BE A ONE-DOSE [DRUG], IN COMBINATION WITH ANOTHER MOLECULE.”

“What’s exciting about this molecule is obviously the fact that it has the ability to be a one-dose [drug], in combination with another molecule to cure blood stage malaria,” says Kevin Read, a drug researcher also at the University of Dundee and a co-author of the study. The fact that the compound has the ability to block transmission and protect against infection is equally thrilling. But the way in which DDD107498 kills malaria might be its most interesting feature. It halts the production of proteins — which are necessary for the parasite’s survival. No other malaria drug does that right now, Read says. “So, in principle, there’s no resistance out there already to this mechanism.”

The drug hasn’t been tested in humans yet, so it may not be nearly as good in the field. But Read says DDD107498 looks promising. “From all the pre-clinical or non-clinical data we’ve generated, it is comparable or better than any of the current marketed anti-malarials in those studies.” And at $1 per treatment, the price of the drug should fall “within the range of what’s acceptable,” he says.

“It looks like an excellent study, and the results look very important,” says Philip Rosenthal, a malaria drug researcher at The University of California-San Francisco who didn’t participate in the study. This is a big shift for Rosenthal’s field. Five years ago, “we had very little going on in anti-malarial drug discovery,” he says. Now, there’s quite a bit going on for malaria researchers, and a number of promising compounds are moving along. DDD107498 “is another player, and it’s got a number of positive features,” he says.

OTHER TREATMENTS HAVE TO BE TAKEN FOR A FEW DAYS

One of the features is the drug’s potency. It’s very active against cultured malaria parasites, Rosenthal says. But what’s perhaps most intriguing about DDD107498 is that the drug works against the mechanism that enables protein synthesis the malaria parasite’s cells. No other malaria drug does that right now, Read says. “Considering challenges of treating malaria, which is often in rural areas and developing countries, a single dose would be a big plus,” he says. “In addition, because of it’s long half life, it may also work to prevent malaria with once a week dosing, which is also a benefit.”

Still, no drug is perfect. The data suggests that DDD107498 doesn’t kill malaria as quickly as some other drugs, Rosenthal says. And when the researchers tested it to see how long it might take for resistance to develop, the results weren’t as promising as he would like. The parasites figured out a way to become resistant to the compound “relatively easily,” he says. That shouldn’t be “deal-killer,” however. “Its slow onset of action probably means it should be combined with a faster-acting drug,” he says.

BUT IT’S SLOW-ACTING

The compound is going through safety testing now. If everything goes well, it should hit human trials within the next year, Read says. Chances are, it will have to be used in combination with other malaria drugs, Gilbert says. “All anti-malarials are given in combination because it slows down resistance.”

“When you’re treating infectious diseases, you know that drug resistance is always a potential problem, so having a number of choices to treat malaria is a good thing,” Rosenthal says. In this case, the drug’s new mode of action may hold lead to an entirely new weapon against malaria. “Obviously it’s got a long way to go,” Read says. But the compound is “very exciting,” nonetheless.

// https://tpc.googlesyndication.com/pagead/js/r20160906/r20110914/abg.js//

//

PATENT
str1 str2 str3 str4
Example 16-Fluoro-2-[4-(morpholinomethyl)phenyl]-N-(2-pyrrolidin-1-ylethyl)quinoline-4-carboxamide, Example compound 1 in Scheme 2
str1
In a sealed microwave tube, a suspension of 2-chloro-6-fluoro-N-(2-pyrrolidin-1-ylethyl)quinoline-4-carboxamide (preparation 4) (2.00 g, 6 mmol), [4-(morpholinomethyl)phenyl]boronic acid, hydrochloride, available from UORSY, (3.20 g, 12 mmol), potassium phosphate (2.63 g, 12 mmol) and tetrakis(triphenylphosphine)palladium (0) (0.21 g, 0.19 mmol) in DMF/Water 3/1 (40 ml) was heated at 130° C. under microwave irradiation for 30 min. The reaction was filtered through Celite™ and solvents were removed under reduced pressure. The resulting residue was taken up in DCM (150 ml) and washed twice with NaHCO3 saturated aqueous solution (2×100 ml). The organic layer was separated, dried over MgSO4 and concentrate to dryness under reduced pressure. The reaction crude was purified by flash column chromatography using an 80 g silica gel cartridge and eluting with DCM (Solvent A) and MeOH (Solvent B) and the following gradient: 1 min hold 100% A, followed by a 30 min ramp to 10% B, and then 15 min hold at 10% B. The fractions containing product were pooled together and concentrated to dryness under vacuum to obtain the desired product as an off-white solid (1 g). The product was dissolved in methanol (100 ml) and 3-mercaptopropyl ethyl sulfide Silica (Phosphonics, SPM-32, 60-200 uM) was added. The suspension was stirred at room temperature over for 2 days and then at 50° C. for 1 h. After cooling to room temperature, the scavenger was filtered off and washed with methanol (30 ml). The solvent was removed under reduced pressure and the product was further purified by preparative HPLC. The fractions containing product were pooled together and freeze dried to obtain the desired product as a white solid (0.6 g, 1.3 mmol, Yield 20%).
1H NMR (500 MHz; CDCl3) δ 1.81-1.84 (m, 4H), 2.50-2.52 (m, 4H), 2.63 (brs, 4H), 2.82 (t, 2H, J=5.9 Hz), 3.61 (s, 2H), 3.71 (dd, 2H, J=5.4 Hz, J=11.4 Hz), 3.74-3.76 (m, 4H), 6.84 (brs, 1H), 7.52-7.57 (m, 3H), 7.97-8.00 (m, 2H), 8.13 (d, 2H, J=8.2 Hz), 8.21 (dd, 1H, J=5.5 Hz, J=9.2 Hz) ppm. 19F NMR (407.5 MHz; CDCl3) δ−111.47 ppm.
Purity by LCMS (UV Chromatogram, 190-450 nm) 99%, rt=5.7 min, m/z 463 (M+H)+ HRMS (ES+) found 463.2501 [M+H]+, C27H32F1N4O2 requires 463.2504.
Example 26-Fluoro-2-[4-(morpholinomethyl)phenyl]-N-(2-pyrrolidin-1-ylethyl)quinoline-4-carboxamide; fumaric acid salt, compound (IB) in Scheme 2
str1
The starting free base (example 1) (0.58 g, 1 mmol) was dissolved in dry ethanol (10 ml) and added dropwise to a stirred solution of fumaric acid (0.15 g, 1 mmol) in dry ethanol (9 ml). The mixture was stirred at room temperature for 1 h. The white precipitate was filtered, washed with ethanol (20 ml) and then dissolved in 10 ml of water and freeze dried to obtain the desired salt as a white solid (0.601 g, 1 mmol, Yield 82%).
1H NMR (500 MHz; d6-DMSO) δ 1.83-1.86 (m, 4H), 2.41 (brs, 4H), 2.94 (brs, 4H), 3.03 (t, 2H, J=6.2 Hz), 3.57 (s, 2H), 3.60-3.65 (m, 6H), 6.47 (s, 2H), 7.51 (d, 2H, J=8.25), 7.74-7.78 (m, 1H), 8.06 (dd, 1H, J=2.9 Hz, J=10.4 Hz), 8.17 (dd, 1H, J=5.7 Hz, J=9.3 Hz), 8.24-8.26 (m, 3H), 9.24 (t, 1H, J=5.5 Hz) ppm. 19F NMR (407.5 MHz; d6-DMSO) δ-112.30 ppm.
Purity by LCMS (UV Chromatogram, 190-450 nm) 99%, rt=5.3 min, m/z 463 (M+H)+
Example 1AAlternative synthesis of 6-fluoro-2-[4-(morpholinomethyl)phenyl]-N-(2-pyrrolidin-1-ylethyl)quinoline-4-carboxamide, Example compound 1A in Scheme 4
str1
To a stirred suspension of 6-fluoro-2-[4-(morpholinomethyl)phenyl]quinoline-4-carboxylic acid (preparation 7) (2.20 g, 6 mmol) in DCM (100 ml) at room temperature, 2-chloro-4,6-dimethoxy-1,3,5-triazine (CDMT) (1.26 g, 7 mmol) and 4-methylmorpholine (NMO) (1.33 ml, 12 mmol) were added. The reaction mixture was stirred at room temperature for 1 h and then 2-pyrrolidin-1-ylethanamine (0.77 ml, 6 mmol) was added and stirred at room temperature for further 3 h. The reaction mixture was washed with NaHCO3 saturated aqueous solution (2×100 ml) and the organic phase was separated, dried over MgSO4 and concentrated under reduced pressure. The resulting residue was absorbed on silica gel and purified by flash column chromatography using an 80 g silica gel cartridge and eluting with DCM (Solvent A) and MeOH (Solvent B) and the following gradient: 2 min hold 100% A followed by a 30 min ramp to 10% B and then 15 min hold at 10% B. The desired fractions were concentrated to dryness under vacuum to obtain the crude product as a yellow solid (95% purity by LCMS). The sample was further purified by a second column chromatography using a 40 g silica gel cartridge, eluting with DCM (Solvent A) and 10% NH3-MeOH in DCM (Solvent B) and the following gradient: 2 min hold 100% A, followed by a 10 min ramp to 23% B and then 15 min hold at 23% B. The desired fractions were concentrated to dryness under vacuum to obtain product as a white solid (1 g). Re-crystallisation form acetonitrile (18 ml) yielded the title compound as a white solid (625 mg, 1.24 mmol, 20%).
1H NMR (500 MHz; CDCl3) δ 1.81-1.84 (m, 4H), 2.50-2.52 (m, 4H), 2.63 (brs, 4H), 2.82 (t, 2H, J=5.9 Hz), 3.61 (s, 2H), 3.71 (dd, 2H, J=5.4 Hz, J=11.4 Hz), 3.74-3.76 (m, 4H), 6.84 (brs, 1H), 7.52-7.57 (m, 3H), 7.97-8.00 (m, 2H), 8.13 (d, 2H, J=8.2 Hz), 8.21 (dd, 1H, J=5.5 Hz, J=9.2 Hz) ppm.
1H NMR (500 MHz; d6-DMSO) δ 1.72-1.75 (m, 4H), 2.41 (brs, 4H), 2.56 (brs, 4H), 2.67 (t, 2H, J=6.6 Hz), 3.49-3.52 (m, 2H), 3.56 (s, 2H), 3.60-3.61 (m, 4H), 7.52 (d, 2H, J=8.3 Hz), 7.73-7.77 (m, 1H), 8.07 (dd, 1H, J=2.9 Hz, J=10.4 Hz), 8.18-8.21 (m, 2H), 8.26 (d, 2H, J=8.3 Hz), 8.85 (t, 1H, J=6.6 Hz) ppm.
13C NMR (125 MHz; d6-DMSO3) δ 23.2, 38.4, 53.2, 53.5, 54.5, 62.1, 66.2, 109.0, 109.1, 117.3, 120.1, 120.3, 124.1, 124.2, 127.1, 129.4, 132.2, 132.3, 136.8, 139.9, 142.8, 145.2, 155.3, 159.0, 161.0, 166.1 ppm.
19F NMR (500 MHz; d6-DMSO) δ-112.47 ppm.
Purity by LCMS (UV Chromatogram, 190-450 nm) 99%, rt=5.0 min, m/z 463 (M+H)+
PATENT
WO 2016033635
Patent
WO 2013153357

SCHEME 1

Figure imgf000018_0001

SCHEME 2

Figure imgf000019_0001

Preparation 4Yield: 54% Preparation 3

Yield: 27%

Figure imgf000019_0002

SCHEME 4 B

Figure imgf000021_0001

Yield: 72% Yield: 70% Preparation 6

Figure imgf000021_0002

Example 1 : 6-Fluoro-2-r4-(morpholinomethyl)phenyll-N-(2-pyrrolidin-1-ylethyl)quinoline- 4-carboxamide, Example compound 1 in Scheme 2

Figure imgf000050_0002

In a sealed microwave tube, a suspension of 2-chloro-6-fluoro-N-(2-pyrrolidin-1- ylethyl)quinoline-4-carboxamide (preparation 4) (2.00 g, 6 mmol), [4- (morpholinomethyl)phenyl]boronic acid, hydrochloride, available from UORSY, (3.20 g, 12 mmol), potassium phosphate (2.63 g, 12 mmol) and tetrakis(triphenylphosphine)palladium (0) (0.21 g, 0.19 mmol) in DMF/Water 3/1 (40 ml) was heated at 130°C under microwave irradiation for 30 min. The reaction was filtered through Celite™ and solvents were removed under reduced pressure. The resulting residue was taken up in DCM (150 ml) and washed twice with NaHC03 saturated aqueous solution (2 x 100 ml). The organic layer was separated, dried over MgS04and concentrate to dryness under reduced pressure. The reaction crude was purified by flash column chromatography using an 80 g silica gel cartridge and eluting with DCM (Solvent A) and MeOH (Solvent B) and the following gradient: 1 min hold 100% A, followed by a 30 min ramp to 10 % B, and then 15 min hold at 10% B. The fractions containing product were pooled together and concentrated to dryness under vacuum to obtain the desired product as an off-white solid (1 g). The product was dissolved in methanol (100 ml) and 3-mercaptopropyl ethyl sulfide Silica (Phosphonics, SPM-32, 60- 200 uM) was added. The suspension was stirred at room temperature over for 2 days and then at 50°C for 1 h. After cooling to room temperature, the scavenger was filtered off and washed with methanol (30 ml). The solvent was removed under reduced pressure and the product was further purified by preparative HPLC. The fractions containing product were pooled together and freeze dried to obtain the desired product as a white solid (0.6 g, 1.3 mmol, Yield 20%).

1 H NMR (500 MHz; CDCI3) δ 1.81-1.84 (m, 4H), 2.50-2.52 (m, 4H), 2.63 (brs, 4H), 2.82 (t, 2H, J = 5.9 Hz), 3.61 (s, 2H), 3.71 (dd, 2H, J = 5.4 Hz, J = 1 1.4 Hz), 3.74-3.76 (m, 4H), 6.84 (brs, 1 H), 7.52-7.57 (m, 3H), 7.97-8.00 (m, 2H), 8.13 (d, 2H, J = 8.2 Hz), 8.21 (dd, 1 H, J = 5.5 Hz, J = 9.2 Hz) ppm . 19 F NMR (407.5 MHz; CDCI3) δ -11 1.47 ppm. Purity by LCMS (UV Chromatogram, 190-450nm) 99 %, rt = 5.7 min, m/z 463 (M+H)+ HRMS (ES+) found 463.2501 [M+H]+, C27H32F1 N402 requires 463.2504.

Example 2: 6-Fluoro-2-[4-(morpholinomethyl)phenyl1-N-(2-pyrrolidin-1-ylethyl)quinoline- 4-carboxamide; fumaric acid salt, compound (IB) in Scheme 2

Figure imgf000051_0001

The starting free base (example 1) (0.58 g, 1 mmol) was dissolved in dry ethanol (10 ml) and added dropwise to a stirred solution of fumaric acid (0.15 g, 1 mmol) in dry ethanol (9 ml). The mixture was stirred at room temperature for 1 h. The white precipitate was filtered, washed with ethanol (20 ml) and then dissolved in 10 ml of water and freeze dried to obtain the desired salt as a white solid (0.601 g, 1 mmol, Yield 82%).

1 H NMR (500 MHz; d6-DMSO) δ 1.83-1.86 (m, 4H), 2.41 (brs, 4H), 2.94 (brs, 4H), 3.03 (t, 2H, J = 6.2 Hz), 3.57 (s, 2H), 3.60-3.65 (m, 6H), 6.47 (s, 2H), 7.51 (d, 2H, J = 8.25), 7.74-7.78 (m, 1 H), 8.06 (dd, 1 H, J = 2.9 Hz, J = 10.4 Hz), 8.17 (dd, 1 H, J = 5.7 Hz, J = 9.3 Hz), 8.24-8.26 (m, 3H), 9.24 (t, 1 H, J = 5.5 Hz) ppm. 19 F NMR (407.5 MHz; d6- DMSO) δ -112.30 ppm.

Purity by LCMS (UV Chromatogram, 190-450nm) 99 %, rt = 5.3 min, m/z 463 (M+H)+

Example 1A: Alternative synthesis of 6-fluoro-2-[4-(morpholinomethyl)phenyl1-N-(2- pyrrolidin-1-ylethyl)quinoline-4-carboxamide, Example compound 1A in Scheme 4

Figure imgf000052_0001

To a stirred suspension of 6-fluoro-2-[4-(morpholinomethyl)phenyl]quinoline-4-carboxylic acid (preparation 7) (2.20 g, 6 mmol) in DCM (100 ml) at room temperature, 2-chloro- 4,6-dimethoxy-1 ,3,5-triazine (CDMT) (1.26 g, 7 mmol) and 4-methylmorpholine (NMO) (1.33 ml, 12 mmol) were added. The reaction mixture was stirred at room temperature for 1 h and then 2-pyrrolidin-1-ylethanamine (0.77 ml, 6 mmol) was added and stirred at room temperature for further 3 h. The reaction mixture was washed with NaHC03 saturated aqueous solution (2x 100 ml) and the organic phase was separated, dried over MgS04 and concentrated under reduced pressure. The resulting residue was absorbed on silica gel and purified by flash column chromatography using an 80 g silica gel cartridge and eluting with DCM (Solvent A) and MeOH (Solvent B) and the following gradient: 2 min hold 100% A followed by a 30 min ramp to 10 %B and then 15 min hold at 10%B. The desired fractions were concentrated to dryness under vacuum to obtain the crude product as a yellow solid (95% purity by LCMS). The sample was further purified by a second column chromatography using a 40 g silica gel cartridge, eluting with DCM (Solvent A) and 10% NH3-MeOH in DCM (Solvent B) and the following gradient: 2 min hold 100% A, followed by a 10 min ramp to 23 % B and then 15 min hold at 23% B. The desired fractions were concentrated to dryness under vacuum to obtain product as a white solid (1 g). Re-crystallisation form acetonitrile (18 ml) yielded the title compound as a white solid (625 mg, 1.24 mmol, 20%).

1 H NMR (500 MHz; CDCI3) δ 1.81-1.84 (m, 4H), 2.50-2.52 (m, 4H), 2.63 (brs, 4H), 2.82 (t, 2H, J = 5.9 Hz), 3.61 (s, 2H), 3.71 (dd, 2H, J = 5.4 Hz, J = 1 1.4 Hz), 3.74-3.76 (m, 4H), 6.84 (brs, 1 H), 7.52-7.57 (m, 3H), 7.97-8.00 (m, 2H), 8.13 (d, 2H, J = 8.2 Hz), 8.21 (dd, 1 H, J = 5.5 Hz, J = 9.2 Hz) ppm .

1 H NMR (500 MHz; d6-DMSO) δ 1.72-1.75 (m, 4H), 2.41 (brs, 4H), 2.56 (brs, 4H), 2.67 (t, 2H, J = 6.6 Hz), 3.49-3.52 (m, 2H), 3.56 (s, 2H), 3.60-3.61 (m, 4H), 7.52 (d, 2H, J = 8.3 Hz), 7.73-7.77 (m, 1 H), 8.07 (dd, 1 H, J = 2.9 Hz, J = 10.4 Hz), 8.18-8.21 (m, 2H), 8.26 (d, 2H , J = 8.3 Hz), 8.85 (t, 1 H, J = 6.6 Hz) ppm.

13C NMR (125 MHz; d6-DMS03) 5 23.2, 38.4, 53.2, 53.5, 54.5, 62.1 , 66.2, 109.0, 109.1 , 1 17.3, 120.1 , 120.3, 124.1 , 124.2, 127.1 , 129.4, 132.2, 132.3, 136.8, 139.9, 142.8, 145.2, 155.3, 159.0, 161 .0, 166.1 ppm.

19 F NM R (500 MHz; d6-DMSO) δ -1 12.47 ppm.

Purity by LCMS (UV Chromatogram, 190-450nm) 99 %, rt = 5.0 min, m/z 463 (M+H)+

PAPER
A Quinoline Carboxamide Antimalarial Drug Candidate Uniquely Targets Plasmodia at Three Stages of the Parasite Life Cycle
Angewandte Chemie, International Edition (2015), 54, (46), 13504-13506
original image

Putting a stop to malaria: Phenotypic screening against malaria parasites, hit identification, and efficient lead optimization have delivered the preclinical candidate antimalarial DDD107498. This molecule is distinctive in that it has potential for use as a single-dose cure for malaria and shows a unique broad spectrum of activity against the liver, blood, and mosquito stages of the parasite life cycle.

 Prof. P. M. O’Neill Department of Chemistry, University of Liverpool Liverpool, L69 7ZD (UK) E-mail: pmoneill@liverpool.ac.uk Prof. S. A. Ward Liverpool School of Tropical Medicine, Pembroke Place Liverpool, L3 5QA (UK)
 str1

Professor Ian Gilbert FRSC

Design and synthesis of potential therapeutic agents
Position:
Professor of Medicinal Chemistry and Head of the Division of Biological Chemistry and Drug Discovery
Address:
College of Life Sciences, University of Dundee, Dundee
Full Telephone:
+44 (0) 1382 386240, int ext 86240

Dr Neil Norcross

Position:
Medicinal Chemist
Address:
College of Life Sciences, University of Dundee, Dundee
Full Telephone:
(0) , int ext
Image result for Beatriz Baragana Ruibal
La investigadora asturiana Beatriz Baragaña, en La Pola. / PABLO NOSTI
Image result for Achim Porzelle

Achim Porzelle

REFERENCES

///////////DDD107498, DDD 107498, PRECLINICAL, DUNDEE, MALARIA, DDD 498, Achim Porzelle, Ian Gilbert, MERCK SERENO, Beatriz Baragaña, Medicines for Malaria Venture,  University of Dundee, Neil Norcross, 1469439-69-7, 1469439-71-1 , SUCCINATE

Fc1ccc2nc(cc(c2c1)C(=O)NCCN1CCCC1)-c1ccc(cc1)CN1CCOCC1


Filed under: Malaria, Preclinical drugs Tagged: 1469439-69-7, 1469439-71-1, Achim Porzelle, Beatriz Baragaña, DDD 107498, DDD 498, DDD107498, DUNDEE, Ian Gilbert, Malaria, Medicines for Malaria Venture, MERCK SERENO, Neil Norcross, preclinical, SUCCINATE, University of Dundee

PF-04745637

$
0
0

str1

 

Graphical abstract: The discovery of a potent series of carboxamide TRPA1 antagonists

PF-04745637

cas 1917294-46-2

MW 509.00, MF C27 H32 Cl F3 N2 O2

Cyclopentanecarboxamide, 1-(4-chlorophenyl)-N-[2-[4-hydroxy-4-(trifluoromethyl)-1-piperidinyl]-3-phenylpropyl]-

rac-1-(4-Chlorophenyl)-N-f2-r4-hvdroxy-4-(trifluoromethyl)piperidin-1-vn-3-phenylpropyDcyclopentanecarboxamide

PRODUCT PATENT WO-2016067143-A1
Applicants: PFIZER INC. [US/US]; 235 East 42nd Street New York, New York 10017 (US)
Inventors: SWAIN, Nigel Alan; (GB).
PRYDE, David Cameron; (GB).
RAWSON, David James; (GB).
RYCKMANS, Thomas; (GB).
SKERRATT, Sarah Elizabeth; (GB).
AMATO, George Salvatore; (US).
MARRON, Brian Edward; (US).
REISTER, Steven Michael; (US).

Image result for PFIZER

TrpA1 is a member of the Transient Receptor Potential (Trp) family of ion channels. It was first described as being activated in response to noxious cold. It is activated by a number of exogenous chemical compounds and some endogenous inflammatory mediators. It has also been reported to be activated in response to mechanical stress.

There is substantial evidence for the involvement of TrpA1 in the physiology of pain, including neuropathic and inflammatory pain, and in pruritus (itch). For example, see:

Bautista, D.M. et al., “TRPA 1: A Gatekeeper for Inflammation” , Annu. Rev. Physiol.2013, 75, 181-200;

Bishnoi, M. & Premkumar, L.S., “Changes in TRP Channels Expression in Painful

Conditions”, Open Pain Journal 2013, 6(Suppl. 1), 10-22;Brederson, J.-D. et al., “Targeting TRP channels for pain relief, Eur. J. Pharmacol.2013, 716, 61-76;

Radresa, O. et al., “Roles of TRPAI in Pain Pathophysiology and Implications for the Development of a New Class of Analgesic Drugs”, Open Pain Journal 2013, 6(Suppl. 1), 137-153; and Toth, B.I. & Biro, T., “TRP Channels and Pruritus” , Open Pain Journal 2013, 6(Suppl.1), 62-80.

There is a continuing interest in finding new compounds that interact with TrpA1.

Image result for SWAIN, Nigel AlanNigel Swain

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2016067143&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

E8 that is 1-(4-chlorophenyl)-/V-[2-(4-hydroxy-4-(trifluoromethyl)piperidin-1-yl)-3-phenylpropyl]-cyclopentanecarboxamide, or a pharmaceutically acceptable salt thereof. This compound is represented by formula (lE).

Example 1

rac-1-(4-Chlorophenyl)-N-f2-r4-hvdroxy-4-(trifluoromethyl)piperidin-1-vn-3-phenylpropyDcyclopentanecarboxamide

Method 1

To a solution of rac-1-(1-amino-3-phenylpropan-2-yl)-4-(trifluoromethyl)piperidin-4-ol (Preparation 2, 50 mg, 0.214 mmol) in DMF (1 mL) was added 1-(4-chlorophenyl)cyclopentanecarboxylic acid (37 mg, 0.165 mmol), DIPEA (0.035 mL, 0.198 mmol) and EDCI (38 mg, 0.198 mmol), followed by HOBt (30 mg, 0.198 mmol) and the reaction was stirred at room temperature for 18 hours. Water was added and the reaction stirred for a further 2 hours. DCM was added with further stirring for 1 hour followed by elution through a phase separation cartridge. The organic filtrate was concentrated in vacuo. The residue was dissolved in MeOH and treated with ethereal HCI with standing for 18 hours. The resulting suspension was filtered and triturated with EtOAc, heptanes and TBME to afford the title compound as the hydrochloride salt (69 mg, 82%).

1H NMR (400MHz, DMSO-d6): δ ppm 1.50-1.60 (m, 4H), 1.70-1.90 (m, 4H), 2.15-2.25 (m, 2H), 2.40-2.48 (m, 2H), 2.70-2.80 (m, 1 H), 3.05-3.25 (m, 6H), 3.47-3.62 (m, 2H), 6.38 (br s, 1 H), 7.20-7.40 (m, 9H), 7.80 (br m, 1 H).

MS m/z 509 [M+H]+

Example 1 may also be prepared according to the following method:

A mixture of 1-(4-chlorophenyl)cyclopentanecarboxylic acid (25.7 g, 114 mmol), 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium-3-oxid-hexafluoro phosphate (49.4 g, 130 mmol) and N,N-diisopropylethylamine (40 mL, 229 mmol) in DMF (475 mL) was stirred at room temperature for 15 minutes. To this mixture was added a solution of 1-(1-amino-3-phenylpropan-2-yl)-4-(trifluoromethyl)piperidin-4-ol (Preparation 2, 31.4 g, 104 mmol) in DMF (200 mL). The reaction was stirred at room temperature for 18 hours before partitioning between EtOAc (600 mL) and saturated aqueous sodium hydrogen carbonatesolution (600 mL). The aqueous layer was washed with EtOAc (2 x 600 mL). The combined organic layers were washed with water (600 mL), brine (600 mL), dried over sodium sulphate and concentrated in vacuo. The residue was purified using silica gel column chromatography eluting with 0: 1 to 1 : 1 EtOAc: heptanes to afford the title compound (44 g, 76%).

1H NMR (400MHz, CDCI3): δ ppm 1.35 (br s, 1 H), 1.49-1.85 (m, 6H), 1.90-1.99 (m, 2H), 2.25-2.55 (m, 7H), 2.56-2.70 (m, 1 H), 2.75-3.00 (m, 4H), 3.23-3.31 (m, 1 H), 5.87 (br s, 1 H), 7.07 (d, 2H), 7.16-7.30 (m, 7H).

MS m/z 509 [M+H]+

Examples 2 and 3

IS) and (R)-1-(4-Chlorophenyl)-N-f2-r4-hvdroxy-4-(trifluoromethyl)piperidin-1-vn-3-phenylpropyl)cyclopentanecarboxamide

Example 2

To a suspension of (S)-1-(1-amino-3-phenylpropan-2-yl)-4-(trifluoromethyl)piperidin-4-ol (Preparation 3, 70 mg, 0.232 mmol) and 1-(4-chlorophenyl)cyclopentanecarboxylic acid (57.3 mg, 0.255 mmol) in acetonitrile (0.8 mL) was added triethylamine (0.133 mL, 0.928 mmol) followed bypropylphosphonic anhydride (50% wt solution in EtOAc, 0.21 mL, 0.35 mmol). The reaction was stirred at room temperature for 1.5 hours after which the solution was purified directly by silica gel column chromatography eluting with 0-30% EtOAc in heptanes to afford the title compound (75 mg, 64%).

[a]D20 = +9.6 in DCM [20 mg/mL]

ee determination:

Column: ChiralTech AD-H, 250×4.6 mm, 5 micron.

Mobile phase A: CO2; Mobile phase B: MeOH with 0.2% ammonium hydroxide Gradient: 5% B at 0.00 mins, 60% B at 9.00 mins; hold to 9.5 mins and return to 5% B at 10 mins. Flow rate 3 mL/min.

Rt = 5.047 minutes, ee = 95%

Example 2 may also be prepared from rac-1-(4-chlorophenyl)-N-{2-[4-hydroxy-4- (trifluoromethyl)piperidin-1-yl]-3-phenylpropyl}cyclopentanecarboxamide(Example 1).

The racemate was separated into two enantiomers using preparative chiral chromatography as described below:

Chiralpak IA, 4.6x250mm, 5 micron.

Mobile phase: Hexane:DCM:EtOH:DEA 90:8:2:0.1

Flow rate: 1 mL/min

Rt = 8.351 minutes and Rt = 10.068 minutes

The first eluting isomer is Example 2: (S)-1-(4-chlorophenyl)-N-{2-[4-hydroxy-4-(trifluoromethyl)piperidin-1-yl]-3-phenylpropyl}cyclopentanecarboxamide. ee = 100% The second eluting isomer is Example 3: (R)-1-(4-chlorophenyl)-N-{2-[4-hydroxy-4-(trifluoromethyl)piperidin-1-yl]-3-phenylpropyl}cyclopentanecarboxamide. ee = 99.62% The compound of Example 2 prepared from the chiral separation method is identical by a-rotation and retention time to the compound of Example 2 prepared as the single enantiomer described above.

MS m/z 509 [M+H]+

1H NMR (400MHz, DMSO-d6): δ 1.30-1.80 (m, 10H), 2.20-2.30 (m, 1 H), 2.35-2.60 (m, 6H), 2.65-2.85 (m, 4H), 3.00-3.15 (m, 1 H), 5.50 (br s, 1 H), 6.95-7.00 (m, 1 H), 7.05-7.15 (m, 2H), 7.20-7.35 (m, 6H) ppm

PAPER

The discovery of a potent series of carboxamide TRPA1 antagonists

D. C. Pryde,*a   B. Marron,b   C. G. West,b   S. Reister,b   G. Amato,b  K. Yoger,b   K. Padilla,b   J. Turner,c   N. A. Swain,a   P. J. Cox,c  S. E. Skerratt,a   T. Ryckmans,d   D. C. Blakemore,a  J. Warmuse and   A. C. Gerlachb  
*Corresponding authors
aPfizer Worldwide Medicinal Chemistry, Neuroscience and Pain Research Unit, Portway Building, Granta Park, Great Abington, UK
bIcagen, Inc., 4222 Emperor Boulevard, Suite 350, Durham, USA
cNeuroscience and Pain Research Unit, Portway Building, Granta Park, Great Abington, UK
dPfizer Worldwide Medicinal Chemistry, Ramsgate Road, Sandwich, UK
ePfizer Worldwide Medicinal Chemistry, Neuroscience and Pain Research Unit, Groton, USA
Med. Chem. Commun., 2016, Advance Article

DOI: 10.1039/C6MD00387G, http://pubs.rsc.org/en/Content/ArticleLanding/2016/MD/C6MD00387G?utm_source=feedburner&utm_medium=feed&utm_campaign=Feed%3A+rss%2FMD+%28RSC+-+Med.+Chem.+Commun.+latest+articles%29#!divAbstract

. Please note PF-6667294 is Compound 4 and PF-4746537 is Compound 8.

A series of potent and selective carboxamide TRPA1 antagonists were identified by a high throughput screen. Structure–activity relationship studies around this series are described, resulting in a highly potent example of the series. Pharmacokinetic and skin flux data are presented for this compound. Efficacy was observed in a topical cinnamaldehyde flare study, providing a topical proof of pharmacology for this mechanism. These data suggest TRPA1 antagonism could be a viable mechanism to treat topical conditions such as atopic dermatitis.

Graphical abstract: The discovery of a potent series of carboxamide TRPA1 antagonists
str1  str2
 hydrochloride salt (69 mg, 82%). 1 H NMR (400 MHz, DMSO-d6): δ ppm 1.50–1.60 (m, 4H), 1.70– 1.90 (m, 4H), 2.15–2.25 (m, 2H), 2.40–2.48 (m, 2H), 2.70–2.80 (m, 1H), 3.05–3.25 (m, 6H), 3.47–3.62 (m, 2H), 6.38 (br s, 1H), 7.20–7.40 (m, 9H), 7.80 (br m, 1H). MS m/z 509 [M + H]+ .

 

Image result for The discovery of a potent series of carboxamide TRPV1 antagonists

Discovery and development of TRPV1 antagonists

https://en.wikipedia.org/wiki/Discovery_and_development_of_TRPV1_antagonists

/////////////PF-04745637, PF 04745637, PF04745637, PFIZER, PRECLINICAL, TRPV1 antagonists,  atopic dermatitis, 1917294-46-2

c1(ccccc1)CC(CNC(=O)C3(c2ccc(cc2)Cl)CCCC3)N4CCC(CC4)(O)C(F)(F)F


Filed under: Preclinical drugs, Uncategorized Tagged: 1917294-46-2, PF-04745637, PF04745637

IPI-549

$
0
0

img

IPI-549

CAS 1693758-51-8
MF : C30H24N8O2
Molecular Weight: 528.576

(S)-2-amino-N-(1-(8-((1-methyl-1H-pyrazol-4-yl)ethynyl)-1-oxo-2-phenyl-1,2-dihydroisoquinolin-3-yl)ethyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide

2-amino-N-[(1S)-1-[8-[2-(1-methylpyrazol-4-yl)ethynyl]-1-oxo-2-phenylisoquinolin-3-yl]ethyl]pyrazolo[1,5-a]pyrimidine-3-carboxamide

Company Infinity Pharmaceuticals Inc.
Description Small molecule inhibitor of phosphoinositide 3-kinase (PI3K) gamma
Molecular Target Phosphoinositide 3-kinase (PI3K) gamma
Mechanism of Action Phosphoinositide 3-kinase (PI3K) gamma inhibitor
Therapeutic Modality Small molecule
Latest Stage of Development Phase I
Standard Indication Solid tumors
Indication Details Treat solid tumors
  • Originator Intellikine
  • Developer Infinity Pharmaceuticals
  • ClassAntineoplastics; Small molecules
  • Mechanism of ActionPhosphatidylinositol 3 kinase delta inhibitors; Phosphatidylinositol 3 kinase gamma inhibitors
  • Phase I Solid tumours

Image result for Intellikine

Most Recent Events

  • 18 Apr 2016 Pharmacodynamics data from a preclinical study in Solid tumours presented at the 107th Annual Meeting of the American Association for Cancer Research (AACR-2016)
  • 01 Dec 2015 Phase-I clinical trials in Solid tumours (Monotherapy, Combination therapy, Late-stage disease, Second-line therapy or greater) in USA (PO)

IPI-549 is a potent and selective phosphoinositide-3-kinase (PI3Kγ) Inhibitor as an Immuno-Oncology Clinical Candidate (Kd = 0.29 nM). Bioactivity data of IPI-549: biochemcial IC50 (nM) for PI3K isoform: 3200 (α); 3500 (β); 16 (γ); and >8400 (δ) respectively. Cellar IC50 (nM) of IPI549 for PI3K isoform: 250 (α); 240 (β); 1.6 (γ); and 180 (δ) respectively. IPI-549 shows >100-fold selectivity over other lipid and protein kinases. IPI-549 demonstrates favorable pharmacokinetic properties and robust inhibition of PI3K-γ mediated neutrophil migration in vivo and is currently in Phase 1 clinical evaluation in subjects with advanced solid tumors.

SCHEMBL16629991.png

Image result for IPI-549

Patent

WO 2015051244

https://www.google.co.in/patents/WO2015051244A1?cl=en

Scheme 1

Scheme 2

Example 1

[00657] Compound 4 was prepared in 3 steps from compound A according to the following procedures:

Compound A was prepared according to Method A. It was coupled to 2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid according to the following procedure: Compound A (27.4 mmol, 1.0 equiv), HOBt hydrate (1.2 equiv), 2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (1.05 equiv) and

EDC (1.25 equiv) were added to a 200 mL round bottomed flask with a stir bar. N,N-Dimethylformamide (50 mL) was added and the suspension was stirred at RT for 2 min. Hunig’s base (4.0 equiv) was added and after which the suspension became homogeneous and was stirred for 22h resulting in the formation of a solid cake in the reaction flask. The solid mixture was added to water (600 mL) and stirred for 3h. The resulting cream colored solid was filtered and washed with water (2 x 100 mL) and dried. The solid was then dissolved in methylene chloride (40 mL) after which trifluoroacetic acid (10 equiv, 20 mL) was added and the reaction was stirred for 30 min at RT after which there is no more starting material by LC/MS analysis. The solution was then concentrated and coevaporated with a mixture of methylene choride/ethanol (1 : 1 v/v) and then dried under high vacuum overnight. The resulting solid was triturated with 60 mL of ethanol for lh and then collected via vacuum filtration. The beige solid was then neutralized with sodium carbonate solution (100 mL) and then transferred to a separatory funnel with methylene chloride (350 mL). The water layer was extracted with an additional 100 mL of methylene chloride. The combined organic layers were dried over sodium sulfate, filtered and concentrated under vacuum to provide a pale yellow solid that was purified using flash silica gel chromatography (Combiflash, 24g column, gradient of 0-5% methanol/methylene chloride) to provide amide B. ESI-MS m/z: 459.4 [M+H]+.

[00658] Amide B was placed in a sealed tube (0.67 mmol, 1.0 equiv) followed by dichlorobis(acetonitrile)palladium (15 mol%), X-Phos (45 mol%), and cesium carbonate (3.0 equiv) Propionitrile (5 mL) was added and the mixture was bubbled with Ar for 1 min. 4-Ethynyl-l -methyl- lH-pyrazole (1.24 equiv) was added and the resulting orange mixture was sealed and stirred in an oil bath at 85 oC for 1.5h. The resulting brownish-black mixture was allowed to cool at which point there was no more SM by LC/MS analysis. The mixture was then filtered through a short plug of cotton using acetonitrile and methylene chloride. The combined filtrates were concentrated onto silica gel and purified using flash silica gel chromatography (Combiflash, 4g column, gradient of 0-5% methylene chloride/methanol). The resulting material was further purified by reverse phase HPLC (15-90%o acetonitrile with 0.1%o formic acid/water with 0.1%o formic water) to provide desired compound 4. ESI-MS m/z: 529.5 [M+H]+.

PAPER

WO 2015143012

https://www.google.com/patents/WO2015143012A1?cl=en

PAPER

IPI-549 NMR 1H

IPI-549 13C NMR

IPI-549 ASSAY

Compound 1 is coupled to 4-ethynyl-1-methyl-1H-pyrazole using the general procedure outlined above to provide compound 26 IPI-549, in 70% yield with >98% enantiomeric purity.

IPI-549

1H NMR (400 MHz, DMSO-d6) δ 8.92 (dd, J = 6.8, 1.7 Hz, 1H), 8.55 (dd, J = 4.5, 1.7 Hz, 1H), 8.00 (d, J=6.8 Hz, 1H), 8.00 (s, 1H), 7.69 – 7.54 (m, 5H), 7.53 – 7.43 (m, 3H), 7.41 – 7.35 (m, 1H), 7.01 (dd, J = 6.7, 4.5 Hz, 1H), 6.74 (s, 1H), 6.42 (s, 2H), 4.56 (quin, J = 6.8 Hz, 1H).), 3.82 (s, 3H), 1.35 (d, J = 6.8 Hz, 3H).

13C NMR (101 MHz, DMSO-d6) δ 162.73, 161.19, 160.93, 150.06, 147.51, 146.74, 141.05, 138.09, 137.81, 135.42, 133.66, 132.56, 131.90, 129.51, 129.24, 129.20, 129.17, 128.50, 126.16, 123.41, 123.31, 107.88, 102.44, 101.15, 90.40, 87.06, 85.94, 44.88, 38.62, 20.69.

ESI-HRMS: calcd for 529.2095 C30H25N8O2 (M+H)+ , found 529.2148.

[]D 22: +447.8o (c 1.007, DMSO)

COMPD1

compound 1 in 95% yield.

1H NMR (400 MHz, CDCl3) 8.41 (dd, J = 6.8, 1.7 Hz, 1H), 8.37 (dd, J = 4.4, 1.7 Hz, 1H), 7.90 (d, J = 7.0 Hz, 1H), 7.50-7.34 (m, 5H), 7.34-7.27 (m, 2H), 6.76 (dd, J = 7.1, 4.9 Hz, 1H), 6.57 (s, 1H), 5.54 (broad s, 2H), 4.79 (quin, J = 6.9 Hz, 1H), 1.36 (d, J = 6.5 Hz, 3H);

ESI-HRMS: calcd for C24H20ClN6O2 459.1331 (M+H)+ , found 459.1386. HPLC Purity: 96% AUC.

Abstract Image

Optimization of isoquinolinone PI3K inhibitors led to the discovery of a potent inhibitor of PI3K-γ (26 or IPI-549) with >100-fold selectivity over other lipid and protein kinases. IPI-549 demonstrates favorable pharmacokinetic properties and robust inhibition of PI3K-γ mediated neutrophil migration in vivo and is currently in Phase 1 clinical evaluation in subjects with advanced solid tumors.

Discovery of a Selective Phosphoinositide-3-Kinase (PI3K)-γ Inhibitor (IPI-549) as an Immuno-Oncology Clinical Candidate

http://pubs.acs.org/doi/abs/10.1021/acsmedchemlett.6b00238

Image result for IPI-549

CLIP

Infinity Expands Pipeline with Addition of IPI-549, an Immuno-Oncology Development Candidate for the Treatment of Solid Tumors

– IPI-549, a Selective PI3K-Gamma Inhibitor, Targets the Immune-Suppressive Tumor Microenvironment –

– Preclinical Data for IPI-549 Presented at CRI-CIMT-EATI-AACR – The Inaugural International Cancer Immunotherapy Conference –

September 18, 2015 07:41 AM Eastern Daylight Time

CAMBRIDGE, Mass.–(BUSINESS WIRE)–Infinity Pharmaceuticals, Inc. (NASDAQ: INFI) today announced the expansion of its pipeline with the addition of IPI-549, an orally administered immuno-oncology development candidate that selectively inhibits phosphoinositide-3-kinase gamma (PI3K-gamma), for the treatment of solid tumors. Preclinical data demonstrating the potential of IPI-549 to disrupt the immune-suppressive tumor microenvironment and enable a heightened anti-tumor immune response are being presented today at CRI-CIMT-EATI-AACR – The Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival Meeting in New York City. IPI-549 was discovered at Infinity and is expected to enter Phase 1 clinical development in early 2016.

“Infinity is committed to developing first-in-class and best-in-class medicines, and the expansion of our pipeline with the addition of IPI-549 represents an important step toward fulfilling our vision of building a sustainable biopharmaceutical company that brings meaningful medicines to patients”

Tweet this

“Infinity is committed to developing first-in-class and best-in-class medicines, and the expansion of our pipeline with the addition of IPI-549 represents an important step toward fulfilling our vision of building a sustainable biopharmaceutical company that brings meaningful medicines to patients,” stated Vito Palombella, Ph.D., Infinity’s chief scientific officer. “Infinity’s ability to internally develop a selective PI3K-gamma inhibitor provides us with a unique opportunity to explore the impact that PI3K-gamma inhibition has on disrupting the tumor microenvironment. We look forward to initiating the first clinical study of IPI-549 in patients with solid tumors.”

“I have had the pleasure of collaborating with Infinity’s discovery team and am excited to have worked with IPI-549 in my laboratory,” Jedd Wolchok, M.D., Ph.D., chief of Melanoma and Immunotherapeutics Service, Lloyd J. Old/Ludwig Chair in Clinical Investigation Department of Medicine and Ludwig Center, at Memorial Sloan Kettering Cancer Center and the principal investigator for the planned Phase 1 clinical study of IPI-549. “IPI-549 is a novel, small molecule immuno-oncology agent, and I am looking forward to leading the Phase 1 study for this program.”

IPI-549 inhibits immune suppressive macrophages within the tumor microenvironment, whereas other immunotherapies such as checkpoint modulators more directly target immune effector cell function. As such, IPI-549 may have the potential to treat a broad range of solid tumors and represents a potentially complementary approach to restoring anti-tumor immunity in combination with other immunotherapies such as checkpoint inhibitors.

Preclinical Data for IPI-549 Presented at CRI-CIMT-EATI-AACR – The Inaugural International Cancer Immunotherapy Conference

Today at the AACR meeting in New York City Infinity researchers are presenting preclinical data for IPI-549 in a poster entitled, “The potent and selective phosphoinositide-3-kinase-gamma inhibitor, IPI-549, inhibits tumor growth in murine syngeneic solid tumor models through alterations in the immune suppressive microenvironment.”

In vitro data showed that IPI-549 blocks both the migration of murine myeloid cells and the differentiation of myeloid cells to the M2 phenotype, which is a type of myeloid cell known to promote cancer growth and suppress anti-tumor immune responses. In vivo data in murine solid tumor models demonstrated that IPI-549 treatment also decreased tumor-associated myeloid cells found in the immune suppressive microenvironment. Additionally, IPI-549 treatment increased the number of intratumoral CD8+T-cells, which are known to play a role in inhibiting tumor growth.

IPI-549 has demonstrated dose-dependent, single-agent, anti-tumor activity in multiple solid tumor models, including murine models of lung, colon and breast cancer. Additionally, mice treated with IPI-549 in combination with checkpoint inhibitors showed greater tumor growth inhibition than either treatment as a monotherapy. Preclinical in vivo data also demonstrated that T-cells are required for the anti-tumor activity of IPI-549, which is a hallmark of immunotherapy.

Further details about the IPI-549 development program will be provided at Infinity’s R&D Day on Tuesday, October 6, 2015. R&D Day will be held in New York City from 7:30 a.m. to 12:00 p.m. ET. The event will be webcast beginning at 8:00 a.m. ET and can be accessed in the Investors/Media section of Infinity’s website, www.infi.com. A replay of the event will also be available.

Infinity is also developing duvelisib, an investigational, oral, dual inhibitor of PI3K-delta and PI3K-gamma. The PI3K pathway is also known to play a critical role in regulating the growth and survival of certain types of blood cancers. The investigational agent is being evaluated in registration-focused studies, including DYNAMOTM, a Phase 2 study in patients with refractory indolent non-Hodgkin lymphoma, DYNAMO+R, a Phase 3 study in patients with previously treated follicular lymphoma, and DUOTM, a Phase 3 study in patients with relapsed/refractory chronic lymphocytic leukemia. Duvelisib is an investigational compound and its safety and efficacy have not been evaluated by the U.S. Food and Drug Administration or any other health authority.

About Infinity Pharmaceuticals, Inc.

Infinity is an innovative biopharmaceutical company dedicated to discovering, developing and delivering best-in-class medicines to people with difficult-to-treat diseases. Infinity combines proven scientific expertise with a passion for developing novel small molecule drugs that target emerging disease pathways. For more information on Infinity, please refer to the company’s website at www.infi.com.

Clip

IPI-549-01-A phase 1/1b first in human study of IPI-549, a PI3K-γ inhibitor, as monotherapy and in combination with pembrolizumab in subjects with advanced solid tumors.

Subcategory:
Category:
Developmental Therapeutics—Immunotherapy
Session Type and Session Title:
Poster Session, Developmental Therapeutics—Immunotherapy
Abstract Number: TPS3111
Poster Board Number:
Board #425a
Citation:
J Clin Oncol 34, 2016 (suppl; abstr TPS3111)
Author(s):
Anthony W. Tolcher, David S. Hong, Ryan J. Sullivan, James Walter Mier, Geoffrey Shapiro, Joseph Pearlberg, Les H. Brail, Jahnavi Kharidia, Lixin Han, Claudio Dansky Ullmann, Howard Marvin Stern, Jedd D. Wolchok; START San Antonio, San Antonio, TX; Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, TX; Massachusetts General Hospital, Boston, MA; Department of Medicine, Dana-Farber/Harvard Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA; Dana-Farber Cancer Institute, Boston, MA; Infinity Pharmaceuticals, Inc., Cambridge, MA; Infinity Pharmaceuticals Inc., Cambridge, MA; Infinity Pharmaceuticals, Cambridge, MA; Memorial Sloan Kettering Cancer Center, New York, NY

Abstract Disclosures

Abstract:

Background: IPI-549 is a potential first-in-class potent and selective PI3K-γ inhibitor being developed as a novel orally administered immuno-oncology therapeutic in multiple cancer indications. Preclinical studies demonstrate a role for PI3K-γ in polarization of immune suppressive myeloid cells in the tumor microenvironment. Inhibition of PI3K-γ by IPI-549 enhanced antitumor immune responses and inhibited tumor growth in syngeneic solid tumor preclinical models. In addition, IPI-549 in combination with immune checkpoint inhibitors showed increased tumor growth inhibition compared to each single agent in multiple pre-clinical models. These data served as the scientific foundation for initiating a clinical trial testing IPI-549 as a potential immuno-oncology therapy. This first-in-human clinical study will evaluate the safety and tolerability, and determine the recommended Phase 2 dose (RP2D) of IPI-549 when administered as a monotherapy and in combination with pembrolizumab (NCT02637531) in solid tumors. Methods: This multi-part Phase 1/1b open-label trial will initiate with monotherapy dose escalation cohorts consisting of an accelerated dose escalation phase followed by a standard phase with a 3+3 design. Evaluation of the PK, PD, and safety data in these cohorts will lead to the determination of the maximum tolerated dose (MTD) and RP2D of IPI-549 monotherapy. Subsequently, combination dose escalation cohorts will be initiated in which the combination of IPI-549 and pembrolizumab will be evaluated. Expansion cohorts evaluating the safety, PK, PD, and preliminary clinical activity of IPI-549 as a monotherapy and in combination with pembrolizumab will occur following the dose escalation phase. All subjects in the trial will have advanced and/or metastatic carcinoma or melanoma, and will have failed to respond to standard therapies. Combination expansion cohorts will recruit subjects with non-small cell lung cancer or melanoma who must have received an anti-PD-1 or anti-PD-L1 antibody as their most recent treatment. This trial is currently enrolling patients in the US. Clinical trial information: NCT02637531

REFERENCES

Discovery of a Selective Phosphoinositide-3-Kinase (PI3K)-γ Inhibitor (IPI-549) as an Immuno-Oncology Clinical Candidate
Catherine A. Evans, Tao Liu, André Lescarbeau, Somarajan J. Nair, Louis Grenier, Johan A. Pradeilles, Quentin Glenadel, Thomas Tibbitts, Ann M. Rowley, Jonathan P. DiNitto, Erin E. Brophy, Erin L. O’Hearn, Janid A. Ali, David G. Winkler, Stanley I. Goldstein, Patrick O’Hearn, Christian M. Martin, Jennifer G. Hoyt, John R. Soglia, Culver Cheung, Melissa M. Pink, Jennifer L. Proctor, Vito J. Palombella, Martin R. Tremblay, and Alfredo C. Castro
Publication Date (Web): July 22, 2016 (Letter)
DOI: 10.1021/acsmedchemlett.6b00238

Patent ID Date Patent Title
US2015290207 2015-10-15 HETEROCYCLIC COMPOUNDS AND USES THEREOF
US2015225410 2015-08-13 HETEROCYCLIC COMPOUNDS AND USES THEREOF
US2015111874 2015-04-23 HETEROCYCLIC COMPOUNDS AND USES THEREOF

///////immuno-oncology,  IPI-549,  isoform selectivity,  neutrophil migration,  phosphoinositide-3-kinase,  PI3K-gamma inhibitor, IPI 549,  IPI549. PRECLINICAL

O=C1N(C2=CC=CC=C2)C([C@@H](NC(C3=C(N=CC=C4)N4N=C3N)=O)C)=CC5=CC=CC(C#CC6=CN(C)N=C6)=C51


Filed under: Preclinical drugs Tagged: Immuno-oncology, IPI-549, IPI549. PRECLINICAL, isoform selectivity, neutrophil migration, phosphoinositide-3-kinase, PI3K-gamma inhibitor

(3S)-3-[(2-amino-5-methoxypyrimidin-4-yl)amino]heptan-1-ol for the treatment of viral infections

$
0
0

SCHEMBL13195144.png

str0

(3S)-3-[(2-amino-5-methoxypyrimidin-4-yl)amino]heptan-1-ol

for the treatment of viral infections

Molecular Formula: C12H22N4O2
Molecular Weight: 254.32868 g/mol

To the use of pyrimidine derivatives in the treatment of viral infections, immune or inflammatory disorders, whereby the modulation, or agonism, of toll-like-receptors (TLRs) is involved. Toll-Like Receptors are primary transmembrane proteins characterized by an extracellular leucine rich domain and a cytoplasmic extension that contains a conserved region. The innate immune system can recognize pathogen-associated molecular patterns via these TLRs expressed on the cell surface of certain types of immune cells. Recognition of foreign pathogens activates the production of cytokines and upregulation of co-stimulatory molecules on phagocytes. This leads to the modulation of T cell behaviour.

It has been estimated that most mammalian species have between ten and fifteen types of Toll-like receptors. Thirteen TLRs (named TLR1 to TLR13) have been identified in humans and mice together, and equivalent forms of many of these have been found in other mammalian species. However, equivalents of certain TLR found in humans are not present in all mammals. For example, a gene coding for a protein analogous to TLR10 in humans is present in mice, but appears to have been damaged at some point in the past by a retrovirus. On the other hand, mice express TLRs 11, 12, and 13, none of which are represented in humans. Other mammals may express TLRs which are not found in humans. Other non-mammalian species may have TLRs distinct from mammals, as demonstrated by TLR14, which is found in the Takifugu pufferfish. This may complicate the process of using experimental animals as models of human innate immunity.

For detailed reviews on toll-like receptors see the following journal articles. Hoffmann, J. A., Nature, 426, p 33-38, 2003; Akira, S., Takeda, K., and Kaisho, T., Annual Rev. Immunology, 21, p 335-376, 2003; Ulevitch, R. J., Nature Reviews: Immunology, 4, p 512-520, 2004.

Compounds indicating activity on Toll-Like receptors have been previously described such as purine derivatives in WO 2006/117670, adenine derivatives in WO 98/01448 and WO 99/28321, and pyrimidines in WO 2009/067081. However, there exists a strong need for novel Toll-Like receptor modulators having preferred selectivity, higher potency, higher metabolic stability, and an improved safety profile compared to the compounds of the prior art.

In the treatment of certain viral infections, regular injections of interferon (IFNα) can be administered, as is the case for hepatitis C virus (HCV), (Fried et. al. Peginterferon-alfa plus ribavirin for chronic hepatitis C virus infection, N Engl J Med 2002; 347: 975-82). Orally available small molecule IFN inducers offer the potential advantages of reduced immunogenicity and convenience of administration. Thus, novel IFN inducers are potentially effective new class of drugs for treating virus infections. For an example in the literature of a small molecule IFN inducer having antiviral effect see De Clercq, E.; Descamps, J.; De Somer, P. Science 1978, 200, 563-565.

IFNα is also given in combination with other drugs in the treatment of certain types of cancer (Eur. J. Cancer 46, 2849-57, and Cancer Res. 1992, 52, 1056). TLR 7/8 agonists are also of interest as vaccine adjuvants because of their ability to induce pronounced Th1 response (Hum. Vaccines 2010, 6, 1-14; Hum. Vaccines 2009, 5, 381-394).

50d

Paper

Abstract Image

Toll-like receptor (TLR) 7 and 8 agonists can potentially be used in the treatment of viral infections and are particularly promising for chronic hepatitis B virus (HBV) infection. An internal screening effort identified a pyrimidine Toll-like receptor 7 and 8 dual agonist. This provided a novel alternative over the previously reported adenine and pteridone type of agonists. Structure–activity relationship, lead optimization, in silico docking, pharmacokinetics, and demonstration of ex vivo and in vivo cytokine production of the lead compound are presented.

Novel Pyrimidine Toll-like Receptor 7 and 8 Dual Agonists to Treat Hepatitis B Virus

Janssen Infectious Diseases Diagnostics BVBA, Turnhoutseweg 30, 2340 Beerse, Belgium
Villapharma Research S.L., Parque Tecnológico de Fuente Álamo. Ctra. El Estrecho-Lobosillo, Km. 2.5, Av. Azul 30320 Fuente Álamo de Murcia, Murcia, Spain
J. Med. Chem., 2016, 59 (17), pp 7936–7949
*For D.M.: phone, +32 6414 1019; E-mail, dmcgowan@its.jnj.com., *For F.H.: phone, +32 6414 1644; E-mail, fherschk@its.jnj.com.
PATENT
Preparation of Compounds.
Compounds of formula (I), where R1 is hydrogen atom are prepared according to scheme 1
Figure US20140045849A1-20140213-C00006
Figure US20140045849A1-20140213-C00007
Figure US20140045849A1-20140213-C00008
Figure US20140045849A1-20140213-C00009
Figure US20140045849A1-20140213-C00010
Compounds of formula (I), when R1 is alkyl, cycloalkyl, trifluoromethyl, or alkoxy and where R2 is aromatic or aliphatic, can be prepared according scheme 4. This reaction scheme begins with a crossed-Claisen reaction where an acyl chloride reacts with ester intermediate A (shown in scheme 1) to form intermediates (G) as in scheme 3. From intermediate G, the reaction scheme follows the same pathway to the products as in scheme 3. This is a general scheme using methods known to a skilled person, see for instance The Journal of American Chemical Society volume 127, page 2854 (2005).
Synthetic Scheme for the Preparation of AA-9
Figure US20140045849A1-20140213-C00028
A solution of AA-6 (38 g, 116.75 mmol) and 10% Pd/C in methanol (200 mL) was hydrogenated under 50 PSI hydrogen at 50° C. for 24 hours. The reaction mixture was filtered and the solvent was evaporated to give crude product AA-7 (17 g).
The crude product was dissolved in dichloromethane (200 mL), triethylamine (26.17 g, 259.1 mmol) and di-tert-butyl dicarbonate (84.7 g, 194.4 mmol) was added at 0° C. The resulting mixture was stirred at room temperature for 16 hours. The mixture was partitioned between dichloromethane and water. The organic phase was washed with brine, dried and evaporated. The residue was purified by silica gel chromatography eluting with 20% ethyl acetate in petroleum ether to give AA-8 (13 g) as colorless oil.
1H NMR (400 MHz, CDCl3): δ ppm 4.08-4.03 (br, 1H), 3.68 (m, 1H), 3.58-3.55 (m, 2H), 3.20-2.90 (br, 1H), 1.80-1.73 (m, 1H), 1.42-1.17 (m, 15H), 0.85-0.82 (t, J=6.8 Hz, 3H).
AA-8 (42 g, 0.182 mol) was dissolved in dioxane (200 mL) and dioxane/HCl (4M, 200 mL) was added at 0° C. The resulting mixture was stirred at room temperature for 2 h. The solvent was evaporated to afford the crude product. A dichloromethane/petroleum ether mixture (50 mL, 1:1, v/v) was added to the crude product, and the supernatant was decanted. This procedure was repeated two times to obtain an oil, AA-9 (26.6 g).
1H NMR (400 MHz, DMSO-d6): δ ppm 8.04 (s, 3H), 3.60-3.49 (m, 2H), 3.16-3.15 (m, 1H), 1.71-1.67 (m, 2H), 1.60-1.55 (m, 2H), 1.33-1.26 (m, 4H), 0.90-0.87 (t, J=6.8 Hz, 3H)
Procedure for Preparation of Intermediate C-1.Reaction Scheme:
Figure US20140045849A1-20140213-C00013
A suspension of intermediate B-1 (160 g, 0.74 mol) in POCl3 (900 mL) was heated to 100° C. under N2 with stirring for 5 hours. The reaction mixture was cooled to room temperature. The excess POCl3 was removed under reduced pressure, the oil residue was poured into cold, sat. aq. NaHCO3 (2 L) that was stirred for 30 minutes. The mixture was extracted with ethyl acetate (3×1.5 L). The combined organic layers were separated and washed with brine (1 L), dried over sodium sulfate, the solids were removed via filtration, and the solvents of the filtrate were concentrated to afford intermediate C-1 (70 g) as a yellow solid. The product was used in the next step without further purification.
Patent ID Date Patent Title
US2015274676 2015-10-01 ACYLAMINOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF VIRAL INFECTIONS AND FURTHER DISEASES
US2014045849 2014-02-13 PYRIMIDINE DERIVATIVES FOR THE TREATMENT OF VIRAL INFECTIONS

///////////////  treatment of viral infections, Pyrimidine Toll-like Receptor 7,  Toll-like Receptor 8 Dual Agonists, Treat Hepatitis B Virus, PRECLINICAL

n1c(nc(c(c1)OC)N[C@@H](CCCC)CCO)N


Filed under: Preclinical drugs Tagged: preclinical, Pyrimidine Toll-like Receptor 7, Toll-like Receptor 8 Dual Agonists, Treat Hepatitis B Virus, treatment of viral infections

JNJ 54166060

$
0
0

SCHEMBL16035899.png

img

JNJ 54166060

JNJ-54166060; JNJ 54166060; JNJ54166060.

(R)-(2-chloro-3-(trifluoromethyl)phenyl)(1-(5-fluoropyridin-2-yl)-4-methyl-1,4,6,7-tetrahydro-5H-imidazo[4,5-c]pyridin-5-yl)methanone

[2-chloro-3-(trifluoromethyl)phenyl]-[(4R)-1-(5-fluoropyridin-2-yl)-4-methyl-6,7-dihydro-4H-imidazo[4,5-c]pyridin-5-yl]methanone

CAS 1627900-42-8
Chemical Formula: C20H15ClF4N4O
Exact Mass: 438.0871

JNJ-54166060 is a potent P2X7 antagonist. Bioactivity data of JNJ-54166060: rP2X7 IC50=4 nM; rP2X7 IC50=115nM; HLM/RLM = 0.35/0.64, ED50 = 2.3 mg/kg in rats. JNJ-54166060 shows high oral bioavailability and low-moderate clearance in preclinical species, acceptable safety margins in rats, and a predicted human dose of 120 mg of QD. Additionally, JNJ-54166060 possesses a unique CYP profile and was found to be a regioselective inhibitor of midazolam CYP3A metabolism.

The P2X7 receptor is a ligand-gated ion channel and is present on a variety of cell types, largely those known to be involved in the inflammatory and/ or immune process, specifically, macrophages and monocytes in the periphery and predominantly in glial cells (microglia and astrocytes) of the CNS. (Duan and Neary, Glia 2006, 54, 738-746; Skaper et al., FASEB J 2009, 24, 337-345;

Surprenant and North, Annu. Rev. Physiol. 2009, 71, 333-359). Activation of the P2X7 receptor by extracellular nucleotides, in particular adenosine triphosphate, leads to the release of proinflammatory cytokines IL-1 β and IL-18 (Muller, et. Al. Am. J. Respir. Cell Mol. Biol. 201 1 , 44, 456-464), giant cell formation

(macrophages/ microglial cells), degranulation (mast cells) and L-selectin shedding (lymphocytes) (Ferrari et al., J. Immunol. 2006, 176, 3877-3883; Surprenant and North, Annu. Rev. Physiol. 2009, 71, 333-359). P2X7 receptors are also located on antigen-presenting cells (keratinocytes, salivary acinar cells (parotid cells)), hepatocytes, erythrocytes, erythroleukaemic cells, monocytes, fibroblasts, bone marrow cells, neurones, and renal mesangial cells.

The importance of P2X7 in the nervous system arises primarily from experiments using P2X7 knock out mice. These mice demonstrate the role of P2X7 in the development and maintenance of pain as these mice were protected from the development of both adjuvant-induced inflammatory pain and partial nerve ligation induced neuropathic pain (Chessell et al., Pain 2005, 114, 386-396). In addition P2X7 knock out mice also exhibit an anti-depressant phenotype based on reduced immobility in forced swim and tail suspension tests (Basso et al., Behav. Brain Res. 2009, 798, 83-90.). Moreover, the P2X7 pathway is linked to the release of the pro-inflammatory cytokine, IL-1 β, which has been linked to precipitation of mood disorders in humans (Dantzer, Immunol. Allergy Clin. North Am. 2009, 29, 247-264; Capuron and Miller, Pharmacol. Ther. 201 1 , 730, 226-238). In addition, in murine models of Alzheimer’s disease, P2X7 was upregulated around amyloid plaques indicating a role of this target in such pathology as well (Parvathenani et al., J. Biol. Chem. 2003, 278, 13309-13317).

In view of the clinical importance of P2X7, the identification of compounds that modulate P2X7 receptor function represents an attractive avenue into the development of new therapeutic agents. Such compounds are provided herein.

PAPER

Identification of (R)-(2-Chloro-3-(trifluoromethyl)phenyl)(1-(5-fluoropyridin-2-yl)-4-methyl-6,7-dihydro-1H-imidazo[4,5-c]pyridin-5(4H)-yl)methanone (JNJ 54166060), a Small Molecule Antagonist of the P2X7 receptor

Janssen Pharmaceutical Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121 United States
Janssen Research & Development, Discovery Sciences, A Division of Janssen-Cilag, Jarama 75, 45007 Toledo, Spain
J. Med. Chem., Article ASAP
*Tel: 1-858-320-3306. E-mail: dswanso1@its.jnj.com.
Abstract Image

The synthesis and SAR of a series of 4,5,6,7-tetrahydro-imidazo[4,5-c]pyridine P2X7 antagonists are described. Addressing P2X7 affinity and liver microsomal stability issues encountered with this template afforded methyl substituted 4,5,6,7-tetrahydro-imidazo[4,5-c]pyridines ultimately leading to the identification of 1 (JNJ 54166060). 1 is a potent P2X7 antagonist with an ED50 = 2.3 mg/kg in rats, high oral bioavailability and low-moderate clearance in preclinical species, acceptable safety margins in rats, and a predicted human dose of 120 mg of QD. Additionally, 1 possesses a unique CYP profile and was found to be a regioselective inhibitor of midazolam CYP3A metabolism.

PATENT

https://www.google.com/patents/WO2014152604A1?cl=en

Example 40. (R* -(2-chloro-3-(trifluoromethyl phenyl (l-(5-fluoropyridin-2-yl -4-methyl- -dihydro-lH-imidazor4,5-c1pyridin-5(4H -yl methanone.

Figure imgf000145_0001

The title compound, absolute configuration unknown, was obtained as a single enantiomer by Chiral SFC purification of Example 11 performed using CHIRALCEL OD-H (5μιη, 250x20mm) and a mobile phase of 70% CO2, 30% EtOH. The enantiomeric purity was confirmed by analytical SFC using a CHIRALCEL OD-H (250×4.6mm) and a mobile phase of 70% CO2, 30% EtOH over 7 minutes. (100% single enantiomer, 2.29 min retention time). MS (ESI): mass calculated for C2oH15ClF4N40, 438.1; m/z found, 439.3 [M+H]+.

Example 11. (2-Chloro-3-(trifluoromethyl)phenyl)(l-(5-fluoropyridin-2-yl)-4-methyl-6,7- dihvdro-lH-imidazor4,5-c1pyridin-5(4H)-yl)methanone.

Figure imgf000128_0001

Step A. (2-Chloro-3 -(trifluoromethyl)phenylX 1 -(5 -fluoropyridin-2-yl)-4-methyl- 1 H- imidazor4.5-c1pyridin-5(4H)-yl)methanone.

To a solution of Intermediate 1 (0.70 g, 3.27 mmol) in THF (20 mL) was added

Intermediate 12 (0.87 g, 3.60 mmol) dropwise. The reaction was allowed to stir for 1 h then cooled to – 78 °C. To the cooled solution was added 3M MeMgBr in Et20 (1.31 mL, 3.92 mmoL) and the reaction was let come to room temperature. The mixture was then quenched with IN NaOH (50 mL) and extracted with EtOAc (3 x 30 mL). The organic layers were combined, dried (Na2S04), and concentrated. Chromatography of the resulting residue (Si02; MeOH (NH3):DCM) gave the title compound (770 mg, 54%). XH NMR (400 MHz, CDC13) δ 8.43 – 8.34 (m, 1H), 7.92 – 7.73 (m, 2H), 7.70 – 7.33 (m, 4H), 6.08 (dtd, J = 19.7, 11.7, 8.0 Hz, 3H), 1.54 (t, J = 7.0 Hz, 3H). MS (ESI): mass calculated for C2oH13ClF4 40, 436.07; m/z found 437.1 [M+H]+.

Step B. (2-Chloro-3-(trifluoromethyl)phenyl)(l-(5-fluoropyridin-2-yl)-4-methyl-6J- dihydro-lH-imidazo[4.5-clpyridin-5(4H)-yl)methanone.

To a solution of (2-chloro-3-(trifluoromethyl)phenyl)(l-(5-fluoropyridin-2-yl)-4-methyl- lH-imidazo[4,5-c]pyridin-5(4H)-yl)methanone (0.80 g, 1.83 mmol) in degassed EtOH (25 mL) was added 10% palladium on carbon (0.20 g, 0.19 mmol). The reaction was placed under an atmosphere of hydrogen and let stir for 48 h. The reaction was diluted with DCM and filtered through a pad of Celite ©. The solvent was concentrated and chromatography of the resulting residue (Si02; MeOH (NH3):DCM) gave the title compound (500 mg, 62%). ¾ NMR (500 MHz, CDC13) δ 8.45 – 8.30 (m, 1H), 7.94 (dd, J = 18.2, 10.7 Hz, 1H), 7.76 (d, J = 5.7 Hz, 1H), 7.67 – 7.43 (m, 3H), 7.43 – 7.30 (m, 1H), 5.81 (dd, J = 13.3, 6.7 Hz, 1H), 5.07 (d, J = 5.6 Hz, 1H), 4.52 (d, J = 6.7 Hz, 1H), 3.61 – 3.31 (m, 1H), 3.08 – 2.69 (m, 1H), 1.63 – 145 (m, 3H). MS (ESI): mass calculated for C20H15ClF4N4O, 438.08; m/z found 439.1 [M+H]+.

Intermediate 12: 2-Chloro-3-(trifluoromethyl)benzoyl chloride.

Figure imgf000122_0001

To a suspension of 2-chloro-3-(trifluoromethyl)benzoic acid (15 g, 67 mmol) and catalytic DMF (0.06 mL, 0.67 mmol) in DCM (150 mL) was added oxalyl chloride (6.8 mL, 80 mmol) dropwise. The reaction was let stir (vigorous bubbling) for 4 h and concentrated to an oily solid which became solid after overnight drying on high vacuum.

Intermediate 1 : l-(5-Fluoropvridin-2-vl)-lH-imidazor4,5-clpvridine.

Figure imgf000118_0001

A solution of 5-azabenzimidazole (1.00 g, 8.40 mmol), 2-bromo-5-fluoropyridine (1.48 g, 8.40 mmol), copper (I) oxide (0.13 g, 0.84 mmol), 8-hydroxyquinoline (0.24 g, 1.68 mmol), and CS2CO3 (5.47 g, 16.8 mmol) in DMSO (4 mL) was irradiated in a microwave apparatus for 1 hour at 140 °C. The reaction was diluted with H2O (100 mL) and extracted with EtOAc (75 mL x 3). The organic layers were combined, dried (Na2S04), and concentrated. Chromatography of the resulting residue (S1O2; MeOH (NH3):DCM) gave the title compound (0.45 g, 25%). MS (ESI): mass calculated for C11H7FN4, 214.07; m/z found 215.1 [M+H]+.

Patent ID Date Patent Title
US2016039809 2016-02-11 P2X7 MODULATORS
US2015322062 2015-11-12 P2X7 MODULATORS
US2014275015 2014-09-18 P2X7 MODULATORS

///////JNJ 54166060, JNJ-54166060,  JNJ54166060, 1627900-42-8, P2X7 antagonists

ClC1=C(C(N2CCC(N(C3=CC=C(F)C=N3)C=N4)=C4[C@H]2C)=O)C=CC=C1C(F)(F)F


Filed under: Preclinical drugs Tagged: 1627900-42-8, JNJ 54166060, JNJ54166060, P2X7 antagonists

Synthesis of 4-Heteroaryl–Quinazoline Derivatives as Potential Anti-breast Cancer Agents

$
0
0

Image result for Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt

Banner

Figure 1.

Figure 2.

Ethyl 2-[(6,7-dimethoxyquinazolin-4-yl)amino]-5,6,7,8-tetrahydro-4H-cyclohepta[b]thiophene-3-carboxylate (15b)

Yield: 76%; mp: 254–256°C; IR (cm−1): 3200 (NH), 2974, 2854 (CH-aliphatic), 1656 (C=O); 1H NMR (DMSO-d6) δ ppm 1.03 (t, 3H, CH3CH2, J = 7.2 Hz), 1.21 (t, 2H, CH2, J = 6.9 Hz), 2.88 (t, 2H, CH2, J = 6.9 Hz), 3.40 (q, 2H, CH2, J = 6.9 Hz), 3.88 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 3.96 (m, 4H, 2 CH2), 4.24 (q, 2H, CH3CH2, J = 7.2 Hz), 7.40 (s, 1H, Ar-H), 7.62 (s, 1H, Ar-H), 8.99 (s, 1H, Ar-H), 12.00 (s, 1H, NH, D2O exchangeable); Anal. Calcd for C22H25N3O4S: C, 61.81; H, 5.89; N, 9.83. Found: C, 61.93; H, 5.96; N, 9.98.

General procedure for the synthesis of compounds 15a,15b

A mixture of 4-chloro-6,7-dimethoxyquinazoline (1) (0.22 g, 1 mmol) and ethyl 2-amino-4,5,6,7-tetrahydro-1-benzothiophene-3-carboxylate (14a) or ethyl 2-amino-5,6,7,8-tetrahydro-4H-cyclohepta[b]thiophene-3-carboxylate (14b) (1 mmol) in isopropanol (15 mL) was heated under reflux for 10 h. The reaction was cooled, and the solid formed was filtered, dried, and crystallized from isopropanol.

Synthesis of 4-Heteroaryl–Quinazoline Derivatives as Potential Anti-breast Cancer Agents

A. E. Kassab, E. M. Gedawy, H. B. El-Nassan+

+Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt

E-mail: hala_bakr@hotmail.com

str1

Asmaa Elsayed Abd Ellatief Kassab( A. E. Kassab)

4-Heteroaryl or heteroalkyl–quinazoline derivatives were prepared as dual epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor-2 (VEGFR-2) inhibitors. The new compounds were tested for their dual enzyme inhibition as well as their cytotoxic activity on MCF7 cell line. The results indicated that almost all the compounds showed moderate dual inhibition of both enzymes. Compound 3 (methyl piperidine-4-carboxylate derivative) showed the highest inhibitory activity against both enzymes with IC50 97.6 and 64.0 µM against EGFR and VEGFR-2 kinases, respectively. Most of the test compounds showed potent to moderate antitumor activity on MCF7 cell line. Five compounds (3, 9c, 11, 13, and 15b) showed potent cytotoxic activity with IC50values between 10 and 17 µM.

Scheme 4.

Scheme 4.

Scheme 3.

Scheme 3.

Scheme 2.

Scheme 2.

Scheme 1.

Scheme 1.

Map of cairo university

Image result for UNIVERSITY OF CAIRO

CAIRO UNIVERSITY

LIBRARY.CAIRO UNIV


DOWNTOWN CAIRO

Image result for Faculty of Pharmacy, Cairo University

Image result for Faculty of Pharmacy, Cairo University


Dean of faculty of pharmacy, Cairo University, Dr. Azza Agha during the opening of the first international day at Faculty of Pharmacy.

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

//////////4-Heteroaryl–Quinazoline Derivatives,  Anti-breast Cancer Agents


Filed under: Preclinical drugs Tagged: 4-Heteroaryl–Quinazoline Derivatives, Anti-breast Cancer Agents

Novel Autotaxin Inhibitors for the Treatment of Osteoarthritis Pain from Lilly Research Laboratories

$
0
0

SCHEMBL15875396.png

str1Figure imgf000023_0002

2-(2-(1H-1,2,3-triazol-5-yl)ethoxy)-1-(2-((2,3-dihydro-1H-inden-2-yl)amino)-5,7-dihydro-6Hpyrrolo[3,4-d]pyrimidin-6-yl)ethan-1-one

l-[2-(2,3-dihydro- lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]-2-[2-(lH- l ,2,3-triazol-4-yl)ethoxy]ethanone.

CAS 1619971-30-0

1-[2-(2,3-dihydro-1H-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]-2-[2-(1H-1,2,3-triazol-4-yl)ethoxy]ethanone;
Molecular Formula: C21H23N7O2
Molecular Weight: 405.45302 g/mol

US2014200231

Scheme A

Scheme B

Scheme C

VI

Scheme E

Autotaxin is an enzyme reported to be the source of lysophosphatidic acid (LPA) which up-regulates pain-related proteins through one if its cognate receptors, LPAi. LPA is an intracellular lipid mediator which influences a multiplicity of biological and biochemical processes. Targeted inhibition of autotaxin-mediated LPA biosynthesis may provide a novel mechanism to prevent nerve injury-induced neuropathic pain.

Compounds that inhibit autotaxin are desired to offer a potential treatment option for patients in need of treatment for pain.

Pain associated with osteoarthritis (OA) is reported to be the primary symptom leading to lower extremity disability in OA patients. Over 20 million Americans have been diagnosed with OA, the most common of the arthropathies. The currently approved treatments for OA pain may be invasive, lose efficacy with long term use, and may not be appropriate for treating all patients. Additional treatment options for patients suffering from pain associated with OA are desired. Compounds that inhibit autotaxin represent another possible treatment option for patients with pain associated with OA.

U.S. Patent 7,524,852 (‘852) discloses substituted bicyclic pyrimidine derivatives as anti-inflammatory agents.

PCT/US2011/048477 discloses indole compounds as autotoxin inhibitors.

There is a need for novel compounds that provide autotaxin inhibition. The present invention provides novel compounds which are autotaxin inhibitors. The present invention provides certain novel compounds that inhibit the production of LPA.

Autotaxin inhibitor compounds are desired to provide treatments for autotaxin mediated conditions, such as pain and pain associated with OA.

PAPER

Abstract Image

In an effort to develop a novel therapeutic agent aimed at addressing the unmet need of patients with osteoarthritis pain, we set out to develop an inhibitor for autotaxin with excellent potency and physical properties to allow for the clinical investigation of autotaxin-induced nociceptive and neuropathic pain. An initial hit identification campaign led to an aminopyrimidine series with an autotaxin IC50 of 500 nM. X-ray crystallography enabled the optimization to a lead compound that demonstrated favorable potency (IC50 = 2 nM), PK properties, and a robust PK/PD relationship.

Image result for Lilly Research Laboratories

Novel Autotaxin Inhibitors for the Treatment of Osteoarthritis Pain: Lead Optimization via Structure-Based Drug Design

Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, Indiana 46285, United States
ACS Med. Chem. Lett., 2016, 7 (9), pp 857–861
DOI: 10.1021/acsmedchemlett.6b00207
*E-mail: jonessp@lilly.com. Tel: +1-317-277-5543.

http://pubs.acs.org/doi/abs/10.1021/acsmedchemlett.6b00207

Spencer Jones

Spencer Jones

Senior Research Scientist at Eli Lilly and Company

2-(2-(1H-1,2,3-triazol-5-yl)ethoxy)-1-(2-((2,3-dihydro-1H-inden-2-yl)amino)-5,7-dihydro-6Hpyrrolo[3,4-d]pyrimidin-6-yl)ethan-1-one (9)

………… Purified the resulting residue by silica gel chromatography (gradient elution: 0-9% methanol in ethyl acetate ) to give the title compound……..

1H NMR (400 MHz, CDCl3): 60:40 mixure of rotamers * indicates minor rotamer δ 8.18 (bs, 0.6H), *8.13 (bs, 0.4H), 7.49 (s, 1H), 7.21-7.09 (m, 4 H), 5.70-5.50 (m, 1H), 4.87-4.78 (m, 1H), 4.75 (s, 1.2H), *4.67 (s, 0.8H), 4.64 (s, 1.2H) *4.53 (s, 0.8H), *4.30 (s, 0.8H), 4.28 (s, 1.2H), 3.93 (t, J = 5.6 Hz, 2H), 3.43 (dd, J = 16.2, 7.1 Hz, 2H), 3.10 (t, J = 5.6 Hz, 2H), 2.89 (dd, J = 16.2, 4.9 Hz, 2H).

13C NMR (400 MHz, CDCl3): * indicates minor δ *169.3, 16 169.2, 167.0, *166.8, *162.4, 162.2, 152.8, *152.3, 141.1, 137.8, 130.9, 126.7, 124.9, 115.9, 69.8, 69.3, *69.0, 52.7, *52.5, 51.2, 49.0, *47.9, 40.1, 24.7.

LC/MS (ESI+ ): (m/z) 406 (C21H24N7O2 = (M+1)+ ).

PATENT

WO-2014110000-A1

Example 2

Synthesis of l-[2-(2,3-dihydro- lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]-2-[2-(lH- l ,2,3-triazol-4-yl)ethoxy]ethanone.

Figure imgf000023_0002

Stir a mixture of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid 2,2,2-trifluoroacetic acid

(20.22 g; 70.90 mmol), N-(2,3-dihydro- lH-inden-2-yl)-6,7-dihydro-5H-pyrrolo[3,4- d]pyrimidin-2-amine dihydrochloride hydrate (27.99 g; 81.54 mmol) and triethylamine (98.83 mL; 709.03 mmol) in dimethylformamide (404.40 mL) at 0°C. Add a solution of 1-propanephosphonic acid cyclic anhydride (50% solution in DMF; 51.89 mL; 81.54 mmol) over 30 minutes, and stir the mixture at room temperature for 18 hours.

Concentrate the reaction mixture under reduced pressure to give a residue. Add water (200 mL) and extract the mixture with ethyl acetate (4 x 250 mL) and

dichloromethane (4 x 250 mL). Wash the combined organic layers with saturated aqueous sodium bicarbonate (2 x 100 mL) and brine (100 mL), then dry over anhydrous sodium sulfate. Filter the mixture and concentrate the solution under reduced pressure to give a red solid (25.70 g) that is slurried in ethyl acetate/methanol (9: 1 mixture; 200 mL) for 2 hours at room temperature. Filter the resulting solid and wash with cold ethyl acetate (50 mL) to give a solid (ca.18.2 g) that is re-slurried in ethyl acetate (200 mL) at reflux for 1 hour. On cooling to room temperature, stir the mixture for 1 hour and filter the resulting light pink solid.

Slurry the light pink solid in water/methanol (1 : 1 mixture; 200 mL) and heat the mixture at 50°C for 30 minutes. Add ammonium hydroxide solution (32% ; 50 mL) and continue to heat the mixture at 50°C for 30 minutes. Upon cooling to room temperature, add additional ammonium hydroxide solution (32% ; 50 mL) and continue stirring for 1 hour at room temperature. Filter the resulting light gray solid, dry and slurry again in ethyl acetate (200 mL) for 1 hour to afford a light gray solid that is filtered, washed with ethyl acetate (25 mL), and dried to give the title compound (12.42 g; 43%) as a gray solid. MS (m/z): 406 (M+l).

PATENT

US-20140200231-A1

https://www.google.com/patents/US20140200231

Scheme E

Figure imgf000014_0001

Preparation 7

Synthesis of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid.

Figure imgf000018_0001

Pressurize 1 atmosphere of hydrogen (g) to a flask containing [2-(l-benzyl-lH- l,2,3-triazol-5-yl)ethoxy]acetic acid (10.1 g; 1.00 equiv; 38.66 mmoles) and palladium (II) chloride (3 g; 16.92 mmoles; 3.00 g) in isopropyl alcohol (300 mL) and water (60 mL). Maintain the flask under a hydrogen atmosphere for 3 h, then filter through Celite™ and concentrate. Add toluene (2×50 mL) and concentrate to afford the title compound (7.96 g, 100%). ]H NMR (d6-DMSO): 2.86 (t, / = 7 Hz, 2 H), 3.65 (t, / = 7 Hz, 2 H), 3.98 (s, 2 H), 7,77 (s, 1 H), 13.4 – 13.6 (br s, 2 H).

Example 1

Synthesis of l-[2-(2,3-dihydro-lH-inden-2-ylamino)-7,8-dihydropyrido[4,3-d]pyrimidin- 6(5H)-yl]-2-[2-(lH-l,2,3-triazol-4- l)ethoxy]ethanone.

Figure imgf000018_0002

Add N-indan-2-yl-5,6,7,8-tetrahydropyrido[4,3-d]pyrimidin-2-amine (4.2 g, 15.8 mmol) to a mixture of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid (2.7 g, 15.8 mmol), 1-hydroxybenzotriazole (3.20 g, 23.7 mmol), and dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (5.44 g, 28.4 mmol) in dichloromethane (40 mL) at 25 °C. Add triethylamine (4.40 mL, 31.6 mmol) to the reaction mixture and stir for 16 h. Wash with water (2 x 50 mL) and concentrate the organic layer. Purify by silica gel column chromatography, eluting with ethyl acetate/methanol, to give the title compound (4.0 g, 60%) as a solid. MS (m/z): 420 (M + Η). Preparation 8

Synthesis of 2-chloro-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-7,8-dihydropyrido[4,3- d]pyrimidin-6(5H)-yl]ethanone.

Figure imgf000019_0001

To N-indan-2-yl-5,6,7,8-tetrahydropyrido[4,3-d]pyrimidin-2-amine (11.0 g, 41.3 mmol) and triethylamine (7.48 mL, 53.7 mmol) in dichloromethane (200 mL), add 2- chloroacetyl chloride (3.61 mL, 5.13 g, 45.4 mmol) dropwise over five minutes at 23 °C. Stir for 30 minutes and pour the reaction mixture into 1 : 1 50% saturated aqueous sodium bicarbonate: dichloromethane (75 mL). Separate the organic layer from the aqueous layer and further extract the aqueous layer with dichloromethane (2 x 25 mL). Combine the organic extracts and dry over anhydrous sodium sulfate, filter, and concentrate. Dissolve the residue in chloroform (10 mL) and purify via silica gel column chromatography (gradient elution: 25% ethyl acetate in hexanes to 100% ethyl acetate) to give the title compound (9.75 g, 69%). ]H NMR (CDC13, * = minor amide rotamer) δ 2.77* (t, 2H), 2.84 (dd, 2H), 2.87 (t, 2H), 3.35 (dd, 2H), 3.76 (t, 2H), 3.85* (t, 2H), 4.12 (s, 2H), 4.52* (s, 2H), 4.57 (s, 2H), 4.72-4.82 (m, IH), 5.48-5.64 (m, IH), 7.12-7.21 (m, 4H), 8.03-8.10 (m, IH).

Preparation 9

Synthesis of 2-(but-3-yn-l-yloxy)-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-7,8- dihydropyrido[4,3-d]p rimidin-6(5H)-yl]ethanone.

Figure imgf000019_0002

To sodium hydride (60 wt% in mineral oil, 1.58 g, 39.6 mmol) in tetrahydrofuran (50 mL) at 23 °C, add 3-butyn-l-ol (7.93 g, 8.59 mL, 113.2 mmol) dropwise, then stir at 23 °C for 20 minutes. Add this solution to 2-chloro-l-[2-(2,3-dihydro-lH-inden-2- ylamino)-7,8-dihydropyrido[4,3-d]pyrimidin-6(5H)-yl]ethanone (9.70 g, 28.3 mmol) in tetrahydrofuran (150 mL) at 23 °C and stir for one hour. Pour the reaction mixture into 50% saturated aqueous sodium bicarbonate solution. Separate the organic layer and further extract the aqueous layer with ethyl ether (x 2) and ethyl acetate (x 2). Combine the organic extracts and wash with brine, then dry over anhydrous sodium sulfate, filter, and concentrate. Purify the resulting crude product by silica gel column chromatography (gradient elution: 20% ethyl acetate in hexanes to 100% ethyl acetate) to give the title compound (8.16 g, 77%). MS (m/z): 377 (M + 1).

Example la

Alternative synthesis of l-[2-(2,3-dihydro- lH-inden-2-ylamino)-7,8-dihydropyrido[4,3- d]pyrimidin-6(5H)-yl]-2-[2-(lH- l,2,3-triazol-4- l)ethoxy]ethanone.

Figure imgf000020_0001

Sparge a solution of 2-(but-3-yn- l-yloxy)-l-[2-(2,3-dihydro-lH-inden-2- ylamino)-7,8-dihydropyrido[4,3-d]pyrimidin-6(5H)-yl]ethanone (8.15 g, 21.7 mmol) and L-ascorbic acid sodium salt (8.58 g, 43.3 mmol) in dimethylformamide (60 mL) and water (60 mL) with nitrogen for ten minutes, then evacuate and backfill with nitrogen three times. Add copper (II) sulfate pentahydrate (1.08 g, 4.33 mmol) and heat to 90 °C, then add azidotrimethylsilane (23.1 mL, 20.0 g, 173 mmol) dropwise and stir for one hour. Cool reaction mixture to 23 °C and pour into water (50 mL). Extract this mixture with ethyl acetate (4 x 50 mL). Combine the organic extracts and wash with saturated aqueous sodium chloride, dry over anhydrous sodium sulfate, filter, and concentrate.

Purify the resulting crude product by silica gel column chromatography (gradient elution: 0 to 10% methanol in ethyl acetate) to give the title compound (3.60 g, 40%). MS (m/z): 420 (M + 1). Preparation 10

Synthesis of tert-butyl-2-(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidine-6-carboxylate.

Figure imgf000021_0001

Charge 450 rriL (2.58 mol) of N-ethyl-N-isopropylpropan-2-amine into a 15 °C solution of tert-butyl 2-chloro-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidine-6-carboxylate (220 g, 860.37 mmol) and 2,3-dihydro-lH-inden-2-amine (137.7 g, 1.03 mol) in 1- methylpyrrolidin-2-one (3.6 L). Heat the resulting mixture to 80 °C for 16 h, then cool to 30 °C and transfer the resulting mixture into 5 L of water at 25 °C. Filter the resulting solid and rinse the filter cake with water (2 x 300 rriL). Reslurry the solid in ethyl acetate (350 iriL) for 45 min at 15 °C. Filter the slurry, rinsing with 15 °C ethyl acetate ( 2 x 250 rriL), and dry to give the title compound (226 g, 75%) as an off-white solid. ‘H NMR (d6-DMSO) 1.45 (s, 9 H), 2.87 (dd, /= 7.2, 15.8 Hz, 2 H), 3.24 (dd, /= 7.2, 15.8 Hz, 2 H), 4.36 (d, 10.4 Hz, 2 H), 4.44 (d, /= 12.8 Hz, 2 H), 4.60 (m, 1 H), 7.14 (m, 2 H), 7.20 (m, 2 H), 7.55 (d, /= 6.8 Hz, 1 H), 8.27 (d, /= 7.2 Hz, 1 H).

Preparation 11

Synthesis of N-(2,3-dihydro-lH-inden-2-yl)-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-2- amine dihydrochloride hydrate.

Figure imgf000021_0002

Charge 670 rriL of 5 M hydrochloric acid (3.35 mol) to a solution of tert-butyl 2-

(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro-6H pyrrolo[3,4-d]pyrimidine-6- carboxylate (226 g, 641.25 mmol) in tetrahydrofuran (2.0 L) at 17 °C, maintaining the internal temperature below 26 °C during the addition. Heat the resulting solution to 50 °C for 16 h, cool to 25 °C and dilute with 500 rriL of water and 500 mL of tert- butylmethylether. Separate the resulting layers and extract with tert-butylmethylether (3 x 1 L). Concentrate the water phase down to a reaction volume of ca. 200 mL, and filter the resulting slurry. Rinse the cake with tert-butylmethylether (2 x 200 mL) and dry to give the title product (177 g, 80%) as a light brown solid. MS (m/z): 253.2 (M-2HC1- H20+1).

Preparation 12

Syntheis of tert-butyl 2-but-3-ynox acetate.

Figure imgf000022_0001

Stir a mixture of but-3-yn-l-ol (6.00 g; 85.60 mmol), tetrabutylammonium sulfate (2.07 g; 8.54 mmol) and sodium hydroxide (40% wt/wt; 150 mL) in dichloromethane (150 mL) at 0°C. Add tert-butyl bromoacetate (19.34 mL; 128.40 mmol) dropwise and stir the mixture for 2.5 hours at room temperature. Dilute the reaction mixture with dichloromethane (200 mL) and water (100 mL), separate the layers, and further extract the aqueous layer with dichloromethane (2 x 100 mL). Wash the combined organic layers with brine (100 mL), dry over anhydrous sodium sulfate, and concentrate to afford the crude title compound as a brown oil (11.93 g). Purify the oil by silica gel column chromatography, eluting with hexane: ethyl acetate (0% to 10% mixtures) to give the title compound (11.35 g; 72%) as a colorless oil. ]H NMR (CDCI3) δ 1.48 (s, 9H), 2.00 (m, 1H), 2.52 (m, 2H), 3.67 (m, 2H), 4.01 (bs, 2H).

Preparation 13

Synthesis of tert-butyl 2-[2-(lH-triazol-5- l)ethoxy]acetate.

Figure imgf000022_0002

Stir tert-Butyl 2-but-3-ynoxyacetate (11.34 g; 61.55 mmol) and copper(I)iodide (584 mg; 3.07 mmol) in a mixture of dimethylformamide (56.70 mL) and methanol (11.34 mL) at 0°C. Add azido(trimethyl)silane (12.33 mL; 86.47 mmol) dropwise and heat the mixture at 90°C for 18 hours.

In a second batch, stir tert-butyl 2-but-3-ynoxyacetate (4.38 g; 23.77 mmol) and copper(I)iodide (226 mg; 1.19 mmol) in a mixture of dimethylformamide (22 mL) and methanol (6 mL) at 0°C. Add azido(trimethyl)silane (4.8 mL; 33.66 mmol) dropwise and the mixture heated at 90°C for 18 hours.

Upon cooling to room temperature, combine the crude products from both batches and concentrate the mixture to afford a greenish residue. Purify the crude product by filtration through a plug of silica eluting with dichloromethane: ethyl acetate (75% to 100% mixtures) to afford the title compound (14.15 g, 73%) as a colorless oil. MS (m/z): 228.15 (M+l).

Preparation 14

Synthesis of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid 2,2,2-trifluoroacetic acid.

Figure imgf000023_0001

Stir a mixture of ieri-butyl 2-[2-(lH-triazol-5-yl)ethoxy]acetate (14.15 g; 62.26 mmol) and trifluoroacetic acid (70.75 mL, 935.69 mmol) in dichloromethane (70.75 mL) for 2 hours at room temperature. Concentrate the reaction mixture under reduced pressure to provide the title compound containing additional trifluoroacetic acid (20.22 g, >100%) as a brown solid. MS (m/z): 172.05 (M+l).

Example 2

Synthesis of l-[2-(2,3-dihydro- lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]-2-[2-(lH- l ,2,3-triazol-4-yl)ethoxy]ethanone.

Figure imgf000023_0002

Stir a mixture of 2-[2-(lH-triazol-5-yl)ethoxy]acetic acid 2,2,2-trifluoroacetic acid

(20.22 g; 70.90 mmol), N-(2,3-dihydro- lH-inden-2-yl)-6,7-dihydro-5H-pyrrolo[3,4- d]pyrimidin-2-amine dihydrochloride hydrate (27.99 g; 81.54 mmol) and triethylamine (98.83 mL; 709.03 mmol) in dimethylformamide (404.40 mL) at 0°C. Add a solution of 1-propanephosphonic acid cyclic anhydride (50% solution in DMF; 51.89 mL; 81.54 mmol) over 30 minutes, and stir the mixture at room temperature for 18 hours.

Concentrate the reaction mixture under reduced pressure to give a residue. Add water (200 mL) and extract the mixture with ethyl acetate (4 x 250 mL) and

dichloromethane (4 x 250 mL). Wash the combined organic layers with saturated aqueous sodium bicarbonate (2 x 100 mL) and brine (100 mL), then dry over anhydrous sodium sulfate. Filter the mixture and concentrate the solution under reduced pressure to give a red solid (25.70 g) that is slurried in ethyl acetate/methanol (9: 1 mixture; 200 mL) for 2 hours at room temperature. Filter the resulting solid and wash with cold ethyl acetate (50 mL) to give a solid (ca.18.2 g) that is re-slurried in ethyl acetate (200 mL) at reflux for 1 hour. On cooling to room temperature, stir the mixture for 1 hour and filter the resulting light pink solid.

Slurry the light pink solid in water/methanol (1 : 1 mixture; 200 mL) and heat the mixture at 50°C for 30 minutes. Add ammonium hydroxide solution (32% ; 50 mL) and continue to heat the mixture at 50°C for 30 minutes. Upon cooling to room temperature, add additional ammonium hydroxide solution (32% ; 50 mL) and continue stirring for 1 hour at room temperature. Filter the resulting light gray solid, dry and slurry again in ethyl acetate (200 mL) for 1 hour to afford a light gray solid that is filtered, washed with ethyl acetate (25 mL), and dried to give the title compound (12.42 g; 43%) as a gray solid. MS (m/z): 406 (M+l).

Preparation 15

Synthesis of 2-chloro- l-[2-(2,3-dihydro- lH-inden-2-ylamino)-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl]ethanone.

Figure imgf000024_0001

Stir a suspension of N-(2,3-dihydro-lH-inden-2-yl)-6,7-dihydro-5H-pyrrolo[3,4- d]pyrimidin-2-amine dihydrochloride hydrate (14.4 g, 41.9 mmol) and triethylamine (14.3 g, 19.7 mL, 141.4 mmol) in dichloromethane (200 mL) at 23 °C for 10 minutes, then cool to -30 °C. Add 2-chloroacetyl chloride (5.49 g, 3.86 mL, 48.6 mmol) over two minutes and warm to 23 °C over 10 minutes. Add methanol (5 mL) and remove the solvent in vacuo. Slurry the crude reaction mixture in methanol (30 mL), add 50 g silica gel and remove solvent in vacuo. Load the resulting residue onto a loading column and purify via silica gel column chromatography (gradient elution: 50% ethyl acetate in hexanes to ethyl acetate to 10% methanol in ethyl acetate) to give the title compound (11.5 g, 84%). MS (m/z): 329(M+1).

Preparation 16

Synthesis of 2-(but-3-yn-l-yloxy)-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro- 6H-pyrrolo[3,4-d]pyrimidin-6-yl]ethanone.

Figure imgf000025_0001

To sodium hydride (60 wt% in mineral oil, 2.06 g, 51.4 mmol) in tetrahydrofuran (86 mL) at 0 °C, add 3-butyn-l-ol (4.64 g, 5.03 mL, 64.3 mmol), then stir at 23 °C for 15 minutes. Add this solution to 2-chloro-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-5,7- dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]ethanone (8.45 g, 25.7 mmol) in

tetrahydrofuran (86 mL) at 0 °C and stir for five minutes. Pour reaction mixture into 50% saturated aqueous sodium bicarbonate solution. Separate the organic layer and further extract the aqueous layer with ethyl ether and ethyl acetate (2 x 50 mL each). Combine the organic extracts and wash with brine, then dry over anhydrous sodium sulfate, filter, and concentrate. Combine the crude product with the crude product from a second reaction (run reaction under identical conditions and stoichiometry employing 2-chloro- 1- [2-(indan-2-ylamino)-5,7-dihydropyrrolo[3,4-d]pyrimidin-6-yl]ethanone (3.0 g, 9.1 mmol)) and purify by silica gel column chromatography (gradient elution: 25% ethyl acetate in hexanes to 100% ethyl acetate) to give the title compound (2.90 g, 23%). MS

(m/z): 363(M+1). Example 2a

Alternative synthesis of l-[2-(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro- pyrrolo[3,4-d]pyrimidin-6-yl]-2-[2-(lH-l,2,3-triazol-4-yl)ethoxy]ethanone.

Figure imgf000026_0001

Add dimethylformamide (27 mL) and water (27 mL) to a flask containing 2-(but- 3-yn-l-yloxy)-l-[2-(2,3-dihydro-lH-inden-2-ylamino)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]ethanone (2.90 g, 8.00 mmol). Add copper (II) sulfate pentahydrate (400 mg, 1.60 mmol) and L-ascorbic acid sodium salt (3.17 g, 16.0 mmol). Evacuate flask and backfill with nitrogen (x 2), then add azidotrimethylsilane (7.37 g, 8.53 mL, 64.0 mmol) and heat the reaction to 90 °C for 70 minutes. Cool the reaction mixture to 23 °C and remove all solvent in vacuo. Suspend the residue in methanol/dichloromethane and then add silica gel and remove solvent in vacuo. Load this material onto a loading column and purify via silica gel column chromatography (gradient elution: 0-9% methanol in ethyl acetate) to give the title compound (980 mg, 30%). MS (m/z):

406(M+1).

/////////Autotaxin LPA osteoarthritis tool molecule, lily, Spencer Jones, PRECLINICAL

N1(Cc2cnc(nc2C1)NC3Cc4ccccc4C3)C(=O)COCCc5cnnn5


Filed under: Preclinical drugs Tagged: Autotaxin, lily, LPA, osteoarthritis, preclinical, Spencer Jones, tool molecule

BMS-852927

$
0
0

str1

str1

BMS-852927

CAS 256918-39-4

609.51 MW

C29 H28 Cl2 F2 N2 O4 S MF

2-(2-(2-(2,6-dichlorophenyl)propan-2-yl)-1-(3,3′-difluoro-4′-(hydroxymethyl)-5′-(methylsulfonyl)biphenyl-4-yl)-1H-imidazol-4-yl)propan-2-ol

1H-Imidazole-4-methanol, 2-[1-(2,6-dichlorophenyl)-1-methylethyl]-1-[3,3′-difluoro-4′-(hydroxymethyl)-5′-(methylsulfonyl)[1,1′-biphenyl]-4-yl]-α,α-dimethyl-

Treat metabolic syndrome

Brett Busch, Ph.D.

Brett Busch, Ph.D.

https://www.linkedin.com/in/brettbbusch

Exelixis

Brett B. Busch, William C. Stevens, Jr., Ellen K. Kick, Haiying Zhang, Venkataiah Bollu,Richard Martin, Raju Mohan
Applicant Exelixis, Inc.
Brett B. Busch, William C. Stevens, JR., Ellen K. Kick, Haiying Zhang, Venkataiah Bollu,Richard Martin, Raju Mohan
Bristol-Myers Squibb Company, Exelixis Patent Company Llc
  • Originator Exelixis
  • Developer Bristol-Myers Squibb
  • Class Antihyperlipidaemics; Small molecules
  • Mechanism of Action Liver X receptor modulators
  • Discontinued Atherosclerosis; Hypercholesterolaemia

Most Recent Events

  • 04 Jun 2014 BMS 852927 is still in phase I trials for atherosclerosis and in preclinical development for hypecholesterolaemia in USA
  • 02 Aug 2013 Bristol-Myers Squibb terminates the planned phase I trial for Hypercholesterolaemia in Germany, Canada and Switzerland (NCT01651273)
  • 06 Jul 2012 Bristol-Myers Squibb plans a phase I trial for Hypercholesterolaemia in Germany, Canada and Switzerland (NCT01651273)

1H-NMR (DMSO-d6, 400 MHz) δ 7.94 (m, 2H), 7.63 (dd, 1H, J = 11.29, 1.51 Hz), 7.34 (d, 1H, J = 9.54
Hz), 7.14 (m, 3H), 7.05 (m, 1H), 6.83 (s, 1H), 5.58 (t, 1H, J = 5.27 Hz), 4.96 (d, 2H, J = 4.27 Hz), 4.70
(s, 1H), 3.46 (s, 3H), 1.96 (s, 6H), 1.45 (s, 6H); MS m/e 609.16 (M+H+);

13CNMR (DMSO-d6, 400MHz) 161.42 (d, J=249.49 Hz), 156.85 (d, J=250.25 Hz), 153.18, 148.39, 141.69 (d, J=3.05 Hz), 139.45 (dd, J=9.16, 1.53 Hz), 139.32 (dd, J=8.39, 1.53 Hz), 138.58, 134.68, 131.39, 129.96, 128.40,
127.12 (d, J=17.55 Hz), 125.72 (d, J=12.97 Hz), 123.15 (d, J=2.29 Hz), 122.49 (d, J=3.05 Hz), 119.04
(d, J=25.18 Hz), 116.30, 114.52 (d, J=22.13 Hz), 68.11, 51.97 (d, J=5.34 Hz ), 45.53, 44.78, 44.29,
31.01, 30.53.

19F-NMR (JEOL 500 MHz, CDCl3) -113.55, -116.73.

HPLC (XBridge 5μ C18 4.6x50mm, 4 mL/min, Solvent A: 10 % MeOH/water with 0.2 % H3PO4, Solvent B: 90 % MeOH/water with0.2 % H3PO4, gradient with 0-100 % B over 4 minutes): 2.56 minutes, Purity, 99.7%.

HRMS (m/z,Obs.): 609.12065 [M+H]+; (Calc.): 609.11877. Formula: C29H29Cl2F2N2O4S. Anal. Calcd. for
C29H28N2O4SCl2F2•0.10 C2H6O•0.10 C4H5O2: C, 57.05; H, 4.75; Cl, 11.42; F, 6.10; N, 4.50; S, 5.15.
Found: C, 57.14; H, 4.54; Cl, 11.57; F, 5.94; N, 4.36; S, 5.07. The residual solvents, ethyl acetate (1.39
weight %), ethanol (0.74 weight %), dichloromethane (0.05 weight %), and heptane (< 0.05 weight %)
were identified in the sample by GC/MS and the retention times were matched with the reference standards.

Image result for BMS-852927

Image result for BMS-852927

Liver X receptors (LXRs) belong to a family of nuclear hormone receptors that are endogenously activated by cholesterol and its oxidized derivatives to mediate transcription of genes involved in maintaining glucose, cholesterol, and fatty acid metabolism. LXRa is found predominantly in the liver, with low levels found in kidney, intestine, spleen, and adrenal tissue. LXRp is ubiquitous in mammals and was found in nearly all tissues examined. Given the intricate link between lipid metabolism and cancer cell growth, the ubiquitous expression of LXRp in some types of cancer is unlikely to be coincidental, allowing cancer cells to synthesize lipids and lipoprotein particles to sustain their growth. At the same time, however, such stable basal expression levels make LXRp an ideal therapeutic target.

Figure

Examples of LXR agonists reported in the literature

PATENT

WO 2010138598

PATENT

WO 2012135082

PATENT

WO 2014028461

PATENT

WO 2016100619

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2016100619&recNum=1&maxRec=&office=&prevFilter=&sortOption=&queryString=&tab=PCTDescription

PATENT

https://www.google.com/patents/US8618154?cl=enIt

Figure US08618154-20131231-C00002

Example 9 2-(2-(2-(2,6-dichlorophenyl)propan-2-yl)-1-(3,3′-difluoro-4′-(hydroxymethyl)-5′-(methylsulfonyl)biphenyl-4-yl)-1H-imidazol-4-yl)propan-2-ol

Figure US08618154-20131231-C00021

Example 9a Preparation of 2-(2,6-dichlorophenyl)-2-methylpropanenitrile

Figure US08618154-20131231-C00022

To a 1 M solution of potassium tert-butoxide (403 mL, 403 mmol) at −66° C. (acetone/dry ice) was slowly added 2-(2,6-dichlorophenyl)acetonitrile (25.0 g, 134 mmol) in anhydrous THF (150 mL). The mixture was stirred at −66° C. for 20 minutes. Then, iodomethane (33.6 mL, 538 mmol) was added drop-wise over 25 minutes at −66° C. At this stage, it was exothermic and a large amount of light yellow precipitate was observed. The suspension was stirred at −60° C. for 30 minutes. The reaction mixture was quenched with 200 mL ice water, and extracted with ether (3×150 mL). The organics were combined, washed with 150 mL brine, dried over Na2SO4, and concentrated on a rotary evaporator. The crude product (30 g, yellow oil) was purified by column chromatography (ISCO, 330 g silica, 20% EtOAc in hexanes) to afford 2-(2,6-dichlorophenyl)-2-methylpropanenitrile (28.2 g, 132 mmol, 98% yield) as a light yellowish oil. 1H-NMR (CDCl3, 400 MHz) δ 7.35 (d, 2H, J=8.03 Hz), 7.16 (t, 1H, J=8.0 Hz), 2.09 (s, 6H); 13C-NMR (CDCl3, 126 MHz) δ134.6, 133.8, 131.4, 129.0, 124.1, 38.6, 29.2; MS m/e 214.10 (M+H+); HPLC (XBridge 5μ C18 4.6×50 mm, 4 mL/min, Solvent A: 10% MeOH/water with 0.2% H3PO4, Solvent B: 90% MeOH/water with 0.2% H3PO4, gradient with 0-100% B over 4 minutes): 3.16 minutes.

Example 9b Preparation of N-(4-bromo-2-fluorophenyl)-2-(2,6-dichlorophenyl)-2-methylpropanimidamide

Figure US08618154-20131231-C00023

2-(2,6-Dichlorophenyl)-2-methylpropanenitrile (20 g, 93 mmol) and 4-bromo-2-fluoroaniline (28.4 g, 149 mmol) were dissolved in anhydrous o-xylene (200 mL) and heated to 100° C. under N2. Trimethylaluminum (2 M) in toluene (140 mL, 280 mmol) was added drop-wise (˜0.9 mL per minute) over 2.5 hours while the reaction mixture was stirred at 100° C. After addition, the reaction mixture was stirred at 100° C. for 30 minutes, and then cooled to −5° C. The reaction mixture was very carefully quenched with potassium sodium tartrate (20 g in 100 mL water) (Caution: gas and heat formation). The reaction mixture was filtered through Celite 545. The filtrate was washed with 1N HCl (4×70 mL). The aqueous was neutralized with 2N NaOH and extracted with EtOAc (4×100 mL). The organics were combined, washed with brine, dried with Na2SO4, and concentrated on a rotary evaporator to afford 24 g of crude product. The crude product was recrystallized with 72 mL of MTBE and 240 mL of hexane to give N-(4-bromo-2-fluorophenyl)-2-(2,6-dichlorophenyl)-2-methylpropanimidamide (17.5 g, 43.3 mmol, 46.4% yield) as a white solid (purity: 99%). 1H-NMR (MeOD, 400 MHz) δ 7.42 (d, 2H, J=8.0 Hz), 7.30 (m, 2H), 7.16 (t, 1H, J=8.0 Hz), 6.93 (t, 1H, J=8.0 Hz), 2.11 (s, 6H); 13C-NMR (DMSO-d6, 100 MHz) δ 166.5, 156.1, 153.7, 140.6, 138.5, 135.9, 131.4, 128.6, 128.0, 125.7, 119.5, 112.9, 50.0, 29.2; MS m/e 403.09 (M+H+); HPLC (XBridge 5μ C18 4.6×50 mm, 4 mL/min, Solvent A: 10% MeOH/water with 0.2% H3PO4, Solvent B: 90% MeOH/water with 0.2% H3PO4, gradient with 0-100% B over 4 minutes): 2.32 minutes.

Example 9c Preparation of ethyl 1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophenyl)propan-2-yl)-4-hydroxy-4,5-dihydro-1H-imidazole-4-carboxylate

Figure US08618154-20131231-C00024

To a mixture of N-(4-bromo-2-fluorophenyl)-2-(2,6-dichlorophenyl)-2-methylpropanimidamide (48.0 g, 119 mmol), K2CO3(41.0 g, 297 mmol) in toluene (180 mL) and THF (180 mL) at 55° C. was added slowly a solution of ethyl 3-bromo-2-oxopropanoate (23.3 mL, 166 mmol) in 24 mL of THF over 50 minutes. The reaction mixture was kept at 55° C. for 1.5 hours. A white slurry was observed. The reaction mixture was cooled to 5° C. HCl (0.5N, 450 mL) was added drop-wise (end point pH=9˜10). After addition, the suspension was cooled to 0° C. The solid was collected by filtration, washed with water (2×50 mL), and then dried in a vacuum oven at 60° C. overnight. Ethyl 1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophenyl)propan-2-yl)-4-hydroxy-4,5-dihydro-1H-imidazole-4-carboxylate (59 g, 114 mmol, 96% yield) was obtained as a white solid. 1H-NMR (CDCl3, 400 MHz) δ 7.11 (m, 3H), 6.96 (m, 2H), 6.72 (t, 1H, J=8.28 Hz), 4.35 (m, 2H), 4.25 (d, 1H, J=10.5 Hz), 3.80 (d, 1H, J=10.8 Hz), 1.98 (s, 3H), 1.93 (s, 3H), 1.38 (t, 3H, J=7.03 Hz); 13C-NMR (CDCl3, 126 MHz) δ 173.0, 171.5, 159.8, 157.8, 137.3, 135.7, 132.1, 131.1, 128.1, 127.4, 125.6, 122.2, 120.1, 93.5, 62.5, 45.5, 30.2, 14.0; MS m/e 517.05 (M+H+); HPLC (XBridge 5μ C18 4.6×50 mm, 4 mL/min, Solvent A: 10% MeOH/water with 0.2% H3PO4, Solvent B: 90% MeOH/water with 0.2% H3PO4, gradient with 0-100% B over 4 minutes): 2.74 minutes.

Example 9d Preparation of ethyl 1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophenyl)propan-2-yl)-1H-imidazole,4-carboxylate

Figure US08618154-20131231-C00025

To a mixture of ethyl 1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophenyl)propan-2-yl)-4-hydroxy-4,5-dihydro-1H-imidazole-4-carboxylate (38 g, 73 mmol) in EdOH (200 mL) was added TFA (25.0 g, 220 mmol). The mixture was stbsequently heated tn 95° C. HPLC analysis after 2.5 hours showed <1% of alcohol intermediate remaining The mixture was diluted with 300 mL of CH2Cl2 and cooled to approximately 5° C. with an ice bath. The mixture was neutralized with 1N NaOH (120 mL) and the organic layer was separated. The aqueous layer was dxtracted with CH2Cl2 (2×100 mL). The combined organic layers were concentrated on a rotary evaporator to give crude material. Recrystallization in EtOH (5 mL/1 g) provided 32 g of ethyl 1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophdnyl)propan-2-yl)-1H-imidazole-4-carboxylate as `n off-white solhd (86% yield). 1H-NMR (DMSO-d6, 400 MHz) δ 7.92 (s, 1H), 7.16 (d, 1H, J=8.0 Hz), 7.22 (m, 3H), 7.11 (m, 1H), 7.04 (t, 1H, J=12.0 Hz), 4.25 (q, 2H, J=8.0 Hz), 1.94 (s, 6H(, 1.27 (t, 3H, J=8.0 Hz); MS m/e 502.68 (M+H+); HPLC (XBridge 5μ C18 4.6×50 mm, 4 mL/min, Solvent A: 10% MeOH/water with 0.2% H3PO4, Solvent B: 90% MeOH/water with 0.2% H3PO4, gradient with 0-100% B over 4 minutes): 3.87 minutes.

Example 9e Prepar`tion of 2-(1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophenyl)propan-2-yl)-1H-imidazol-4-yl)propan-2-ol

Figure US08618154-20131231-C00026

To a mixture of methylmagnesium bromide (60.0 mL, 180 mmol, 3M in ether) in 120 ml, of THF cooled with an ice/salt bath (−15 to −17° C.) was added slowly a solution of ethyl 1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophenyl)propan-2-yl)-1H-imidazole-4-carboxylate (30 g, 60 mmol) in 65 mL of CH2Cl2 and 87 mL of THF over 45 minutes. The internal temperature was carefully kept below 0° C. A further 2×20 mL of CH2Cl2 was used to wash forward the residual material. The reaction mixture temperature was maintained below 0° C. for 1 hour with stirring. Then the reaction mixture was diluted with 100 mL of CH2Cl2, and saturated NH4Cl was added slowly. The resulting mixture was extracted with CH2Cl2 (2×80 mL). Organics were combined, washed with brine, dried with Na2SO4, and concentrated on a rotary evaporator to afford 2-(1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophenyl)propan-2-yl)-1H-imidazol-4-yl)propan-2-ol (28.5 g, 58.6 mmol, 98% yield) as a white solid. 1H-NMR (CDCl3, 400 MHz) δ 7.13 (dd, 1H, J=9.03, 2.01 Hz), 7.09 (s, 1H), 7.07 (s, 1H), 6.93 (m, 2H), 6.75 (t, 1H, J=8.16 Hz), 6.55 (s, 1H), 3.18 (s, 1H), 2.00 (s, 6H), 1.58 (s, 6H); 13C-NMR (CDCl3, 126 MHz) δ 158.1, 156.1, 154.5, 147.8, 139.3, 135.7, 131.3, 130.3, 127.8, 126.9, 122.7, 119.8, 115.1, 68.7, 44.8, 31.1, 29.9; MS m/e 485.05 (M+H+); HPLC (XBridge 5μ C18 4.6×50 mm, 4 mL/min, Solvent A: 10% MeOH/water with 0.2% H3PO4, Solvent B: 90% MeOH/water with 0.2% H3PO4, gradient with 0-100% B over 4 minutes): 2.78 minutes.

Example 9 Preparation of 2-(2-(2-(2,6-dichlorophenyl)propan-2-yl)-1-(3,3′-difluoro-4′-(hydroxymethyl)-5′-(methylsulfonyl)biphenyl-4-yl)-1H-imidazol-4-yl)propan-2-ol

Figure US08618154-20131231-C00027

To a 1 L 3-necked round bottom flask under nitrogen was added 2-(1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophenyl)propan-2-yl)-1H-imidazol-4-yl)propan-2-ol (12.0 g, 24.7 mmol), [2-fluoro-6-methanesulfonyl-4-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-phenyl]-methanol (9.78 g, 29.6 mmol), K2CO3 (10.2 g, 74 mmol), DME (120 mL) and water (12 mL). The mixture was heated to 60° C., and then 1,1′-bis(diphenylphosphino)ferrocene palladium (II) chloride complex (4.06 g, 4.94 mmol) was added under nitrogen. The reaction mixture was heated to 80° C. for 30 minutes. The resulting darkly colored mixture was cooled with an ice bath, and partitioned in 200 mL of CH2Cl2 and 200 mL of water. The organic layers were combined and dried with Na2SO4. After concentration, the crude product was purified by flash chromatography (ISCO, 330 g silica, 0% to 100% EtOAc in hexanes) to afford 12.79 g of crude product (85% yield) as a light yellow solid.

Recrystallization was carried out by dissolving 9.5 g of crude product in acetone (80 mL) at 65° C. The resulting solution was cooled slowly to 25° C. over 5 hours, and then cooled to 0° C. for an additional 30 minutes. Crystals began to form at 45° C. The solid was collected by filtration and rinsed with cold acetone. After drying in an oven at 45° C. under vacuum for 14 hours, 4.9 g of pure product was obtained. To recover additional crystalline product, the mother liquid was concentrated to approximately 10 mL and passed through a silica pad. EtOAc (100 mL) was used to elute the compound. The filtrate was concentrated under vacuum to give a crude solid. The crude solid was recrystallized in acetone following the procedure above to afford an additional 2.5 g of product. The combined recovery for the two crops after recrystallization was a 78% yield. 1H-NMR (DMSO-d6, 400 MHz) δ 7.94 (m, 2H), 7.63 (dd, 1H, J=11.29, 1.51 Hz), 7.34 (d, 1H, J=9.54 Hz), 7.14 (m, 3H), 7.05 (m, 1H), 6.83 (s, 1H), 5.58 (t, 2H, J=5.27 Hz), 4.96 (d, 2H, J=4.27 Hz), 4.70 (s, 1H), 3.46 (s, 3H), 1.96 (s, 6H), 1.45 (s, 6H); MS m/e 609.16 (M+H+); HPLC (XBridge 5μ C18 4.6×50 mm, 4 mL/min, Solvent A: 10% MeOH/water with 0.2% H3PO4, Solvent B: 90% MeOH/water with 0.2% H3PO4, gradient with 0-100% B over 4 minutes): 2.56 minutes.

Alternatively, Example 9 was prepared as follows:

To a 1 L 3-necked round bottom flask under nitrogen was added methyltetrahydrofuran (“MeTHF”, 6.9 kg), 2-(1-(4-bromo-2-fluorophenyl)-2-(2-(2,6-dichlorophenyl)propan-2-yl)-1H-imidazol-4-yl)propan-2-ol (1.994 kg, 4.1 moles) and (2-fluoro-6-(methylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)methanol (1.38 kg, 4.19 moles). The mixture was agitated at 23° C. for 15 min until all the solids dissolved. At the conclusion of this period, (oxydi-2,1-phenylene)bis(diphenylphosphine) (0.022 kg, 0.041 moles) and Pd(OAc)2 (0.01 kg, 0.045 moles) were added as a slurry via a subsurface line. Upon completion of addition, the mixture was rinsed with additional MeTHF (1.65 kg). The resulting mixture was evacuated to less than 80 Torr and backfilled with nitrogen. This process was repeated two more times. After completion of the degassing sequence, the reaction mixture was agitated for at least 15 min and a clear, golden color was observed. In a separate reaction vessel, a solution of potassium hydroxide (0.352 kg) in water (10.00 kg) was prepared and degassed by sparging the solution with nitrogen gas for at least 15 min prior to use. The KOH solution (10.35 kg) was transferred into the reactor by vacuum. The reaction temperature exhibited a known exotherm from 20° C. to 29° C. Upon completion of addition, the resulting biphasic mixture was degassed by a series of pressure swings. The mixture was warmed to between 45-50° C. where it was stirred for at least 2 h. After this time, the reaction mixture was analyzed by HPLC, which indicated the reaction was complete. The reaction mixture was cooled to 23° C. and the stirring was stopped. The mixture was allowed to separate for 30 min and the lower spent KOH stream was removed. The product rich organic was passed through a column of thiourea functionalized silica gel (0.782 kg) (Silicycle) at ˜0.1 kg per min to remove the palladium. The product rich organic phase was washed with a 5% NaHCO3 solution (5 vol) and the phases separated. The organic phase was washed with water (5 vol) and the organic and aqueous phases separated.

The product rich organic phase was polish filtered into a clean reaction vessel and then concentrated to ˜8 volumes (˜16 L) under vacuum (80 Torr, Tjacket=60° C.). Once at the prescribed volume, the reaction mixture was allowed to cool to 25° C. Once at the prescribed temperature the reaction mixture was seeded with 2-(2-(2-(2,6-dichlorophenyl)propan-2-yl)-1-(3,3′-difluoro-4′-(hydroxymethyl)-5′-(methylsulfonyl)biphenyl-4-yl)-1H-imidazol-4-yl)propan-2-ol (0.5%, 0.008 kg). The resulting slurry was stirred at 25° C. for about 18 h. At the conclusion of this period, the reaction mixture was concentrated to ˜8 L under vacuum (150 Torr, Tjacket=60° C.). Once at the prescribed volume, the reaction mixture was heated to 50° C. and isopropyl acetate (IPAc, 13.90 kg) was added to the reactor during a 90 min period. Upon completion of addition, the reaction mixture was cooled to 25° C. during a 3 h period. Once at the prescribed temperature the reaction mixture was stirred at room temperature for about 16 h. At the conclusion of this period, the reaction mixture was filtered, deliquored, and washed with additional IPAc (10.4 kg). The filter cake was dried via suction on the filter under a stream of dry nitrogen to yield a white solid. The white solid was transferred to a dryer and dried at 50° C. under full vacuum to afford 2.03 kg of product (81% yield, 99.40 AP, 98 wt %).

PAPER

Abstract Image

Introducing a uniquely substituted phenyl sulfone into a series of biphenyl imidazole liver X receptor (LXR) agonists afforded a dramatic potency improvement for induction of ATP binding cassette transporters, ABCA1 and ABCG1, in human whole blood. The agonist series demonstrated robust LXRβ activity (>70%) with low partial LXRα agonist activity (<25%) in cell assays, providing a window between desired blood cell ABCG1 gene induction in cynomolgus monkeys and modest elevation of plasma triglycerides for agonist 15. The addition of polarity to the phenyl sulfone also reduced binding to the plasma protein, human α-1-acid glycoprotein. Agonist 15 was selected for clinical development based on the favorable combination of in vitroproperties, excellent pharmacokinetic parameters, and a favorable lipid profile.

Discovery of Highly Potent Liver X Receptor β Agonists

Department of Discovery Chemistry, Department of Cardiovascular Biology, #Pharmaceutical Candidate Optimization, Research & Development, Bristol-Myers Squibb, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
Exelixis Inc., 210 East Grand Avenue, South San Francisco, California 94080, United States
ACS Med. Chem. Lett., Article ASAP
DOI: 10.1021/acsmedchemlett.6b00234
Publication Date (Web): October 23, 2016
Copyright © 2016 American Chemical Society
*Tel: 609 466-5053. E-mail: ellen.kick@bms.com.

http://pubs.acs.org/doi/full/10.1021/acsmedchemlett.6b00234

WO2007002563A1 Jun 26, 2006 Jan 4, 2007 Exelixis, Inc. Imidazole based lxr modulators
WO2008073825A1 Dec 7, 2007 Jun 19, 2008 Exelixis, Inc. Lxr and fxr modulators
Citing Patent Filing date Publication date Applicant Title
US8901106 Mar 26, 2012 Dec 2, 2014 Bristol-Myers Squibb Company Imidazole prodrug LXR modulators
US20140163081 * Nov 21, 2013 Jun 12, 2014 Exelixis Patent Company Llc Lxr modulators
US20150299136 * May 4, 2015 Oct 22, 2015 Bristol-Myers Squibb Company Lxr modulators

///////////Discovery, Highly Potent,  Liver X Receptor β Agonists, ABCA1 ABCG1 Liver X receptor LXRα LXRβ α-1-acid glycoproteinBMS-852927, BMS 852927

CS(=O)(=O)c1cc(cc(F)c1CO)c2cc(F)c(cc2)n3cc(nc3C(C)(C)c4c(Cl)cccc4Cl)C(C)(C)O


Filed under: Preclinical drugs Tagged: ABCA1, ABCG1, BMS-852927, discovery, Highly Potent, α-1-acid glycoprotein, Liver X receptor, Liver X Receptor β Agonists, LXRα, LXRβ

Design and synthesis of indoline thiohydantoin derivatives based on enzalutamide as antiproliferative agents against prostate cancer

$
0
0

str0

str0

4-(6-chloro-1-oxo-3-thioxo-9,9a-dihydro-1H-imidazo[1,5-a]indol-2(3H)-yl)-2-(trifluoromethyl)-benzonitrile

WILL BE UPDATED………

Prostate cancer, one of the most malignant tumors worldwide, is the second leading cause of cancer deaths among men in America . Although androgen deprivation therapy (ADT) has been proved to be effective initially, the tumor will eventually progress and develop into the lethal castration resistant prostate cancer (CRPC) . The androgen receptor (AR) is a ligand-dependent transcription factor belonging to the nuclear receptor superfamily and plays a critical role in the progression of normal prostate cells. However, overexpression of AR was found in most CRPC, which is essential for CRPC to adapt to the low levels of androgens. As AR contributes significantly to the resistance to castration, it has been recognized as an attractive target for the treatment of CRPC

Reagents and conditions: (i) HNO3 , H2SO4 , -5 oC, 3 h; (ii) SOCl2 , MeOH, reflux, 12 h; (iii) H2 , Pd/C, MeOH, rt, 12 h; (iv) (CH3CO)2O, TEA, 50 oC, 6 h; (v) H2 , Pd/C, MeOH, rt, 12 h; (vi) acetone, HCl (6 mol/L), -10 oC, 0.5 h, NaNO2 , H2O, -10 oC, 1 h, CuCl/CuBr/KI, 0 oC, 3 h; (vii) HCl, 50 oC, 3 h; (viii) SOCl2 , MeOH, reflux, 12 h; (ix) 2, DMF, TEA, 60 oC, 1 h.

4-(6-chloro-1-oxo-3-thioxo-9,9a-dihydro-1H-imidazo[1,5-a]indol-2(3H)-yl)-2-(trifluoromethyl)-benzonitrile (48c). It was obtained as a yellow solid

m.p. 220-222 oC;

1H-NMR (300 MHz,DMSO-d6): δ 8.40 (d, J = 8.1 Hz, 1H, Ar-H), 8.19 (s, 1H, Ar-H), 8.02-7.92 (m, 2H, Ar-H), 7.49-7.46 (m, 1H, Ar-H), 7.34-7.32 (m, 1H, Ar-H), 5.56 (t, J = 9.6 Hz, 1H, -CH-), 3.58 (d, J = 9.6 Hz, 2H, -CH2-) ppm;

13C-NMR (75 MHz, DMSO-d6): δ 184.1, 172.1, 142.3, 138.5, 136.8, 134.7, 131.9, 131.5, 128.0, 126.1 (q, J = 267.9 Hz, CF3), 117.2, 115.4, 66.9, 39.9 ppm;

IR (KBr): 3094, 2232, 1763, 1607, 1499, 1270, 1136, 1052, 998, 786 cm-1;

HRMS (ESI): m/z, calculated for C18H9ClF3N3OS 408.0180 (M + H)+ , found 408.0173.

Paper

A series of indoline thiohydantoin derivatives were synthesized and evaluated in vitro.The most potent compound 48c shows comparable ability with enzalutamide in proliferation inhibition of LNCaP cells.Compound 48c has less cytotoxic to AR-negative cells compared with Enzalutamide.

The bicalutamide-resistant mechanism was clarified and overcome by compound 48c.


Abstract

A novel scaffold of indoline thiohydantoin was discovered as potent androgen receptor (AR) antagonist through rational drug designation. Several compounds showed good biological profiles in AR binding and higher selective toxicity than enzalutamide toward LNCaP cells (AR-rich) versus DU145 cells (AR-deficient). In addition, the docking studies supported the rationalization of the biological evaluation. Among these compounds, the representative compound 48c exhibited the strongest inhibitory effect on LNCaP growth and also acted as a competitive AR antagonist. Further preliminary mechanism study confirmed that 48c exerted its AR antagonistic activity through impairing AR nuclear translocation. All these results indicated that the novel scaffold compounds demonstrated AR antagonistic behaviour and promising candidates for future development were identified.


Graphical abstract

Image 1
Research paper

Design and synthesis of indoline thiohydantoin derivatives based on enzalutamide as antiproliferative agents against prostate cancer

  • Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
  • zhiyuli@cpu.edu.cn

http://www.sciencedirect.com/science/article/pii/S0223523416309114

http://dx.doi.org/10.1016/j.ejmech.2016.10.049

////////Prostate cancer, Androgen receptor, Antagonist, Indoline thiohydantoin derivatives, indoline thiohydantoin derivatives, enzalutamide, antiproliferative agents, prostate cancer

c1c(cc(c(c1)C#N)C(F)(F)F)N2C(C3Cc4ccc(cc4N3C2=S)Cl)=O


Filed under: Preclinical drugs Tagged: androgen receptor, Antagonist, antiproliferative agents, enzalutamide, Indoline thiohydantoin derivatives, Prostate cancer

CEP 33779

$
0
0

img

CEP-33779, CEP33779
CAS 1257704-57-6
Chemical Formula: C24H26N6O2S
Molecular Weight: 462.57
Elemental Analysis: C, 62.32; H, 5.67; N, 18.17; O, 6.92; S, 6.93

N-(3-(4-methylpiperazin-1-yl)phenyl)-8-(4-(methylsulfonyl)phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-amine

PRECLINICAL Treatment of Rheumatoid Arthritis, Agents for Colorectal Cancer Therapy Systemic Lupus Erythematosus,

Jak2 Inhibitors

Image result for teva logo

Matthew A. Curry, Bruce D. Dorsey, Benjamin J. Dugan, Diane E. Gingrich, Eugen F. Mesaros, Karen L. Milkiewicz,
Applicant Cephalon, Inc.

Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380, United States

Image result for Cephalon, Inc.

Matt Curry

 Matthew A. Curry

Bruce Dorsey

Bruce Dorsey

Image result for Cephalon, Inc. Benjamin J. Dugan

Benjamin Dugan

Benjamin J. Dugan received a B.S. degree in Chemistry from the University of Delaware in 1993 under the tutelage of the late Dr. Cynthia McClure. He began his career at FMC Corporation in the agricultural products division. In 2006, he moved to Cephalon, Inc., acquired by Teva Pharmaceutical Industries Ltd. in 2011, and engaged in oncology research focused on small molecule, ATP competitive, kinase inhibitors culminating with the discovery of CEP-33779. He is currently a Research Scientist focused on the development of novel, bioactive small molecules for treatment of central nervous system disorders.

Cephalon Inc.
Malvern, United States

Image result for Cephalon, Inc. Diane E. Gingrich

Members of the Cephalon research team that discovered CEP-5214 and CEP-7055 include (from left) Hudkins, Thelma S. Angeles, Bruce A. Ruggeri, and Diane E. Gingrich. CEPHALON PHOTO

Eugen F. Mesaros

Cephalon Inc.
Malvern, United States
Image result for cephalon Karen L. Milkiewicz

Lupus (systemic lupus erythematosus, SLE) is a chronic autoimmune disease characterized by the presence of activated T and B cells, autoantibodies and chronic inflammation that attacks various parts of the body including the joints, skin, kidneys, CNS, cardiac tissue and blood vessels. In severe cases, antibodies are deposited in the cells (glomeruli) of the kidneys, leading to inflammation and possibly kidney failure, a condition known as lupus nephritis.

Although the cause of lupus remains unknown, manifestations of the disease have been linked to genetic polymorphisms, environmental toxins and pathogens (Morel;

Fairhurst, Wandstrat et al. 2006). In addition, gender, hormonal influences and cytokine dysregulation have been tightly linked to the development of lupus (Aringer and Smolen 2004; Smith-Bouvier, Divekar et al. 2008). Lupus affects nine times as many women as men. It may occur at any age, but appears most often in people between the ages of 10 and 50 years. African Americans and Asians are affected more often than people from other races.

There is no cure for lupus. Current treatments for lupus are aimed at controlling symptoms and are limited to toxic and immunosuppressive agents with severe side-effects such as high dose glucocorticoids and/or hydroxchloroquine. Severe disease (e.g., patients that have signs of renal involvement) require more aggressive drugs including

mycophenolate mofetil (MMF), azathioprine (AZA) and/or cyclophosphamide (CTX) (Bertsias and Boumpas 2008). CTX, AZA and MMF are very toxic and

immunosuppressive, and only 50% of treated patients enter complete remission, with relapse rates up to 30% over a 2-year period.

Memory B cells, and more important, long-lived plasma cells (LL-PCs) which differentiate from memory B cells, are key cell types involved in lupus (Neubert, Meister et al. 2008; Sanz and Lee 2010). Long-lived plasma cells synthesize and secrete large quantities of high-affinity isotype switched antibodies (Meister, Schubert et al. 2007;

Muller, Dieker et al. 2008). Circulating antinuclear antibodies (ANAs) increase the chances of antibody depositing onto self tissues, forming immune-complexes and eventually leading to tissue destruction, epitope spreading and involvement of other organ systems. LL-PCs are commonly found to be chemo- and radio-resistant, over expressing various heat shock proteins and drug pumps (Obeng, Carlson et al. 2006; Neubert, Meister et al. 2008). In addition, LL-PCs primarily reside in the bone marrow where they are protected from current lupus therapies such as cyclophosphamide and glucocorticoids.

A need exists for new treatments for lupus, including lupus nephritis. A need particularly exists for lupus treatments that can target and reduce LL-PCs.

str0

CEP-33779 is a highly selective, orally active, small-molecule inhibitor of JAK2. CEP-33779 induced regression of established colorectal tumors, reduced angiogenesis, and reduced proliferation of tumor cells. Tumor regression correlated with inhibition of STAT3 and NF-κB (RelA/p65) activation in a CEP-33779 dose-dependent manner. The ability of CEP-33779 to suppress growth of colorectal tumors by inhibiting the IL-6/JAK2/STAT3 signaling suggests a potential therapeutic utility of JAK2 inhibitors in multiple tumors types, particularly those with a strong inflammatory component.

str0

{[8-(4-Methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-1-yl)-phenyl]-amine} (1)

LC/MS: (M+H+)+ = 463.2;
1H NMR (DMSO, 400 MHz) δ 9.61 (s, 1H), 8.85 (d, J = 6.8 Hz, 1H), 8.43 (d, J = 6.8 Hz, 2H), 8.06 (d, J = 6.8 Hz, 2H), 7.96 (d, J = 7.5 Hz, 1H), 7.59 (s, 1H), 7.17 (t, J = 6.8 Hz, 1H), 7.11 (t, J = 8.0 Hz, 1H), 7.05 (d, J = 8.6 Hz 1H), 6.49 (d, J = 8.0 Hz, 1H), 3.30 (s, 3H), 3.13 (m, 4H), 2.48 (m, 4H), 2.24 (s, 3H).
CEP-33779 Diglycolate Salt
1H NMR (DMSO, 400 MHz) δ 9.61 (s, 1H), 8.85 (d, J = 6.7 Hz, 1H), 8.43 (d, J = 6.7 Hz, 2H), 8.06 (d, J = 6.7 Hz, 2H), 7.97 (d, J = 7.5 Hz, 1H), 7.59 (s, 1H), 7.18 (d, J = 6.7 Hz, 1H), 7.11 (m, 1H), 7.05 (d, J = 8.6 Hz, 1H), 6.50 (d, J = 8.0 Hz, 1H), 3.89 (s, 4H), 3.30 (s, 3H), 3.13 (m, 4H), 2.48 (m, 4H), 2.24 (s, 3H).
DSC: Endotherm onset at 153.0 °C; Peak at 155.8 °C.

PATENT

WO 2010141796

https://www.google.com/patents/WO2010141796A3?cl=en

Example 35 [8-(4-Methanesulfonyl-phenyl)-[ 1 ,2,4]triazolo[ 1 ,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-

1 -yl)-phenyl]-amine

Figure imgf000156_0001

35 a) l-(3-Bromo-phenyl)-4-methyl-piperazine was prepared from l-(3-bromo-phenyl)- piperazine (1.33 g, 5.52 mmol) in a manner analogous to Step 32a. The reaction product was isolated as a pale yellow oil (1.4 g, 100%). 1H NMR (400 MHz, CDCl3, δ, ppm): 7.10 (dd, J=8.2, 8.2 Hz, IH), 7.04 (dd, J=2.1, 2.1 Hz, IH), 6.95 (ddd, J=I. S, 1.7, 0.7 Hz, IH), 6.83 (ddd, J=8.3, 2.4, 0.6 Hz, IH), 3.23-3.18 (m, 4H), 2.58-2.54 (m, 4H), 2.35 (s, 3H). MS = 255, 257 (MH)+. 35b) [8-(4-Methanesulfonyl-phenyl)-[ 1 ,2,4]triazolo[ 1 ,5-a]pyridin-2-yl]-[3-(4-methyl- piperazin-l-yl)-phenyl]-amine was prepared from 8-(4-methanesulfonyl-phenyl)- [l,2,4]triazolo[l,5-a]pyridin-2-ylamine (75.0 mg, 0.260 mmol) and l-(3-bromo-phenyl)-4- methyl-piperazine (80.0 mg, 0.314 mmol) with 2,2′-bis-dicyclohexylphosphanyl-biphenyl (30.0 mg, 0.0549 mmol) as the ligand in a manner analogous to Step 2d and was isolated as a yellow solid (0.072 g, 60%).

MP = 232-234 0C.

1H NMR (400 MHz, CDCl3, δ, ppm): 8.49 (d, J=I 2 Hz, IH), 8.25 (d, J=I .5 Hz, 2H), 8.08 (d, J=I .9 Hz, 2H), 7.65 (d, J=I .1 Hz, IH), 7.38 (s, IH), 7.27-7.20 (m, IH), 7.04-6.95 (m, 2H), 6.84 (s, IH), 6.60 (d, J=8.0 Hz, IH), 3.30-3.25 (m, 4H), 3.10 (s, 3H), 2.63-2.58 (m, 4H), 2.38 (s, 3H).

MS = 463 (MH)+.

PATENT

WO 2012078504

PATENT

WO 2012078574

https://google.com/patents/WO2012078574A2?cl=da

COMPOUND A is a JAK2 inhibitor with the chemical name [8-(4-methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-1-yl)-phenyl]-amine. COMPOUND A has the following structure:

COMPOUND A

COMPOUND A was prepared in a manner analogous to the five-step method described below (see Example 35 of International Application No. PCT/US10/37363):

Step 1 : To a solution of 1-(3-bromo-phenyl)-piperazine (about 1 g) and acetic acid (about 0.4 mL) in methanol (about 25 mL) is added 37% formaldehyde in water/methanol (about 56.7:37:6.3, water:formaldehyde:methanol; about 5 mL). The mixture is stirred at room temperature for about 18 hours. The suspension is cooled to about 5°C in an ice/water bath and sodium cyanoborohydride (about 5 g) is added in small portions. The mixture is stirred and warmed to room temperature for about 18 hours. The mixture is slowly poured into saturated aqueous ammonium chloride (about 200 mL) and stirred for about 1 hour. The mixture is extracted with dichloromethane (3 x about 75 mL). The combined organic layers are dried over magnesium sulfate, filtered and evaporated. The material is placed under high vacuum for about 18 hours to yield 1-(3-bromo-phenyl)-4-methyl-piperazine as a pale yellow oil (about 1 g). 1H NMR (400 MHz, CDCl3, δ, ppm): 7.10 (dd, J=8.2, 8.2 Hz, 1H), 7.04 (dd, J=2.1, 2.1 Hz, 1H), 6.95 (ddd, J=7.8, 1.7, 0.7 Hz, 1H), 6.83 (ddd, J=8.3, 2.4, 0.6 Hz, 1H), 3.23-3.18 (m, 4H), 2.58-2.54 (m, 4H), 2.35 (s, 3H). MS = 255, 257 (MH)+.

Step 2: To a solution of 3-bromo-pyridin-2-ylamine (about 10 g) in 1,4-dioxane (about 100 mL) is added dropwise ethoxycarbonyl isothiocyanate (about 7 mL). The mixture is stirred under an atmosphere of nitrogen for about 18 hours. The volatiles are evaporated to yield a waxy solid. The recovered material is triturated with hexane (about 250 mL). N-(3-bromo-2-pyridinyl)-N’-carboethoxy-thiourea is isolated and used without further purification. 1H NMR (400 MHz, (D3C)2SO, δ, ppm): 11.46 (s, 1H), 11.43 (s, 1H), 8.49 (dd, J=4.6, 1.5 Hz, 1H), 8.18 (dd, J=8.0, 1.5 Hz, 1H), 7.33 (dd, J=8.0, 4.7 Hz, 1H), 4.23 (q, J=7.1 Hz, 2H), 1.27 (t, J=7.2 Hz, 3H). MS = 215 (MH)+.

Step 3: To a stirred suspension of hydroxylamine hydrochloride (about 17 g) and Ν,Ν-diisopropylethylamine (about 26 mL) in a mixture of methanol (about 70 mL) and

ethanol (about 70 mL) is added N-(3-bromo-2-pyridinyl)-N’-carboethoxy-thiourea. The mixture is stirred for about 2 hours at room temperature then heated to about 60°C for about 18 hours. The suspension is cooled to room temperature, filtered and rinsed with methanol, water then methanol. 8-Bromo-[1,2,4]triazolo[1,5-a]pyridin-2-ylamine is isolated as an off-white solid (about 8 g). 1H NMR (400 MHz, (D3C)2SO, δ, ppm): 8.58 (d, J=6.4 Hz, 1H), 7.73 (d, J=7.6 Hz, 1H), 6.80 (t, J=7.0 Hz, 1H), 6.25 (s, 2H). MS = 213, 215 (MH)+.

Step 4: An oven dried tube is charged with palladium acetate (about 0.2 g) and triphenylphosphine (about 0.6 g). The tube is evacuated under high vacuum and backflushed under a stream of nitrogen for about 5 minutes. A suitable solvent such as

1,4-dioxane (about 10 mL) is added and the mixture is stirred under nitrogen for a suitable time (e.g., for about 10 minutes). 8-Bromo-[1,2,4]triazolo[1,5-a]pyridin-2-ylamine (about 0.75 g), (4-methylsulfonylphenyl)boronic acid (about 1 g), a suitable solvent, such as N,N-dimethylformamide (about 10 mL) and a suitable base, such as about 1.5 M of sodium carbonate in water (about 10 mL) are added. The mixture is stirred for about 2 minutes at room temperature under nitrogen then the tube is sealed and heated at about 80°C for about 18 hours. The mixture is transferred to a round bottom flask and the volatiles are evaporated under reduced pressure. The product is isolated in a suitable manner. For example, water (about 100 mL) may be added and the mixture stirred. The solid may then be collected by filtration, and optionally rinsed with water, air dried, triturated with ether/dichloromethane (about 4: 1; about 10 mL), filtered and rinsed with ether. 8-(4-methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-ylamine is isolated as a tan solid (about 0.6 g). MP = 236-239 °C. 1H NMR (400 MHz, (D3C)2SO, δ, ppm): 8.63 (d, J=6.3 Hz, 1H), 8.38 (d, J=7.9 Hz, 2H), 8.03 (d, J=7.9 Hz, 2H), 7.84 (d, J= 7.3 Hz, 1H), 7.03 (t, J=7.0 Hz, 1H), 6.21 (br s, 2H), 3.28 (s, 3H). MS = 289 (MH)+.

Step 5: To an oven dried tube is added palladium acetate (about 10 mg) and 2,2′-bis-dicyclohexylphosphanyl-biphenyl (about 30 mg), 8-(4-methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-ylamine (about 75 mg), 1-(3-bromo-phenyl)-4-methyl-piperazine (about 80 mg), a suitable base, such as cesium carbonate (about 270 mg) and a suitable solvent, such as 1,4-dioxane (about 5 mL). The tube is evacuated and backflushed with nitrogen three times. The tube is sealed and heated at about 80°C for about 72 hours. The mixture is cooled to room temperature and the product isolated in a suitable manner.

For example, the cooled mixture may be diluted with dichloromethane (about 10 mL), filtered through a plug of diatomaceous earth, rinsed with dichloromethane and evaporated. The material may then be purified, e.g., via chromatography, e.g., utilizing an ISCO automated purification apparatus (e.g., amine modified silica gel column 5%→100% ethyl acetate in hexanes). [8-(4-Methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-1-yl)-phenyl]-amine (i.e., COMPOUND A) is isolated as a yellow solid (about 0.07 g). MP = 232-234 °C. 1H NMR (400 MHz, CDCl3, δ, ppm): 8.49 (d, J=7.2 Hz, 1H), 8.25 (d, J=7.5 Hz, 2H), 8.08 (d, J=7.9 Hz, 2H), 7.65 (d, J=7.7 Hz, 1H), 7.38 (s, 1H), 7.27-7.20 (m, 1H), 7.04-6.95 (m, 2H), 6.84 (s, 1H), 6.60 (d, J=8.0 Hz, 1H), 3.30-3.25 (m, 4H), 3.10 (s, 3H), 2.63-2.58 (m, 4H), 2.38 (s, 3H). MS = 463 (MH)+.

PATENT

WO 2015089153

https://www.google.com/patents/WO2015089153A1?cl=un

This disclosure relates to a l,2,4 riazolo[l,5a]pyridine derivative, [8-(4 methanesulfonyl-phenyl)-[ 1 ,2,4]triazoio[1 ,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin- 1 -yl phenyl] -amine, re g structure:

or a pharmaceutical salt thereof, and its use in the treatment of multiple sclerosis.

Compound A is a potent, orally active, small molecule inhibitor of JA 2. See, e.g..International Application No. PCT/USlO/37363, U.S. Patent Nos. 8,501,936 and ,633,173, and U.S. Published Patent Application Nos. 2013/0267535 and 2014/0024655, each of which is incorporated by reference herein. Compound A can be prepared, for example, using methods analogous to Example 35 of International Application No.PCT/US 10/37363.

PAPER

A Selective, Orally Bioavailable 1,2,4-Triazolo[1,5-a]pyridine-Based Inhibitor of Janus Kinase 2 for Use in Anticancer Therapy: Discovery of CEP-33779

Worldwide Discovery Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, Pennsylvania 19380, United States
J. Med. Chem., 2012, 55 (11), pp 5243–5254
DOI: 10.1021/jm300248q
Publication Date (Web): May 10, 2012
Copyright © 2012 American Chemical Society
*Phone: 610-738-6733. Fax: 610-738-6643. E-Mail: bdugan@cephalon.com.

Abstract

Abstract Image

Members of the JAK family of nonreceptor tyrosine kinases play a critical role in the growth and progression of many cancers and in inflammatory diseases. JAK2 has emerged as a leading therapeutic target for oncology, providing a rationale for the development of a selective JAK2 inhibitor. A program to optimize selective JAK2 inhibitors to combat cancer while reducing the risk of immune suppression associated with JAK3 inhibition was undertaken. The structure–activity relationships and biological evaluation of a novel series of compounds based on a 1,2,4-triazolo[1,5-a]pyridine scaffold are reported. Para substitution on the aryl at the C8 position of the core was optimum for JAK2 potency (17). Substitution at the C2 nitrogen position was required for cell potency (21). Interestingly, meta substitution of C2-NH-aryl moiety provided exceptional selectivity for JAK2 over JAK3 (23). These efforts led to the discovery of CEP-33779 (29), a novel, selective, and orally bioavailable inhibitor of JAK2.

[8-(4-Methanesulfonyl-phenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-yl]-[3-(4-methyl-piperazin-1-yl)-phenyl]-amine (29)

 1H NMR (CDCl3) δ 8.49 (dd, J = 6.6, 1.0 Hz, 1H), 8.25 (d, J = 8.4 Hz, 2H), 8.08 (d, J = 8.4 Hz, 2H), 7.66 (dd, J = 7.5, 0.9 Hz, 1H), 7.39–7.36 (m, 1H), 7.23 (t, J = 8.2 Hz, 1H), 7.02 (t, J = 7.1 Hz, 1H), 6.97 (dd, J = 7.8, 1.4 Hz, 1H), 6.88 (s, 1H), 6.60 (dd, J = 8.3, 1.8 Hz, 1H), 3.30–3.25 (m, 4H), 3.10 (s, 3H), 2.63–2.58 (m, 4H), 2.38 (s, 3H).
13C NMR (CDCl3) δ 162.65, 152.28, 148.87, 141.00, 140.91, 140.05, 129.64, 129.29, 128.18, 127.85, 127.76, 124.77, 112.03, 109.40, 108.59, 104.80, 55.19, 49.02, 46.19, 44.59;
mp 208–211 °C.
High resolution mass spectrum (ESI+) m/z 463.1925 [(M + H)+calcd for C24H26N6O2S: 463.1916]. HPLC: 95 A%.

PAPER

An Improved Synthesis of the Free Base and Diglycolate Salt of CEP-33779; A Janus Kinase 2 Inhibitor

Chemical Process Research and Development, Teva Branded Pharmaceutical Products R&D Inc., 383 Phoenixville Pike, Malvern, Pennsylvania 19355, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00311
Publication Date (Web): November 30, 2016
Copyright © 2016 American Chemical Society

Abstract

Abstract Image

CEP-33779 is a triazole that has been reported to show highly selective inhibition of Janus kinase 2 (JAK2). An efficient process to form CEP-33779 will be presented that uses multiple palladium couplings to provide the drug substance in a convergent manner. The existing medicinal chemistry route was modified to avoid chromatographic purification, improve safety, and utilize palladium ligands which are available in quantities amenable to scale-up. Challenges faced during the development of the new process included optimization of conditions for Buchwald–Hartwig and Suzuki couplings, control of homocoupled impurities and removal of residual palladium. In addition, a screen of conditions to form a diglycolate salt of the parent compound are also presented.

REFERENCES

1: Dugan BJ, Gingrich DE, Mesaros EF, Milkiewicz KL, Curry MA, Zulli AL, Dobrzanski P, Serdikoff C, Jan M, Angeles TS, Albom MS, Mason JL, Aimone LD, Meyer SL, Huang Z, Wells-Knecht KJ, Ator MA, Ruggeri BA, Dorsey BD. A selective, orally bioavailable 1,2,4-triazolo[1,5-a]pyridine-based inhibitor of Janus kinase 2 for use in anticancer therapy: discovery of CEP-33779. J Med Chem. 2012 Jun 14;55(11):5243-54. doi: 10.1021/jm300248q. Epub 2012 May 18. PubMed PMID: 22594690.

2: Tagoe C, Putterman C. JAK2 inhibition in murine systemic lupus erythematosus. Immunotherapy. 2012 Apr;4(4):369-72. doi: 10.2217/imt.12.20. PubMed PMID: 22512630.

3: Seavey MM, Lu LD, Stump KL, Wallace NH, Hockeimer W, O’Kane TM, Ruggeri BA, Dobrzanski P. Therapeutic efficacy of CEP-33779, a novel selective JAK2 inhibitor, in a mouse model of colitis-induced colorectal cancer. Mol Cancer Ther. 2012 Apr;11(4):984-93. doi: 10.1158/1535-7163.MCT-11-0951. Epub 2012 Feb 14. PubMed PMID: 22334590.

4: Lu LD, Stump KL, Wallace NH, Dobrzanski P, Serdikoff C, Gingrich DE, Dugan BJ, Angeles TS, Albom MS, Mason JL, Ator MA, Dorsey BD, Ruggeri BA, Seavey MM. Depletion of autoreactive plasma cells and treatment of lupus nephritis in mice using CEP-33779, a novel, orally active, selective inhibitor of JAK2. J Immunol. 2011 Oct 1;187(7):3840-53. doi: 10.4049/jimmunol.1101228. Epub 2011 Aug 31. PubMed PMID: 21880982.

5: Stump KL, Lu LD, Dobrzanski P, Serdikoff C, Gingrich DE, Dugan BJ, Angeles TS, Albom MS, Ator MA, Dorsey BD, Ruggeri BA, Seavey MM. A highly selective, orally active inhibitor of Janus kinase 2, CEP-33779, ablates disease in two mouse models of rheumatoid arthritis. Arthritis Res Ther. 2011 Apr 21;13(2):R68. doi: 10.1186/ar3329. PubMed PMID: 21510883; PubMed Central PMCID: PMC3132063.

/////////////CEP-33779, CEP33779, CEP 33779, 1257704-57-6, PRECLINICAL, TEVA,  Rheumatoid Arthritis, Colorectal Cancer Therapy, Systemic Lupus Erythematosus,

Jak2 Inhibitors

O=S(C1=CC=C(C2=CC=CN3C2=NC(NC4=CC=CC(N5CCN(C)CC5)=C4)=N3)C=C1)(C)=O

str1 str2

str0


Filed under: Preclinical drugs Tagged: 1257704-57-6, CEP-33779, CEP33779, Colorectal Cancer Therapy, Jak2 Inhibitors, preclinical, rheumatoid arthritis, Systemic lupus erythematosus, teva

DNDI-VL-2098

$
0
0

str0

DNDI-VL-2098

CAS 681492-17-1

(R)-2-Methyl-6-nitro-2-(4-trifluoromethoxyphenoxymethyl)-2,3-dihydroimidazo[2,1-b]oxazole

Watch this post, will be updated………..

MF C14 H12 F3 N3 O5,
MW 359.26
Imidazo[2,1-b]oxazole, 2,3-dihydro-2-methyl-6-nitro-2-[[4-(trifluoromethoxy)phenoxy]methyl]-, (2R)-
Image result for OTSUKA
Medicinal Chemistry Research Institute, Otsuka Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan, and Microbiological Research Institute, Otsuka Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
Image result for OTSUKA Hidetsugu Tsubouchi
(left to right) Hidetsugu Tsubouchi, Ph.D., Compliance & Ethics Department, manager; Hirofumi Sasaki, Medicinal Chemistry Research Laboratories, associate head and project OPC; Makoto Matsumoto, Ph.D, Pharmaceutical Business Division, senior director; Hiroyuki Hashizume, Pharmaceutical Marketing Headquarters, Product Planning and Management Group, product management manager; Masanori Kawasaki, TB Projects, associate director
Melting Point: 176-178 °C , Condition: Solvent ethyl acetate; isopropanol

(2R)-2-Methyl-6-nitro-2-(4-trifluoromethoxyphenoxymethyl)-2,3-dihydroimidazo[2,1-b]oxazole

Mp: 169–171 °C; Org. Process Res. Dev., Article ASAP, DOI: 10.1021/acs.oprd.6b00331

HPLC (area %): 99.52%; HPLC (chiral): 99.8% (a/a);

1H NMR (400 MHz, CDCl3): δ 7.57 (s, 1H), 7.14–7.16 (d, 2H, J = 10.0 Hz), 6.83–6.86 (d, 2H, J = 7.2 Hz), 4.48–4.50 (d, 1H, J = 10.0 Hz), 4.22–4.24 (d, 1H, J = 10.0 Hz), 4.05–4.10 (t, 2H, J = 9.6 and 10.4 Hz), 1.79 (s, 3H);

13C NMR (100 MHz, CDCl3): δ 156.0, 155.8, 147.1, 143.5, 122.6, 115.5, 112.6, 122.6, 121.7, and 119.1 (JC–F = 255.1 Hz), 116.6, 92.9, 71.8, 51.3, 23.0;

19F NMR (CDCl3, 376 MHz): δ −58.4;

IR (KBr, cm–1): 3155, 2996, 1607, 1456, 1281, 1106, 978, 921, 834,783, 708;

mass (m/z): 360.3 (M + 1)+;

[α]25589 = (+)8.445 (c 1.00 g/100 mL, CHCl3).

Visceral leishmaniasis (VL), infamously known as kala-azar (black fever) in the Indian subcontinent, is the most lethal form of leishmaniasis and is caused by protozoan parasites. This deadly disease is the second largest parasitic killer in the world, surpassed only by malaria, with a worldwide distribution in Asia, East Africa, South America, and the Mediterranean region. In the search for effective treatments for visceral leishmaniasis, the Drugs for Neglected Diseases initiative (DNDi) recently evaluated fexinidazole a nitroimidazole being developed as a treatment for Human African Trypanosomiasis. Fexinidazole  showed potential as a safe and effective oral drug for the treatment of visceral leishmaniasis and is now in clinical trials.

Figure

fexinidazole (1) and DNDI-VL-2098 (2).

Earlier, through an agreement with TB Alliance and in association with the ACSRC at the University of Auckland (NZ), DNDi screened about 70 other nitroimidazole analogues belonging to four chemical subclasses and investigated them for antileishmanial activity

Image result for DNDI-VL-2098

Image result for DNDI-VL-2098

Paper

http://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.5b01699

Repositioning Antitubercular 6-Nitro-2,3-dihydroimidazo[2,1-b][1,3]oxazoles for Neglected Tropical Diseases: Structure–Activity Studies on a Preclinical Candidate for Visceral Leishmaniasis

Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
§ Laboratory for Microbiology, Parasitology and Hygiene, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow 226031, India
Drugs for Neglected Diseases Initiative, 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
# Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
Global Alliance for TB Drug Development, 40 Wall Street, New York 10005, United States
J. Med. Chem., 2016, 59 (6), pp 2530–2550
DOI: 10.1021/acs.jmedchem.5b01699
*Phone: (+649) 923-6145. Fax: (+649) 373-7502. E-mail: am.thompson@auckland.ac.nz.

Abstract

Abstract Image

6-Nitro-2,3-dihydroimidazo[2,1-b][1,3]oxazole derivatives were initially studied for tuberculosis within a backup program for the clinical trial agent pretomanid (PA-824). Phenotypic screening of representative examples against kinetoplastid diseases unexpectedly led to the identification of DNDI-VL-2098 as a potential first-in-class drug candidate for visceral leishmaniasis (VL). Additional work was then conducted to delineate its essential structural features, aiming to improve solubility and safety without compromising activity against VL. While the 4-nitroimidazole portion was specifically required, several modifications to the aryloxy side chain were well-tolerated e.g., exchange of the linking oxygen for nitrogen (or piperazine), biaryl extension, and replacement of phenyl rings by pyridine. Several less lipophilic analogues displayed improved aqueous solubility, particularly at low pH, although stability toward liver microsomes was highly variable. Upon evaluation in a mouse model of acute Leishmania donovani infection, one phenylpyridine derivative (37) stood out, providing efficacy surpassing that of the original preclinical lead.

Figure

Structures of various antileishmanial or antitubercular agents.

 str1 str0

CLICK ON IMAGE

2-Methyl-6-nitro-2-{[4-(trifluoromethoxy)phenoxy]methyl}-2,3-dihydroimidazo[2,1- b][1,3]oxazole (7).

Method A (Scheme 1B): Reaction of alcohol 88 with NaH, using procedure C, followed by chromatography of the product on silica gel, eluting with CH2Cl2, gave 71 (87%) as a pale yellow solid: mp (CH2Cl2/hexane) 122-124 C (lit.1 mp 126.8-127.9 C); 1 H NMR (CDCl3)  7.56 (s, 1 H), 7.16 (br d, J = 9.1 Hz, 2 H), 6.85 (br d, J = 9.2 Hz, 2 H), 4.48 (d, J = 10.2 Hz, 1 H), 4.23 (d, J = 10.1 Hz, 1 H), 4.09 (d, J = 10.1 Hz, 1 H), 4.05 (d, J = 10.2 Hz, 1 H), 1.79 (s, 3 H); 13C NMR (CDCl3)  156.3 (C-1’), 156.1 (C-7a), 147.4 (C- 6), 143.9 (q, JC-F = 2.1 Hz, C-4’), 122.8 (2 C, C-3’,5’), 120.7 (q, JC-F = 256.5 Hz, 4’-OCF3), 115.8 (2 C, C-2’,6’), 112.8 (C-5), 93.1 (C-2), 72.2 (2-CH2O), 51.6 (C-3), 23.3 (2-CH3). Anal. (C14H12F3N3O5) C, H, N.

Method B (Scheme 2B): Reaction of 2-bromo-1-[(2-methyloxiran-2-yl)methyl]-4-nitro-1Himidazole2 (98) with 4-(trifluoromethoxy)phenol (0.95 equiv) and NaH (1.2 equiv), using procedure I, followed by chromatography of the product on silica gel, eluting with 2:1 and 3:1 CH2Cl2/petroleum ether (foreruns) and then with 3:1 CH2Cl2/petroleum ether and CH2Cl2, S8 gave a crude product, which was crystallized from CH2Cl2/hexane (and the mother liquors further purified by chromatography on silica gel, eluting as before), to give 71 (55%) as a pale yellow solid (see data above). Method C (Scheme 2D): Reaction of 2-chloro-1-[(2-methyloxiran-2-yl)methyl]-4-nitro-1Himidazole1 (109) with 4-(trifluoromethoxy)phenol (1.0 equiv) and NaH, using procedure I, followed by chromatography of the product on silica gel, eluting with 1:1 and 3:2 CH2Cl2/petroleum ether (foreruns) and then with 3:1 CH2Cl2/petroleum ether and CH2Cl2, gave a crude product, which was crystallized from CH2Cl2/hexane (and the mother liquors further purified by chromatography on silica gel, eluting with 1:1 and 3:1 Et2O/petroleum ether and then with Et2O and CH2Cl2), to give 71 (51%) as a pale yellow solid (see data above).

Synthesis of 9 (Scheme 2A): (2R)-2-Methyl-6-nitro-2-{[4-(trifluoromethoxy)phenoxy]methyl}-2,3-dihydroimidazo- [2,1-b][1,3]oxazole (9). Reaction of 2-chloro-1-{[(2R)-2-methyloxiran-2-yl]methyl}-4-nitro- 1H-imidazole3 (96) with 4-(trifluoromethoxy)phenol and NaH, using procedure H, gave 91,3 (36%) as a pale brown solid: mp 170-171 C (lit.1 mp 176.5-178 C); 1 H NMR (CDCl3)  7.56 (s, 1 H), 7.16 (br d, J = 8.8 Hz, 2 H), 6.85 (br d, J = 9.0 Hz, 2 H), 4.48 (d, J = 10.2 Hz, 1 H), 4.23 (d, J = 10.0 Hz, 1 H), 4.09 (d, J = 10.2 Hz, 1 H), 4.05 (d, J = 10.3 Hz, 1 H), 1.79 (s, 3 H); [α] 25 D 9.0 (c 1.002, CHCl3) [lit.1 [α] 28 D 7.67 (c 1.030, CHCl3)]. Anal. (C14H12F3N3O5) C, H, N. HPLC purity: 100%. Chiral HPLC (using a CHIRALPAK AD-H analytical column and eluting with 15% EtOH/hexane at 1 mL/min) determined that the ee of 9 was 98.7%.

Paper

Sasaki, Hirofumi; Journal of Medicinal Chemistry 2006, VOL 49(26), Pg 7854-7860

Synthesis and Antituberculosis Activity of a Novel Series of Optically Active 6-Nitro-2,3-dihydroimidazo[2,1-b]oxazoles

Medicinal Chemistry Research Institute, Otsuka Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan, and Microbiological Research Institute, Otsuka Pharmaceutical Co., Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
J. Med. Chem., 2006, 49 (26), pp 7854–7860
DOI: 10.1021/jm060957y

Abstract

Abstract Image

In an effort to develop potent new antituberculosis agents that would be effective against both drug-susceptible and drug-resistant strains of Mycobacterium tuberculosis, we prepared a novel series of optically active 6-nitro-2,3-dihydroimidazo[2,1-b]oxazoles substituted at the 2-position with various phenoxymethyl groups and a methyl group and investigated the in vitro and in vivo activity of these compounds. Several of these derivatives showed potent in vitro and in vivo activity, and compound 19 (OPC-67683) in particular displayed excellent in vitro activity against both drug-susceptible and drug-resistant strains of M. tuberculosis H37Rv (MIC = 0.006 μg/mL) and dose-dependent and significant in vivo efficacy at lower oral doses than rifampicin in mouse models infected with M. tuberculosis Kurono. The synthesis and structure−activity relationships of these new compounds are presented.

(R)-2-Methyl-6-nitro-2-(4-trifluoromethoxyphenoxymethyl)-2,3-dihydroimidazo[2,1-b]oxazole (8). Mp 176−178 °C.

1H NMR (CDCl3) δ 1.79 (3H, s), 4.06 (1H, d, J = 6.8 Hz), 4.10 (1H, d, J = 6.8 Hz), 4.23 (1H, d, J = 10.1 Hz), 4.49 (1H, d, J = 10.1 Hz), 6.84 (2H, d, J = 9.0 Hz), 7.13 (2H, d, J = 9.0 Hz), 7.56 (1H, s).

MS (DI) m/z 359 (M+). Anal. (C14H12F3N3O5) C, H, N.

PAPER

Abstract Image

A process suitable for kilogram-scale synthesis of (2R)-2-methyl-6-nitro-2-{[4-(trifluoromethoxy)phenoxy]methyl}-2,3-dihydroimidazo[2,1-b][1,3]oxazole (DNDI-VL-2098, 2), a preclinical drug candidate for the treatment of visceral leishmaniasis, is described. The four-step synthesis of the target compound involves the Sharpless asymmetric epoxidation of 2-methyl-2-propen-1-ol, 8. Identification of a suitable synthetic route using retrosynthetic analysis and development of a scalable process to access several kilograms of 2 are illustrated. The process was simplified by employing in situ synthesis of some intermediates, reducing safety hazards, and eliminating the need for column chromatography. The improved reactions were carried out on the kilogram scale to produce 2 in good yield, high optical purity, and high quality.

http://pubs.acs.org/doi/abs/10.1021/acs.oprd.6b00331

Development of a Scalable Process for the Synthesis of DNDI-VL-2098: A Potential Preclinical Drug Candidate for the Treatment of Visceral Leishmaniasis

Process Chemistry Division, Advinus Therapeutics Ltd., 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, Karnataka, India
Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
Drugs for Neglected Diseases initiative (DNDi), 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00331
*Process Chemistry Division, Advinus Therapeutics Ltd., 21 & 22, Phase II, Peenya Industrial Area, Bangalore -560058, Karnataka, India. E-mail: hari.pati@advinus.com. Tel. No.: (+91)9900212096.
 
Hiroyuki Fujiki, Ph.D, New Drug Research Division, Biology and Translational Research Unit, senior research scientist; Yoshitaka Yamamura, Pharmaceutical Business Division, senior director; Youichi Yabuuchi, Ph.D, Otsuka Pharmaceutical Factory, Inc., corporate adviser; Hidenori Ogawa, Ph.D, Medicinal Chemistry Research Laboratories
/////////////preclinical, DNDI-VL-2098, 681492-17-1, Visceral Leishmaniasis

Filed under: Preclinical drugs Tagged: 681492-17-1, DNDI-VL-2098, preclinical, Visceral Leishmaniasis

BMS-960

$
0
0

Figure imgf000099_0001

str1

BMS-960

PRECLINICAL

(S)-1-((S)-2-Hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylic Acid

3-Piperidinecarboxylic acid, 1-[(2S)-2-hydroxy-2-[4-[5-[3-phenyl-4-(trifluoromethyl)-5-isoxazolyl]-1,2,4-oxadiazol-3-yl]phenyl]ethyl]-, (3S)-

(S)-1-((S)-2-Hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylic Acid

CAS 1265321-86-5 FREE FORM

FREE FORM 528.48, C26 H23 F3 N4 O5

CAS 1265323-40-7 HCL SALT

BASIC PATENT WO201117578, 2011, (US Patent 8399451)

Inventors John L. Gilmore, James E. Sheppeck
Applicant Bristol-Myers Squibb Company

Image result for Bristol-Myers Squibb Company

Sphingosine-1-phosphate (S1P) is the endogenous ligand for the sphingosine-1-phophate receptors (S1P1–5) and triggers a number of cellular responses through their stimulation. S1P and its interaction with the S1P receptors play a significant role in a variety of biological processes including vascular stabilization, heart development, lymphocyte homing, and cancer angiogenesis. Agonism of S1P1, especially, has been shown to play an important role in lymphocyte trafficking from the thymus and secondary lymphoid organs, inducing immunosuppression, which has been established as a novel mechanism of treatment for immune diseases and vascular diseases

Sphingosine-1 -phosphate (SlP) has been demonstrated to induce many cellular effects, including those that result in platelet aggregation, cell proliferation, cell morphology, tumor cell invasion, endothelial cell and leukocyte chemotaxis, endothelial cell in vitro angiogenesis, and lymphocyte trafficking. SlP receptors are therefore good targets for a wide variety of therapeutic applications such as tumor growth inhibition, vascular disease, and autoimmune diseases. SlP signals cells in part via a set of G protein-coupled receptors named SlPi or SlPl, SlP2 or S1P2, SlP3 or S1P3, SlP4 Or S1P4, and SlP5 or S1P5 (formerly called EDG-I, EDG-5, EDG-3, EDG-6, and EDG-8, respectively).

SlP is important in the entire human body as it is also a major regulator of the vascular and immune systems. In the vascular system, SlP regulates angiogenesis, vascular stability, and permeability. In the immune system, SlP is recognized as a major regulator of trafficking of T- and B-cells. SlP interaction with its receptor SlPi is needed for the egress of immune cells from the lymphoid organs (such as thymus and lymph nodes) into the lymphatic vessels. Therefore, modulation of SlP receptors was shown to be critical for immunomodulation, and SlP receptor modulators are novel immunosuppressive agents.

The SlPi receptor is expressed in a number of tissues. It is the predominant family member expressed on lymphocytes and plays an important role in lymphocyte trafficking. Downregulation of the SlPi receptor disrupts lymphocyte migration and homing to various tissues. This results in sequestration of the lymphocytes in lymph organs thereby decreasing the number of circulating lymphocytes that are capable of migration to the affected tissues. Thus, development of an SlPi receptor agent that suppresses lymphocyte migration to the target sites associated with autoimmune and aberrant inflammatory processes could be efficacious in a number of autoimmune

Among the five SlP receptors, SlPi has a widespread distribution and is highly abundant on endothelial cells where it works in concert with SIP3 to regulate cell migration, differentiation, and barrier function. Inhibition of lymphocyte recirculation by non-selective SlP receptor modulation produces clinical immunosuppression preventing transplant rejection, but such modulation also results in transient bradycardia. Studies have shown that SlPi activity is significantly correlated with depletion of circulating lymphocytes. In contrast, Sl P3 receptor agonism is not required for efficacy. Instead, SIP3 activity plays a significant role in the observed acute toxicity of nonselective SlP receptor agonists, resulting in the undesirable cardiovascular effects, such as bradycardia and hypertension. (See, e.g., Hale et al, Bioorg. Med. Chem. Lett., 14:3501 (2004); Sanna et al., J. Biol. Chem., 279: 13839 (2004); Anliker et al., J. Biol. Chem., 279:20555 (2004); Mandala et al., J. Pharmacol. Exp. Ther., 309:758 (2004).)

An example of an SlPi agonist is FTY720. This immunosuppressive compound FTY720 (JPI 1080026-A) has been shown to reduce circulating lymphocytes in animals and humans, and to have disease modulating activity in animal models of organ rejection and immune disorders. The use of FTY720 in humans has been effective in reducing the rate of organ rejection in human renal transplantation and increasing the remission rates in relapsing remitting multiple sclerosis (see Brinkman et al., J. Biol. Chem., 277:21453 (2002); Mandala et al., Science, 296:346 (2002); Fujino et al., J.

Pharmacol. Exp. Ther., 305:45658 (2003); Brinkman et al, Am. J. Transplant., 4: 1019 (2004); Webb et al., J. Neuroimmunol, 153: 108 (2004); Morris et al., Eur. J. Immunol, 35:3570 (2005); Chiba, Pharmacology & Therapeutics, 108:308 (2005); Kahan et al., Transplantation, 76: 1079 (2003); and Kappos et al., N. Engl. J. Med., 335: 1124 (2006)). Subsequent to its discovery, it has been established that FTY720 is a prodrug, which is phosphorylated in vivo by sphingosine kinases to a more biologically active agent that has agonist activity at the SlPi, SIP3, SlP4, and SIP5 receptors. It is this activity on the SlP family of receptors that is largely responsible for the pharmacological effects of FTY720 in animals and humans. [0007] Clinical studies have demonstrated that treatment with FTY720 results in bradycardia in the first 24 hours of treatment (Kappos et al, N. Engl. J. Med., 335: 1124 (2006)). The observed bradycardia is commonly thought to be due to agonism at the SIP3 receptor. This conclusion is based on a number of cell based and animal experiments. These include the use of SIP3 knockout animals which, unlike wild type mice, do not demonstrate bradycardia following FTY720 administration and the use of SlPi selective compounds. (Hale et al., Bioorg. Med. Chem. Lett., 14:3501 (2004); Sanna et al., J. Biol. Chem., 279: 13839 (2004); and Koyrakh et al., Am. J. Transplant, 5:529 (2005)).

The following applications have described compounds as SlPi agonists: WO 03/061567 (U.S. Patent Publication No. 2005/0070506), WO 03/062248 (U.S. Patent No. 7,351,725), WO 03/062252 (U.S. Patent No. 7,479,504), WO 03/073986 (U.S. Patent No. 7,309,721), WO 03/105771, WO 05/058848, WO 05/000833, WO 05/082089 (U.S. Patent Publication No. 2007/0203100), WO 06/047195, WO 06/100633, WO 06/115188, WO 06/131336, WO 2007/024922, WO 07/109330, WO 07/116866, WO 08/023783 (U.S. Patent Publication No. 2008/0200535), WO 08/029370, WO 08/114157, WO 08/074820, WO 09/043889, WO 09/057079, and U.S. Patent No. 6,069,143. Also see Hale et al., J. Med. Chem., 47:6662 (2004).

There still remains a need for compounds useful as SlPi agonists and yet having selectivity over Sl P3.

Applicants have found potent compounds that have activity as SlPi agonists. Further, applicants have found compounds that have activity as SlPi agonists and are selective over SIP3. These compounds are provided to be useful as pharmaceuticals with desirable stability, bioavailability, therapeutic index, and toxicity values that are important to their drugability.

SYNTHESIS

Figure

(S)-1-((S)-2-Hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylic acid, HCl (BMS-960). CAS 1265323-40-7

(S)-1-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylic acid, HCl (BMS-960)

1H NMR (400 MHz, DMSO-d6) δ 12.88 (br. s, 1H), 10.5 (br. s, 1H), 8.14 (d, J = 8.6 Hz, 2H), 7.72 (d, J = 8.4 Hz, 2H), 7.69–7.57 (m, 5H), 6.43 (br. s., 1H), 5.37 (d, J = 10.8 Hz, 1H), 3.89–3.60 (m, 2H), 3.50–2.82 (m, 6H), 2.14–1.99 (m, 1H), 1.97–1.75 (m, 1H), 1.63–1.35 (m, 1H);

13C NMR (101 MHz, CDCl3) δ 172.8, 168.5, 164.0, 161.6, 155.4, 156.2, 131.2, 129.0, 128.9, 127.4, 127.2, 125.5, 124.3, 122.2, 111.6, 66.6. 63.0, 52.9, 52.2, 38.8, 25.0, 21.7;

19F NMR (376 MHz, DMSO-d6) δ −54.16;

Anal. calcd for C26H23F3N4O5·HCl: C, 54.71; H, 4.36; N, 9.80. Found: C, 54.76; H, 3.94; N, 9.76;

HRMS (ESI) m/e 529.17040 [(M + H)+, calcd for C26 H24 N4 O5 F3 529.16933].

PATENT

WO 2011017578

Example 14

(S)-l-((S)-2-Hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-l,2,4- oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylic acid

Figure imgf000099_0001

Preparation 14A: (3S)-Ethyl l-(2-(4-cyanophenyl)-2-hydroxyethyl)piperidine-3- carboxylate

Figure imgf000099_0002

(14A)-isomer A (14A)-isomer B [00210] To a mixture of (S)-ethyl piperidine-3-carboxylate (1.3 g, 8.27 mmol) in toluene (50 mL) was added 4-(2-bromoacetyl)benzonitrile (2.4 g, 10.71 mmol). The reaction mixture was stirred overnight. LCMS indicated completion of reaction. MeOH (10 mL) was added to the mixture, followed by the portionwise addition of sodium borohydride (0.313 g, 8.27 mmol). After 1 hour, LCMS show complete reduction to the desired alcohol. The reaction was quenched with water. The reaction mixture was diluted with ethyl acetate and washed with saturated NaCl. The organic layer was dried with MgSO4, filtered, concentrated, and purified on a silica gel cartridge using an EtOAc/hexanes gradient to yield 2.0 g of solid product. The product was separated by chiral HPLC (Berger SFC MGIII instrument equipped with a CHIRALCEL® OJ (25 x 3 cm, 5 μM). Temp: 30 0C; Flow rate: 130 mL/min; Mobile phase: C(V(MeOH +

0.1%DEA) in 9: 1 ratio isocratic:

[00211] Peak 1 (Isomer A): RT = 2.9 min. for (S)-ethyl l-((S)-2-(4-cyanophenyl)-2- hydroxyethyl)piperidine-3-carboxylate (>99% d.e.). The absolute and relative stereochemistry of compound 14A-isomer A was assigned (S,S) by X-ray crystal structure (see Alternative Route data). 1H NMR (400 MHz, CDCl3) δ ppm 7.63 (2 H, m, J=8.35 Hz), 7.49 (2 H, m, J=8.35 Hz), 4.77 (1 H, dd, J=10.55, 3.52 Hz), 4.17 (2 H, q, J=7.03 Hz), 3.13 (1 H, d, J=9.23 Hz), 2.53-2.67 (3 H, m), 2.44 (2 H, dd, J=18.68, 9.89 Hz), 2.35 (1 H, dd, J=12.74, 10.55 Hz), 1.87-2.01 (1 H, m), 1.71-1.82 (1 H, m), 1.52-1.70 (2 H, m), 1.28 (3 H, t, J=7.03 Hz).

[00212] Peak 2 (Isomer B): RT = 3.8 min for (S)-ethyl l-((R)-2-(4-cyanophenyl)-2- hydroxyethyl)piperidine-3-carboxylate (>99% d.e.). The absolute and relative stereochemistry of 14A-isomer B was assigned (S,R) based on the crystal structure of 14A-isomer A. 1H NMR (400 MHz, CDCl3) δ ppm 7.63 (2 H, m, J=8.35 Hz), 7.49 (2 H, m, J=8.35 Hz), 4.79 (1 H, dd, J=10.55, 3.52 Hz), 4.16 (2 H, q, J=7.03 Hz), 2.69-2.91 (3 H, m), 2.60-2.68 (1 H, m), 2.56 (1 H, dd, J=12.30, 3.52 Hz), 2.36 (1 H, dd, J=12.52, 10.77 Hz), 2.25 (1 H, t, J=8.79 Hz), 1.65-1.90 (3 H, m), 1.52-1.64 (1 H, m, J=12.69, 8.49, 8.49, 4.17 Hz), 1.27 (3 H, t, J=7.25 Hz).

[00213] (S)-Ethyl l-((S)-2-(4-cyanophenyl)-2-hydroxyethyl)piperidine-3-carboxylate (14A-isomer A) was carried forward to make Example 14 and (S)-ethyl l-((R)-2-(4- cyanophenyl)-2-hydroxyethyl)piperidine-3-carboxylate (14A-isomer B) was carried forward to make Example 15.

Preparation 14B: (S)-Ethyl l-((S)-2-hydroxy-2-(4-((Z)-N’-hydroxycarbamimidoyl) phenyl)ethyl)piperidine-3 -carboxylate

Figure imgf000100_0001

[00214] To a mixture of ((S)-ethyl l-((S)-2-hydroxy-2-(4-((Z)-N’- hydroxycarbamimidoyl) phenyl)ethyl)piperidine-3 -carboxylate (14A-Isomer A) (58 mg, 0.192 mmol) and hydroxylamine hydrochloride (26.7 mg, 0.384 mmol) in 2-propanol (10 mL) was added sodium bicarbonate (64.5 mg, 0.767 mmol). The reaction mixture was heated at 85 0C. The reaction mixture was diluted with ethyl acetate and washed with sat NaCl. The organic layer was dried with MgSO4, filtered, and concentrated to yield 56 mg. MS (M+l) = 464. HPLC Peak RT = 1.50 minutes.

Preparation 14C: (S)-Ethyl l-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl) isoxazol-5-yl)-l,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylate

Figure imgf000101_0001

[00215] 3-Phenyl-4-(trifluoromethyl)isoxazole-5-carbonyl fluoride, InM-G (214 mg, 0.78 mmol) was dissolved in acetonitrile (5.00 mL). DIEA (0.272 mL, 1.555 mmol) and (S)-ethyl- 1 -((S)-2-hydroxy-2-(4-((Z)-N’-hydroxycarbamimidoyl) phenyl)ethyl)- piperidine-3-carboxylate (261 mg, 0.778 mmol) were added. The reaction mixture was stirred for 2 hours, then IM TBAF in THF (0.778 mL, 0.778 mmol) was added. The reaction mixture was stirred overnight at room temperature. The reaction mixture was filtered and purified by HPLC in three batches. HPLC conditions: PHENOMENEX® Luna C18 5 micron column (250 x 30mm); 25-100% CH3CN/water (0.1% TFA); 25 minute gradient; 30 mL/min. Isolated fractions with correct mass were partitioned between EtOAc and saturated NaHCO3 with back extracting aqueous layer once. The organic layer was dried with MgSO4, filtered, and concentrated to give 155mg of (S)- ethyl l-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-l,2,4- oxadiazol-3-yl)phenyl)ethyl) piperidine-3-carboxylate. 1H NMR (400 MHz, MeOH-d3) δ ppm 8.04 (2 H, d, J=8.13 Hz), 7.55-7.60 (2 H, m), 7.41-7.54 (5 H, m), 4.81 (1 H, ddd, J=8.35, 4.06, 3.84 Hz), 3.96-4.10 (2 H, m), 2.82-3.08 (1 H, m), 2.67-2.82 (1 H, m), 2.36- 2.61 (3 H, m), 2.08-2.33 (2 H, m), 1.73-1.87 (1 H, m, J=8.54, 8.54, 4.45, 4.17 Hz), 1.32- 1.70 (3 H, m), 1.09-1.19 (3 H, m). MS (m+l) = 557. HPLC Peak RT = 3.36 minutes. Purity = 99%.

Example 14: [00216] (S)-Ethyl l-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5- yl)-l,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylate (89 mg, 0.16 mmol) was heated at 50 0C in 6N HCl (5 mL) in acetonitrile (5 mL). The reaction mixture was stirred overnight and then filtered and purified by HPLC. HPLC conditions:

PHENOMENEX® Luna C 18 5 micron column (250 x 30mm); 25-100% CH3CN/water (0.1% TFA); 25 minute gradient; 30 mL/min. Isolated fractions with correct mass were freeze-dried overnight to yield 36 mg of (S)-l-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4- (trifluoromethyl)isoxazol-5-yl)-l,2,4-oxadiazol-3-yl)phenyl)ethyl) piperidine-3- carboxylic acid as a TFA salt. 1H NMR (400 MHz, MeOH-d3) δ ppm 8.23 (2 H, d, J=8.35 Hz), 7.65-7.74 (4 H, m), 7.54-7.65 (3 H, m), 5.29 (1 H, t, J=7.03 Hz), 4.00 (1 H, br. s.), 3.43-3.75 (1 H, m), 3.34-3.41 (2 H, m), 2.82-3.24 (2 H, m), 2.26 (1 H, d, J=I 1.86 Hz), 1.84-2.14 (2 H, m), 1.52-1.75 (1 H, m). MS (m+1) = 529. HPLC Peak RT = 3.24 minutes. Purity = 98%. Example 14-Alternate Synthesis Route 1

Preparation 14D (Alternate Synthesis Route 1): (S)-4-(Oxiran-2-yl)benzonitrile

Figure imgf000102_0001

[00217] To 800 mL of 0.2M, pH 6.0 sodium phosphate buffer in a 2 L flask equipped with an overhead stirrer was added D-glucose (38.6 g, 1.2 eq), β-nicotinamide adenine dinucleotide, free acid (1.6 g, mmol), glucose dehydrogenase (36 mg, 3.2 kU,

CODEXIS® GDH- 102, 90 U/mg), and enzyme KRED-NADH-110 (200 mg,

CODEXIS®, 25 U/mg). The vessels containing the reagents above were rinsed with 200 mL of fresh sodium phosphate buffer and added to the reaction which was stirred to dissolution and then heated to 40 0C. To this mixture was added a solution of 2-bromo- 4′-cyanoacetophenone (40 g, 178.5 mmol) in 100 mL DMSO through an addition funnel in about 30 min. The container was rinsed with 20 mL DMSO and the rinse was added to the reactor. A pH of 5.5-6.0 was maintained by adding 1 M NaOH through a fresh addition funnel (total volume of 200 mL over 6h) after which HPLC showed complete consumption of the starting material. The reaction mixture was extracted with 800 mL MTBE x 2 and the combined extracts were washed with 300 mL of 25% brine. The crude alcohol was transferred to a 3L 3-neck flask and treated with solid NaOtBu (34.3 g, 357 mmol) stirring for 1 h and then additional NaOtBu (6.9 g, 357 mmol) and stirring for 30 min. The reaction mixture was filtered and the solution was washed with 300 mL 0.2 M pH 6.0 sodium phosphate buffer, brine, and then the solvent was removed in vacuo and the resulting white solid was dried in a vacuum oven to give (S)-4-(oxiran-2- yl)benzonitrile (23 g, 90% yield, 100% e.e.). 1H NMR (400 MHz, CDCl3) δ ppm 7.62 (2 H, d), 7.35 (2 H, d), 3.88 (1 H, dd), 3.18 (1 H, app t), 2.73 (1 H, dd) Purity = 99%.

[00218] Chiral HPLC was done on a CHIRALP AK® AD-RH 4.6x150mm (Daicel Chemical Industries Ltd.) column using gradient of solvent A (10 mM NH4OAc in water/acetonitrile, 90: 10) and solvent B (10 mM NH4OAc in water/acetonitrile, 10:90) with 70% to 90% in 40 min at a flow rate of 0.5 ml/min at ambient temperature. The detection employed UV at 235 nm. The retention times are as follows:

[00219] Peak 1 (Isomer A): RT = 16.7 min. for (S)-4-(oxiran-2-yl)benzonitrile

[00220] Peak 2 (Isomer B): RT = 14.0 min. for (R)-4-(oxiran-2-yl)benzonitrile Preparation of 14A-isomer A (Alternate Synthesis Route 1): (S)-Ethyl l-((S)-2-(4- cyanophenyl)-2 -hydroxy ethyl)piperidine-3-carboxylate

Figure imgf000103_0001

(14A)-isomer A

[00221] (S)-4-(Oxiran-2-yl)benzonitrile (10.00 g, 68.9 mmol), (S)-ethyl piperidine-3- carboxylate (10.83 g, 68.9 mmol) and iPrOH (100 mL) was charged into a round bottom flask under N2. After heating at 55 0C for 4 hours, 4-dimethylaminopyridine (1.683 g, 13.78 mmol) was then added. The reaction mixture was then heated to 50 0C for an additional 12 hours. At this time HPLC indicated the starting material was completely converted to the desired product. The reaction mixture was then cooled to room temperature. EtOAc (120 ml) was added, followed by 100 ml of water. The organic layer was separated, extracted with EtOAc (2x 100 mL) and concentrated under vacuo to give a crude product. The crude product was recrystallized from EtOH/EtOAc/H2O (3/2/2) (8ml/lg) to give a crystalline off-white solid 14A-alt (15 g, 72% yield, 99.6% e.e.). The absolute and relative stereochemistry was determined by single X-ray crystallography employing a wavelength of 1.54184 A. The crystalline material had an orthorhombic crystal system and unit cell parameters approximately equal to the following:

a = 5.57 A α = 90.0°

b = 9.7l A β = 90.0°

c = 30.04 A γ = 90.0°

Space group: P212121

Molecules/asymmetric unit: 2

Volume/Number of molecules in the unit cell = 1625 A3

Density (calculated) = 1.236 g/cm3

Temperature 298 K.

Preparation 14E (Alternate Route 1): (S)-Ethyl l-((S)-2-(tert-butyldimethylsilyloxy)-2- (4-cyanophenyl)ethyl)piperidine-3-carboxylate

Figure imgf000104_0001

[00222] To a mixture of (S)-ethyl 1 -((S)-2-(4-cyanophenyl)-2-hydroxy ethyl) piperidine-3-carboxylate (17.0 g, 56.2 mmol) and DIPEA (17.68 ml, 101 mmol) in CH2Cl2 (187 mL) was added tert-butyldimethylsilyl trifluoromethanesulfonate (16 ml, 69.6 mmol) slowly. The reaction was monitored with HPLC. The reaction completed in 2 hours. The reaction mixture (a light brown solution) was quenched with water, the aqueous layer was extracted with DCM. The organic phase was combined and dried with Na2SO4. After concentration, the crude material was further purified on a silica gel cartridge (33Og silica, 10-30% EtOAc/hexanes gradient) to afford a purified product (S)- ethyl 1 -((S)-2-(tert-butyldimethylsilyloxy)-2-(4-cyanophenyl)ethyl) piperidine-3 – carboxylate (22.25 g, 53.4 mmol, 95 % yield). 1H NMR (400 MHz, CDCl3) δ ppm 7.61 (2 H, d), 7.45 (2 H, d), 4.79 (1 H, m), 4.15 (2 H, m), 2.88 (1 H, m), 2.75 (1 H, m), 2.60 (1 H, dd), 2.48 (1 H, m), 2.40 (1 H, dd), 2.33 (1 H, tt), 2.12 (1 H, tt), 1.90 (1 H, m), 1.68 (1 H, dt), 1.52 (1 H, m), 1.48 (1 H, m), 1.27 (3 H, t), 0.89 (9 H, s), 0.08 (3 H, s), -0.07 (3 H, s).

Preparation 14F (Alternate Route 1): (S)-Ethyl l-((S)-2-(tert-butyldimethylsilyloxy)-2- (4-((Z)-N’-hydroxycarbamimidoyl)phenyl)ethyl)piperidine-3-carboxylate

Figure imgf000105_0001

[00223] (S)-Ethyl- 1 -((S)-2-(tert-butyldimethylsilyloxy)-2-(4-cyanophenyl)ethyl) piperidine-3-carboxylate (31.0 g, 74.4 mmol) was dissolved in EtOH (248 mL).

Hydroxylamine (50% aq) (6.84 ml, 112 mmol) was added and stirred at room temperature overnight. Then all volatiles were removed with ROTA VAPOR®. The residue was purified with on a silica gel cartridge (33Og silica, 0-50% EtOAc/hexanes gradient) to give (S)-ethyl l-((S)-2-(tert-butyldimethylsilyloxy)-2-(4-((Z)-N’- hydroxycarbamimidoyl)phenyl)ethyl)piperidine-3-carboxylate (31 g, 68.9 mmol, 93 % yield) as a white foam. 1H NMR (400 MHz, CDCl3) δ ppm 8.38 (1 H, br s), 7.58 (2 H, d), 7.37 (2 H, d), 4.88 (2 H, br s), 4.81 (1 H, m), 4.13 (2 H, m), 2.96 (1 H, m), 2.82 (1 H, m), 2.61 (1 H, dd), 2.51 (1 H, m), 2.42 (1 H, dd), 2.32 (1 H, tt), 2.13 (1 H, dt), 1.91 (1 H, m), 1.66 (1 H, dt), 1.58 (1 H, m), 1.48 (1 H, m), 1.27 (3 H, t), 0.89 (9 H, s), 0.08 (3 H, s), -0.09 (3 H, s). Preparation 14G (Alternate Route 1): (S)-Ethyl l-((S)-2-(tert-butyldimethylsilyloxy)-2- (4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-l,2,4-oxadiazol-3- yl)phenyl)ethyl)piperidine-3-carboxylate

Figure imgf000105_0002

[00224] (S)-Ethyl- 1 -((S)-2-(tert-butyldimethylsilyloxy)-2-(4-((Z)-N’- hydroxycarbamimidoyl)phenyl)ethyl)piperidine-3-carboxylate (32.6g, 72.5 mmol) was dissolved in acetonitrile (145 ml) (anhydrous) and cooled to ~3 0C with ice-bath. 3- phenyl-4-(trifluoromethyl)isoxazole-5-carbonyl chloride (19.98 g, 72.5 mmol) was dissolved in 5OmL anhydrous acetonitrile and added dropwise. The internal temperature was kept below 10 0C during addition. After addition, the reaction mixture was allowed to warm to room temperature. At 30 minutes, HPLC showed completion of the first reaction step. The reaction mixture was re-cooled to below 10 0C. DIEA (18.99 ml, 109 mmol) was added slowly. After the addition, the reaction mixture was heated up to 55 0C for 17 hr s. HPLC/LCMS showed completion of the reaction. The solvents were removed by ROTA VAPOR®. The residue was stirred in 25OmL 20% EtOAc/hexanes and the DIPEA HCl salt precipitated from solution and was removed via filtration. The filtrate was concentrated and purified using a silica gel cartridge (3X33Og silica, 0-50%

EtOAc/hexanes gradient). (S)-ethyl l-((S)-2-(tert-butyldimethylsilyloxy)-2-(4-(5-(3- phenyl-4-(trifluoromethyl)isoxazol-5-yl)-l,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3- carboxylate (43g, 64.1 mmol, 88 % yield) was obtained a light yellow oil. 1H NMR (400 MHz, CDCl3) δ ppm 8.16 (2 H, d), 7.68 (2 H, d), 7.57 (5 H, m), 4.85 (1 H, m), 4.14 (2 H, m), 2.95 (1 H, m), 2.82 (1 H, m), 2.64 (1 H, dd), 2.51 (1 H, m), 2.49 (1 H, dd), 2.35 (1 H, tt), 2.14 (1 H, dt), 1.91 (1 H, m), 1.66 (1 H, dt), 1.57 (1 H, m), 1.48 (1 H, m), 1.27 (3 H, t), 0.92 (9 H, s), 0.11 (3 H, s), -0.05 (3 H, s).

Example 14 (Alternate Route 1): (S)-l-((S)-2-Hydroxy-2-(4-(5-(3-phenyl-4- (trifluoromethyl)isoxazol-5-yl)-l,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3- carboxylic acid

Figure imgf000106_0001

[00225] (S)-Ethyl l-((S)-2-(tert-butyldimethylsilyloxy)-2-(4-(5-(3-phenyl-4- (trifluoromethyl)isoxazol-5-yl)-l,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3- carboxylate (42g, 62.6 mmol) was dissolved in dioxane (150 ml) and treated with 6M HCl (150 ml). The reaction mixture was heated to 65 0C for 6 hours (the reaction was monitored with HPLC, EtOH was distilled out to push the equilibrium forward). Dioxane was removed and the residue was redissolved in ACN/water and lyophilized separately to give crude (S)-l-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl) isoxazol-5-yl)- l,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylic acid, HCl, (37g crude foamy solid). The crude solid (36 g, 63.7 mmol) was suspended in acetonitrile (720 mL) and heated to 60 0C and water (14.4 mL) was added dropwise. A clear solution was obtained, which was cooled to room temperature and concentrated to a viscous oil, treated with ethyl acetate (1.44 L) with vigorously stirring, heated to 60 0C, and cooled to room temperature. (S)-l-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)- l,2,4-oxadiazol-3-yl)phenyl)ethyl) piperidine-3-carboxylic acid, HCl (28g, 49.3 mmol, 77 % yield) was collected and vacuum dried. Characterization of product by 1H NMR and chiral HPLC matched Example 14 prepared in previous synthesis.

Preparation of Intermediate (14A)-isomer A-Alternate Route 2; 2-Steps: (S)-Ethyl 1- ((S)-2-(4-cyanophenyl)-2-hydroxyethyl)piperidine-3-carboxylate

Figure imgf000107_0001

(14A)-isomer A

Step 1 : Preparation (14D) (Alternate Route 2): (S)-Ethyl l-(2-(4-cyanophenyl)-2- oxoethyl)piperidine-3-carboxylate hydrobromide

Figure imgf000107_0002

(14D)-isomer A

[00226] To a solution of commercially available (S)-ethyl piperidine-3-carboxylate (10 g, 63.6 mmol) in 200 mL toluene was added 4-(2-bromoacetyl)benzonitrile (17g, 76 mmol). The reaction mixture was stirred overnight. The next day, the precipitated solid was collected by filtration and washed with ethyl acetate (x3) and dried under vacuum to give 15.2g of (S)-ethyl l-(2-(4-cyanophenyl)-2-oxoethyl)piperidine-3-carboxylate hydrobromide. MS (M+ 1) = 301. HPLC Peak RT = 1.51 minutes.

Step 2: Preparation of 14 A-isomer A (Alternate Route 2): (S)-Ethyl l-((S)-2-(4- cyanophenyl)-2-hydroxyethyl)piperidine-3 -carboxylate

[00227] Phosphate buffer (1100 mL, BF045, pH 7.0, 0. IM) was added into two liter jacketed glass reactor. The temperature of the reactor was adjusted to 20 0C with the help of a circulator and the reaction mixture was stirred with a magnetic stirrer. Dithiothretol (185.2 mg, 1 mM), magnesium sulfate (288.9 mg, 2 mM), and D-glucose (11.343 g, 62.95 m moles) were added into the reactor. (5*)-Ethyl l-(2-(4-cyanophenyl)-2-oxoethyl) piperidine-3 -carboxylate HBr salt (12 g, 31.47 m moles dissolved in 60 mL DMSO) was added into the reactor slowly with continuous stirring, β-nicotinamide adenine dinucleotide phosphate sodium salt (NADP), 918.47 mg, glucose dehydrogenase, 240 mg (total 18360 U, 76.5 U/mg, ~ 15U/mL, Amano Lot. GDHY1050601) and KRED-114, 1.2 g (CODEXIS® assay 7.8 U/mg of solid), were dissolved in 2.0 mL, 2.0 mL and 10 ml of the same buffer, respectively. Next, NADP, GDH and KRED-114 were added to the reactor in that order. The remaining 26 mL of same buffer was used to wash the NADP, GDH and KRED-114 containers and buffer was added into the same reactor. The starting pH of the reaction was 7.0 which decreased with the progress of the reaction and was maintained at pH 6.5 during the course of the reaction (used pH stat, maintained with IM NaOH). The reaction was run for 4.5 hours and immediately stopped and extracted with ethyl acetate. The ethyl acetate solution was evaporated under reduced pressure and weight of the dark brown residue was 12.14 g. The product was precipitated with dichloromethane and heptane to give 9 g of crude product which was further purified by dissolving it in minimum amount of dichloromethane and re-precipitating by the addition of excess amount of heptane to give 5.22 g. The process was repeated to give an additional 2.82 g of highly pure product for a total of 8.02 g of de > 99.5%.

[00228] Chiral HPLC was done on a CHIRALP AK® AD-RH 4.6x150mm (Daicel Chemical Industries Ltd.) column using gradient of solvent A (10 mM NH4OAc in water/acetonitrile, 90: 10) and solvent B (IO mM NH4OAc in water/acetonitrile, 10:90) with 70% to 90% in 40 min at a flow rate of 0.5 ml/min at ambient temperature. The detection was done by UV at 235 nm. The retention times are as follows: [00229] Peak 1 (14A-isomer A): RT = 20.7 min. for (S)-ethyl l-((S)-2-(4- cyanophenyl)-2-hydroxyethyl)piperidine-3-carboxylate.

[00230] Peak 2 (14B-isomer B): RT = 30.4 min. for (S)-ethyl l-((R)-2-(4- cyanophenyl)-2-hydroxyethyl)piperidine-3-carboxylate.

[00231] Compound 14A-isomer A prepared using this asymmetric method was unambiguously assigned since it was identical to the 14A-isomer A (by 1H NMR and chiral HPLC retention time) that was prepared above and determined by X-ray crystallography. Synthesis of Example 14 from this material followed the same route as described above.

paper

Regioselective Epoxide Ring Opening for the Stereospecific Scale-Up Synthesis of BMS-960, A Potent and Selective Isoxazole-Containing S1P1Receptor Agonist

Discovery Chemistry, Bristol-Myers Squibb, Princeton, New Jersey 08540, United States
Chemical & Synthetic Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00366
Abstract Image

This article presents a stereospecific scale-up synthesis of (S)-1-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylic acid (BMS-960), a potent and selective isoxazole-containing S1P1 receptor agonist. The process highlights an enzymatic reduction of α-bromoketone toward the preparation of (S)-bromo alcohol, a key precursor of (S)-4-(oxiran-2-yl)benzonitrile. A regioselective and stereospecific epoxide ring-opening reaction was also optimized along with improvements to 1,2,4-oxadiazole formation, hydrolysis, and crystallization. The improved process was utilized to synthesize batches of BMS-960 for Ames testing and other toxicological studies.

PAPER

Journal of Medicinal Chemistry (2016), 59(13), 6248-6264.

Discovery and Structure–Activity Relationship (SAR) of a Series of Ethanolamine-Based Direct-Acting Agonists of Sphingosine-1-phosphate (S1P1)

Abstract

Abstract Image

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite that regulates a multitude of physiological processes such as lymphocyte trafficking, cardiac function, vascular development, and inflammation. Because of the ability of S1P1 receptor agonists to suppress lymphocyte egress, they have great potential as therapeutic agents in a variety of autoimmune diseases. In this article, the discovery of selective, direct acting S1P1 agonists utilizing an ethanolamine scaffold containing a terminal carboxylic acid is described. Potent S1P1 agonists such as compounds 18a and 19a which have greater than 1000-fold selectivity over S1P3 are described. These compounds efficiently reduce blood lymphocyte counts in rats through 24 h after single doses of 1 and 0.3 mpk, respectively. Pharmacodynamic properties of both compounds are discussed. Compound 19a was further studied in two preclinical models of disease, exhibiting good efficacy in both the rat adjuvant arthritis model (AA) and the mouse experimental autoimmune encephalomyelitis model (EAE).

BASE

(S)-1-((S)-2-Hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl) isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylic Acid (18a)

(S)-ethyl 1-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylate (36%).

1H NMR (400 MHz, MeOH-d3) δ ppm 8.04 (2 H, d, J = 8.13 Hz), 7.55–7.60 (2 H, m), 7.41–7.54 (5 H, m), 4.81 (1 H, ddd, J = 8.35, 4.06, 3.84 Hz), 3.96–4.10 (2 H, m), 2.82–3.08 (1 H, m), 2.67–2.82 (1 H, m), 2.36–2.61 (3 H, m), 2.08–2.33 (2 H, m), 1.73–1.87 (1 H, m, J = 8.54, 8.54, 4.45, 4.17 Hz), 1.32–1.70 (3 H, m), 1.09–1.19 (3 H, m).

MS (M + H)+ at m/z 557. HPLC purity: 99%, tr = 3.36 min (method B).

TFA salt

(S)-1-((S)-2-hydroxy-2-(4-(5-(3-phenyl-4-(trifluoromethyl)isoxazol-5-yl)-1,2,4-oxadiazol-3-yl)phenyl)ethyl)piperidine-3-carboxylic acid, TFA salt (18a, 61%) as a white solid.

1H NMR (400 MHz, MeOH-d3) δ ppm 8.23 (2 H, d, J = 8.35 Hz), 7.65–7.74 (4 H, m), 7.54–7.65 (3 H, m), 5.29 (1 H, t, J = 7.03 Hz), 4.00 (1 H, br s), 3.43–3.75 (1 H, m), 3.34–3.41 (2 H, m), 2.82–3.24 (2 H, m), 2.26 (1 H, d, J = 11.86 Hz), 1.84–2.14 (2 H, m), 1.52–1.75 (1 H, m).

MS (M + H)+ at m/z 529.

HPLC tr = 3.27 min (method B). HPLC purity: 99.4%, tr = 8.78 min (method E); 99.0%, tr = 7.29 min (method F).

HCL SALT

This material was converted to the HCl salt for the following analyses: mp: 219.2 °C. Anal. Calcd for C26H23N4O5F3·HCl: 0.14% water: C, 55.2; H, 4.31; N, 9.87; Cl, 6.25. Found: C, 55.39; H, 4.10; N, 9.88; Cl, 6.34. [α]D20 + 30.47 (c 0.336, MeOH). HPLC with chiral stationary phase (A linear gradient using CO2 (solvent A) and IPA with 0.1% DEA (solvent B); t = 0 min, 30% B, t = 10 min, 55% B was employed on a Chiralcel AD-H 250 mm × 4.6 mm ID, 5 μm column; flow rate was 2.0 mL/min): tr = 5.38 min with >99% ee.

References

Gilmore, J. L.; Sheppeck, J. E.; Watterson, S. H.; Haque, L.; Mukhopadhyay, P.; Tebben, A. J.; Galella, M. A.; Shen, D. R.; Yarde, M.; Cvijic, M. E.; Borowski, V.; Gillooly, K.; Taylor, T.; McIntyre, K. W.; Warrack, B.; Levesque, P. C.; Li, J. P.; Cornelius, G.; D’Arienzo, C.; Marino, A.; Balimane, P.; Salter-Cid, L.; Barrish, J. C.; Pitts, W. J.; Carter, P. H.; Xie, J.; Dyckman, A. J.Discovery and Structure Activity Relationship (SAR) of a Series of Ethanolamine-Based Direct-Acting Agonists of Sphingosine-1-Phosphate (S1P1) J. Med. Chem. 2016, 59, 62486264, DOI: 10.1021/acs.jmedchem.6b00373
Gilmore, J. L.; Sheppeck, J. E. Preparation of 3-(4-(1-hydroxyethyl)phenyl)-1,2,4-oxadiazole derivatives as sphingosine-1-phosphate receptor agonists for the treatment of autoimmune disease and inflammation. PCT Int. Appl. 2011, WO 2011017578.

//////BMS-960, PRECLINICAL, BMS 960

Cl.O=C(O)[C@H]1CCCN(C1)C[C@@H](O)c2ccc(cc2)c3nc(on3)c5onc(c4ccccc4)c5C(F)(F)F


Filed under: Preclinical drugs, Uncategorized Tagged: BMS-960, preclinical

Hoshinolactam, A new antitrypanosomal lactam

$
0
0

Abstract Image
Tropical diseases caused by parasitic protozoa are a threat to human health, mainly in developing countries. Trypanosomiasis (Chagas disease and sleeping sickness) and leishmaniasis, inter alia, are classified as neglected tropical diseases, and over 400 million people are at risk of contracting these diseases.

In addition, a parasite of the Trypanosoma genus, Trypanosoma brucei brucei, is the causative agent of Nagana disease in wild and domestic animals, and this disease is a major obstacle to the economic development of affected rural areas.

Although some therapeutic agents for these diseases exist, they have limitations, such as serious side effects and the emergence of drug resistance. Thus, new and more effective antiprotozoal medicines are needed

Marine natural products have recently been considered to be good sources for drug leads. In particular, secondary metabolites produced by marine cyanobacteria have unique structures and versatile biological activities, and some of these compounds show antiprotozoal activities. For example, coibacin A isolated from cf. Oscillatoria sp. exhibited potent antileishmanial activity, and viridamide A isolated from Oscillatoria nigro-viridis showed antileishmanial and antitrypanosomal activities.

constituents of marine cyanobacteria and reported an antitrypanosomal cyclodepsipeptide, janadolide.

The marine cyanobacterium was collected at the coast near Hoshino, Okinawa.

Image result for OKINAWA

Image result for OKINAWA

Okinawa
沖縄市
Uchinaa
City
Okinawa City downtown.jpg
Flag of Okinawa
Flag

EARLIER MERCK TEAM HAD REPORTED

CAS 159153-15-8
MF C20 H33 N O5
MW 367.48
2-Pyrrolidinone, 3,4-dihydroxy-5-(hydroxymethyl)-3-[3-(2-nonylcyclopropyl)-1-oxo-2-propenyl]-, [3S-[3α,3[E(1S*,2S*)],4β,5α]]-
Image result for AntitrypanosomalImage result for Antitrypanosomal
Antitrypanosomal
Image result for marine cyanobacterium
Marine cyanobacterium
Image result for human fetal lung fibroblast MRC-5 cells
Human fetal lung fibroblast MRC-5 cells
Majusculoic acid.png
Majusculoic acid
Image result for malyngamide A.
Malyngamide A.

PAPER

http://pubs.acs.org/doi/suppl/10.1021/acs.orglett.7b00047

Recently, we isolated a new antitrypanosomal lactam, hoshinolactam (1), from a marine cyanobacterium.Structurally, 1 contains a cyclopropane ring and a γ-lactam ring. So far, some metabolites possessing either a cyclopropane ring or a γ-lactam ring have been discovered from marine cyanobacteria, such as majusculoic acid and malyngamide A. To the best of our knowledge, on the other hand, hoshinolactam (1) is the first compound discovered in marine cyanobacteria that possesses both of these ring systems. In addition, we clarified that 1 exhibited potent antitrypanosomal activity without cytotoxicity against human fetal lung fibroblast MRC-5 cells. Here, we report the isolation, structure elucidation, first total synthesis, and preliminary biological characterization of hoshinolactam (1).

Isolation and Total Synthesis of Hoshinolactam, an Antitrypanosomal Lactam from a Marine Cyanobacterium

Department of Chemistry, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
Research Center for Tropical Diseases, Kitasato Institute for Life Sciences, and §Graduate School of Infection Control Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
Org. Lett., Article ASAP
DOI: 10.1021/acs.orglett.7b00047

Abstract Image

In the search for new antiprotozoal substances, hoshinolactam, an antitrypanosomal lactam, was isolated from a marine cyanobacterium. The gross structure was elucidated by spectroscopic analyses, and the absolute configuration was determined by the first total synthesis. Hoshinolactam showed potent antitrypanosomal activity with an IC50 value of 3.9 nM without cytotoxicity against human fetal lung fibroblast MRC-5 cells (IC50 > 25 μM).

Table 1. 1H and 13C NMR Data for 1 in C6D6
unit position δCa δHb (J in Hz)
HIMP 1 177.8, C
2 44.1, CH 2.51, dq (5.2, 7.6)
3 80.8, CH 4.94, dd (4.6, 5.2)
4 57.3, CH 3.49, ddd (4.6, 4.7, 9.4)
5a 44.6, CH2 1.21, m
5b 1.36, m
6 25.0, CH 1.61, m
7 21.7, CH3 0.74, d (6.2)
8 23.2, CH3 0.76, d (6.3)
9 15.0, CH3 1.33, d (7.6)
NH 7.65, s
PCPA 1 166.0, C
2 117.4, CH 5.88, d (15.5)
3 155.0, CH 6.59, dd (10.3, 15.5)
4 22.4, CH 0.91, m
5 23.3, CH 0.59, m
6 35.7, CH2 0.96, m
7 22.5, CH2 1.20, tq (7.1, 7.3)
8 14.0, CH3 0.78, t (7.3)
9a 16.1, CH2 0.35, ddd (4.5, 6.0, 8.2)
9b 0.42, ddd (4.5, 4.5, 8.8)
aMeasured at 100 MHz.
bMeasured at 400 MHz.
Positive HRESIMS data (m/z 308.2228, calcd for C18H30NO3 [M + H]+ 308.2225). Table 1 shows the NMR data for 1.
An analysis of the 1H NMR spectrum indicated the presence of four methyl groups (δH 0.74, 0.76, 0.78 and 1.33), four protons of the cyclopropane ring (δH 0.35, 0.42, 0.59 and 0.91), and two olefinic protons (δH 5.88 and 6.59).
The 13C NMR and HMQC spectra revealed the existence of two carbonyl groups (δC 166.0 and 177.8) and two sp2 methines (δC 117.4 and 155.0).
Examination of the COSY and HMBC spectra established the presence of two fragments derived from 4-hydroxy-5-isobutyl-3-methylpyrrolidin-2-one (HIMP) and 3-(2-propylcyclopropyl) acrylic acid (PCPA), respectively. The configuration of the C-2–C-3 olefinic bond in the PCPA was determined to be trans on the basis of the coupling constant (3JH2–H3 = 15.5 Hz). The connectivity of the two partial structures was determined from the HMBC correlation (H-3 of HIMP/C-1 of PCPA).
1H, 13C, COSY, HMQC, HMBC, and NOESY NMR spectra in C6D6 and 1H and 13C NMR spectra in CD3OD for hoshinolactam (1)
1H, 13C, COSY, HMQC, HMBC, and NOESY NMR spectra in C6D6

1H and 13C NMR spectra in CD3OD

1H NMR PREDICT

13 C NMR PREDICT

Image result for OKINAWAImage result for OKINAWA

OKINAWA

///////////Hoshinolactam

CC(C)C[C@@H]2NC(=O)[C@H](C)C2OC(=O)/C=C/[C@H]1C[C@@H]1CCC


Filed under: Preclinical drugs, Uncategorized Tagged: Hoshinolactam

AMG-3969

$
0
0

Image result for amg 3969

AMG-3969

M.Wt: 522.46
Cas : 1361224-53-4 , MF: C21H20F6N4O3S

WO 2012027261 PRODUCT PATENT

Inventors Kate Ashton, Michael David Bartberger, Yunxin Bo, Marian C. Bryan, Michael Croghan, Christopher Harold Fotsch, Clarence Henderson Hale, Roxanne Kay Kunz, Longbin Liu, Nobuko Nishimura, Mark H. Norman, Lewis Dale Pennington, Steve Fong Poon, Markian Myroslaw Stec, Jean David Joseph St., Jr., Nuria A. Tamayo, Christopher Michael Tegley, Kevin Chao Yang
Applicant Amgen Inc.

2-[4-[(2S)-4-[(6-Amino-3-pyridinyl)sulfonyl]-2-(1-propyn-1-yl)-1-piperazinyl]phenyl]-1,1,1,3,3,3-hexafluoro-2-propanol)

(S)-2-(4-(4-((6-Aminopyridin-3-yl)sulfonyl)-2-(prop-1-yn-1-yl)piperazin-1-yl)phenyl)-1,1,1,3,3,3-hexafluoropropan-2-ol,

mp 113–123 °C;
[α]D20 = +75.1 (c = 2.2, MeOH).
Agents for Type 2 Diabetes,  PRECLINICAL

AMG-3969, a novel and stable small-molecule disruptor of glucokinase (GK) and glucokinase regulatory protein (GKRP) interaction by the optimization of initial screening hit and AMG-1694. AMG-3969 potently induced the dissociation of the GK-GKRP complex and promoted GK translocation both in-vitro and in-vivo. In rodent model of diabetes, AMG-3969 reduced blood glucose levels without affecting euglycemic animals. The study represents the first successful discovery of a small molecule that targets the GK-GKRP complex as a novel pathway for managing blood glucose levels with reduced hypoglycemic risk.

Image result for AMGEN

 Kate Ashton

Kate Ashton

Senior Scientist at Amgen, Inc

Amgen
Thousand Oaks, United States
Dr. Kate Ashton received a Masters in Chemistry with Industrial Experience from the University of Edinburgh. She conducted her PhD thesis research on the synthesis and structure elucidation of Reidispongiolide A with Prof. Ian Paterson at the University of Cambridge, and her postdoctoral work on SOMO catalysis with Prof. David W. C. MacMillan at both Caltech and Princeton. She has been at Amgen for 6 years and has worked on indications for cancer, Alzheimer’s and diabetes.Dr Fecke works in the area of industrial early drug discovery since 1996. He is currently Group Leader in the Primary Pharmacology department at UCB Pharma (UK) and is involved in the identification and characterization of NCE and NBE drugs in molecular interaction assays for both immunological and CNS diseases. Prior to joining UCB, he worked for Novartis and Siena Biotech in the areas of transplant rejection, neurodegeneration and oncology. He obtained his PhD at the Heinrich-Heine-University Dusseldorf in Germany in 1994.

Image result for amg 3969

(S)-2-(4-(4-((6-Aminopyridin-3-yl)sulfonyl)-2-(prop-1-yn-1-yl)piperazin-1-yl)phenyl)-1,1,1,3,3,3-hexafluoropropan-2-ol, AMG-3969

Glucokinase (GK) is a member of a family of four hexokinases that are critical in the cellular metabolism of glucose. Specifically GK, also known as hexokinase IV or hexokinase D, facilitates glucose induced insulin secretion from pancreatic β-cells as well as glucose conversion into glycogen in the liver. GK has a unique catalytic activity that enables the enzyme to be active within the physiological range of glucose (from 5mM glucose to lOmM glucose).

Genetically modified mouse models support the role of GK playing an important role in glucose homeostasis. Mice lacking both copies of the GK gene die soon after birth from severe hyperglycemia, whereas mice lacking only one copy of the GK gene present with only mild diabetes. Mice that are made to overexpress the GK gene in their livers are hypoglycemic.

Numerous human mutations in the GK gene have been identified, with the vast majority of them resulting in proteins with impaired or absent enzymatic activity. These loss-of-function mutations are thought to contribute to the hyperglycemia seen with maturity-onset diabetes of the young type II (MODY-2). A small fraction of these mutations result in a GK with increased catalytic function. These individuals present with moderate to severe hypoglycemia.

GK activity in the liver is transiently regulated by glucokinase regulatory protein (GKRP). GK catalytic activity is inhibited when GK is bound to GKRP. This interaction is antagonized by increasing concentrations of both glucose and fructose -1 -phosphate (F1P). The complex of the two proteins is localized primarily to the nuclear compartment of a cell. Post prandially as both glucose and fructose levels rise, GK released from GKRP translocates to the cytoplasm. Cytoplasmic GK is now free of the inhibitory effects of GKRP and able to kinetically respond to glucose. Evidence from the Zucker diabetic fatty rat (ZDF) indicates that their glucose intolerance may be a result of this mechanism failing to function properly.

A compound that acts directly on GKRP to disrupt its interaction with GK and hence elevate levels of cytoplasmic GK is a viable approach to modulate GK activity. Such an approach would avoid the unwanted hypoglycemic effects of over stimulation of GK catalytic activity, which has been seen in the

development of GK activators. A compound having such an effect would be useful in the treatment of diabetes and other diseases and/or conditions in which GKRP and/or GK plays a role.

CLIP

Antidiabetic effects of glucokinase regulatory protein small-molecule disruptors
Nature 2013, 504(7480): 437

Image result for Antidiabetic effects of glucokinase regulatory protein small-molecule disruptors.

Image result for Antidiabetic effects of glucokinase regulatory protein small-molecule disruptors.

SYNTHESIS

Figure

aReagents and conditions: (a) 1-propynylmagnesium bromide, THF, 0 °C, 99%; (b) TFA, DCM, then NaBH(OAc)3 77%; (c) NH4OH, EtOH, 120 °C, 88%; (d) chiral SFC, 38%………..Nature 2013,504, 437440

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2012027261

EXAMPLE 241 : 2-(4-(4-((6-AMINO-3-PYRIDINYL)SULFONYL)-2-(l-PROP YN- 1 – YL)- 1 -PIPERAZINYL)PHENYL)- 1,1,1 ,3 ,3 ,3 -HEXAFLUORO-2-PROPANOL

STEP 1 : 4-BENZYL 1 -TERT-BUTYL 2-0X0-1,4-PIPERAZINEDICARBOXYLATE

A 2-L Erlenmeyer flask was charged with 2-piperazinone (36.5 g, 364 mmol, Sigma- Aldrich, St. Louis, MO), sodium carbonate (116 g, 1093 mmol), 600 mL of dioxane, and 150 mL of water. To this was slowly added benzyl chloroformate (62.1 g, 364 mmol, Sigma-Aldrich, St. Louis, MO) at room temperature over 20 min. After the addition was complete, the mixture was stirred for 2 h and then diluted with water and extracted with EtOAc (2 L). The combined organic extracts were dried (MgS04), filtered, and concentrated to give a white solid. To this solid was added 500 mL of DCM, triethylamine (128 mL, 911 mmol), DMAP (4.45 g, 36.4 mmol), and di-tert-butyl dicarbonate (119 g, 546 mmol, Sigma-Aldrich, St. Louis, MO). After 1 h at room temperature, the mixture was diluted with water and the organics were separated. The organics were dried (MgS04), filtered, and concentrated to give a brown oil. To this oil was added 100 mL of DCM followed by 1 L of hexane. The resulting white solid was collected by filtration to give 4-benzyl 1-tert-butyl 2-oxo-l,4-piperazinedicarboxylate (101 g).

STEP 2: BENZYL (2-((TERT-BUTOXYCARBONYL)AMINO)ETHYL)(2-OXO-3 -PENTYN- 1 -YL)CARBAMATE

A 150-mL round-bottomed flask was charged with 4-benzyl 1-tert-butyl

2- oxo-l,4-piperazinedicarboxylate (1.41 g, 4.22 mmol) and THF (5 mL). 1-Propynylmagnesium bromide (0.5 M in THF, 20.0 mL, 10.0 mmol, Sigma-Aldrich, St. Louis, MO) was added at 0 °C slowly. The mixture was stirred at 0 °C for 2 h. Saturated aqueous NH4C1 (40 mL) was added and the aqueous phase was extracted with EtOAc (200 mL, then 2 x 100 mL). The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography (50 g of silica, 0 to 50% EtOAc in hexanes) to afford benzyl (2-((tert-butoxycarbonyl)amino)ethyl)(2-oxo- 3- pentyn-l-yl)carbamate (1.55 g) as a clear oil.

STEP 3: BENZYL 3-(l-PROPYN-l-YL)-l-PIPERAZINECARBOXYLATE

A 3-L round-bottomed flask was charged with 2-((tert-butoxycarbonyl)amino)ethyl)(2-oxo-3-pentyn-l-yl)carbamate (82.2 g, 219 mmol) and 300 mL of DCM. After cooling to -10 °C, TFA (169 mL, 2195 mmol) was added and the resulting dark solution was stirred at room temperature for 15 min. Sodium triacetoxyborohydride (186 g, 878 mmol, Sigma-Aldrich, St. Louis, MO) was then added portion- wise over 10 min. After 2 h, the mixture was

concentrated, diluted with EtOAc (1 L), and neutralized with 5 N NaOH. The layers were separated and the organic extracts were washed with brine, dried (MgS04), filtered and concentrated. The resulting orange oil was purified via column chromatography (750 g of silica gel, 0 to 4.5 % MeOH/DCM) to give benzyl 3-(l-propyn-l-yl)-l-piperazinecarboxylate (43.7 g) as a brown foam.

STEP 4: BENZYL 3-(l-PROPYN-l-YL)-4-(4-(2,2,2-TRIFLUORO-l-HYDROXY- 1 -(TRIFLUOROMETHYL)ETHYL)PHENYL)- 1 -PIPERAZINECARBOXYLATE

A 150-mL reaction vessel was charged with benzyl 3-(prop-l-yn-l-yl)piperazine-l-carboxylate (2.88 g, 11.2 mmol), 2-(4-bromophenyl)-l, 1,1, 3,3,3-hexafluoropropan-2-ol (4.36 g, 13.5 mmol, Bioorg. Med. Chem. Lett. 2002, 12, 3009), dicyclohexyl(2′,6′-diisopropoxy-[ 1 , 1 ‘-biphenyl]-2-yl)phosphine, RuPhos (0.530 g, 1.14 mmol, Sigma- Aldrich, St. Louis, MO), RuPhos Palladacycle (0.417 g, 0.572 mmol, Strem Chemical Inc, Newburyport, MA), sodium tert-butoxide (2.73 g, 28.4 mmol, Strem Chemical Inc, Newburyport, MA) and toluene (35 mL). The mixture was degassed by bubbling Ar through the solution for 10 min. The vessel was sealed and heated at 100 °C for 1.5 h. The reaction mixture was cooled to room temerature and water (100 mL) was added. The aqueous phase was extracted with EtOAc (3 x 100 mL) and the combined organic phases were washed with saturated aqueous sodium chloride (150 mL). The organic extracts were dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography (100 g of silica, 0 to 50% EtOAc in hexanes) to afford benzyl 3-(l-propyn-l-yl)-4-(4-(2,2,2-trifluoro- 1 -hydroxy- 1 -(trifluoromethyl)ethyl)phenyl)- 1 -piperazinecarboxylate as a yellow solid.

STEP 5: 2-(4-(4-((6-CHLORO-3-PYRIDINYL)SULFONYL)-2-(l-PROPYN-l-YL)- 1 -PIPERAZIN YL)PHENYL)- 1,1,1 ,3 ,3 ,3 -HEXAFLUORO-2-PROPANOL

A 500-mL round-bottomed flask was charged with benzyl 3-(l-propyn-l-yl)-4-(4-(2,2,2-trifluoro- 1 -hydroxy- 1 -(trifluoromethyl)ethyl)phenyl)- 1 -piperazinecarboxylate (3.13 g, 6.25 mmol) and TFA (40 mL).

Trifluoromethanesulfonic acid (1.25 mL, 14.1 mmol, Acros/Fisher Scientific, Waltham, MA) was added dropwise at room temperature. After 5 min, additional TfOH (0.45 mL, 5.1 mmol) was added. After an additional 10 min, solid

NaHC03 was carefully added in potions. Saturated aqueous NaHC03 (250 mL) was added slowly to bring pH to approximately 7. The aqueous phase was extracted with EtOAc (100 mL). At this time, more solid NaHC03 was added to the aqueous phase and extracted again with EtOAc (100 mL). The combined organic phases were washed with water (200 mL) and saturated aqueous sodium chloride (200 mL). The combined organic extracts were dried over sodium sulfate, filtered and concentrated in vacuo to afford 3.10 g of tan solid.

A 500-mL round-bottomed flask was charged with this material, triethylamine (5.00 mL, 35.9 mmol) and CH2CI2 (30 mL). 6-Chloropyridine-3-sulfonyl chloride (1.58 g, 7.43 mmol, Organic Process Research & Development 2009, 13, 875) was added in potions at 0 °C. The brown mixture was stirred at 0 °C for 10 min. The volume of the reaction mixture was reduced to approximately 10 mL in vacuo then the mixture was purified twice by column chromatography (100 g of silica, 0 to 50% EtOAc in hexanes) to afford 2-(4-(4-((6-chloro-3-pyridinyl)sulfonyl)-2-( 1 -propyn- 1 -yl)- 1 -piperazinyl)phenyl)- 1,1,1,3,3,3-hexafluoro-2-propanol (3.46 g) as an off-white solid.

STEP 6: 2-(4-(4-((6-AMINO-3-PYRIDINYL)SULFONYL)-2-(l-PROPYN-l-YL)- 1 -PIPERAZIN YL)PHENYL)- 1,1,1 ,3 ,3 ,3 -HEXAFLUORO-2-PROPANOL

A 20-mL sealed tube was charged with 2-(4-(4-((6-chloro-3-pyridinyl)sulfonyl)-2-( 1 -propyn- 1 -yl)- 1 -piperazinyl)phenyl)- 1,1,1,3,3,3-hexafluoro-2-propanol (0.340 g, 0.627 mmol), concentrated ammonium hydroxide (5.00 mL, 38.5 mmol) and EtOH (5 mL). The reaction mixture was heated in an Initiator (Biotage, AB, Uppsala, Sweden) at 120 °C for 1 h. The reaction mixture was further heated in a heating block at 110 °C for 5 h. The reaction mixture was concentrated and purified by column chromatography (25 g of silica, 30 to 80% EtOAc in hexanes) to afford 2-(4-(4-((6-amino-3-pyridinyl)sulfonyl)-2-( 1 -propyn- 1 -yl)- 1 -piperazinyl)phenyl)- 1,1,1,3,3,3-hexafluoro-2-propanol (0.289 g) as a mixture of two enantiomers.

1H NMR (400 MHz, CDC13) δ ppm 8.49 (br. s., 1 H), 7.80 (dd, J= 2.3, 8.8 Hz, 1 H), 7.59 (d, J= 8.8 Hz, 2 H), 6.97 (d, J= 9.0 Hz, 2 H), 6.55 (d, J= 8.8 Hz, 1 H), 5.05 (s, 2 H), 4.46 (br. s., 1 H), 3.85 – 3.72 (m, 2 H), 3.54 (br. s., 1 H), 3.50 – 3.34 (m, 2 H), 2.83 (dd, J= 3.3, 11.0 Hz, 1 H), 2.69 (dt, J= 3.4, 11.0 Hz, 1 H), 1.80 (s, 3 H). m/z (ESI, +ve ion) 523.1 (M+H)+. GK-GKRP IC50 (Binding) = 0.003 μΜ

The individual enantiomers were isolated using chiral SFC. The method used was as follows: Chiralpak® ADH column (21 x 250 mm, 5 μιη) using 35% methanol in supercritical C02 (total flow was 70 mL/min). This produced the two enantiomers with enantiomeric excesses greater than 98%.

2-(4-((2S)-4-((6-amino-3-pyridinyl)sulfonyl)-2-(l -propyn- 1-yl)- 1 -piperazinyl)phenyl)- 1,1,1 ,3 ,3 ,3 -hexafluoro-2-propanol and 2-(4-((2R)-4-((6-amino-3 -pyridinyl)sulfonyl)-2-( 1 -propyn- 1 -yl)- 1 -piperazinyl)phenyl)- 1,1,1,3,3,3-hexafluoro-2-propanol.

FIRST ELUTING PEAK (PEAK #1)

1H NMR (400 MHz, CDC13) δ 8.48 (d, J= 2.3 Hz, 1 H), 7.77 (dd, J= 2.5, 8.8 Hz, 1 H), 7.57 (d, J= 8.8 Hz, 2 H), 6.95 (d, J= 9.2 Hz, 2 H), 6.52 (d, J= 8.8 Hz, 1 H), 4.94 (s, 2 H), 4.44 (br. s., 1 H), 3.82 – 3.71 (m, 2 H), 3.58 – 3.33 (m, 3 H), 2.81 (dd, J= 3.2, 11.1 Hz, 1 H), 2.67 (dt, J= 3.9, 11.0 Hz, 1 H), 1.78 (d, J = 2.2 Hz, 3 H). m/z (ESI, +ve ion) 523.2 (M+H)+. GK-GKRP IC50 (Binding) = 0.002 μΜ.

SECOND ELUTING PEAK (PEAK #2)

1H NMR (400 MHz, CDC13) δ 8.49 (d, J= 1.8 Hz, 1 H), 7.78 (dd, J= 2.3, 8.8 Hz, 1 H), 7.59 (d, J= 8.6 Hz, 2 H), 6.97 (d, J= 9.0 Hz, 2 H), 6.54 (d, J= 8.8 Hz, 1 H), 4.97 (s, 2 H), 4.46 (br. s., 1 H), 3.77 (t, J= 11.7 Hz, 2 H), 3.67 (br. s., 1 H), 3.51 – 3.33 (m, 2 H), 2.82 (dd, J= 3.3, 11.0 Hz, 1 H), 2.68 (dt, J= 3.9, 11.1 Hz, 1 H), 1.79 (d, J= 2.0 Hz, 3 H). m/z (ESI, +ve ion) 523.2 (M+H)+. GK-GKRP IC50 (Binding) = 0.342 μΜ.

Alternative procedure starting after Step 4.

STEP 5 : 2-(4-(4-((6-AMINO-3-PYRIDINYL)SULFONYL)-2-(l-PROPYN-l-YL)- 1 -PIPERAZIN YL)PHENYL)- 1,1,1 ,3 ,3 ,3 -HEXAFLUORO-2-PROPANOL

Alternatively, 2-(4-(4-((6-amino-3-pyridinyl)sulfonyl)-2-( 1 -propyn- 1 -yl)-l-piperazinyl)phenyl)-l,l,l,3,3,3-hexafluoro-2-propanol was synthesized from benzyl 3-( 1 -propyn- 1 -yl)-4-(4-(2,2,2-trifluoro- 1 -hydroxy- 1 -(trifluoromethyl)ethyl)phenyl)- 1 -piperazinecarboxylate as follows.

A 2-L round-bottomed flask was charged with benzyl 3 -(1 -propyn- 1-yl)-4-(4-(2,2,2-trifluoro- 1 -hydroxy- 1 -(trifluoromethyl)ethyl)phenyl)- 1 -piperazinecarboxylate (21.8 g, 43.5 mmol, step 5) and TFA (130 mL).

Trifluoromethanesulfonic acid (11.6 mL, 131 mmol, Acros/Fisher Scientific, Waltham, MA) was added slowly at rt resulting orange cloudy mixture. After stirring at rt for 10 min, the volume of the reaction mixture was reduced to half in vacuo. Solid NaHC03 was added in potions until the mixture became sludge. Saturated aqueous NaHC03(800 mL) was added slowly until the pH was about

8. The aqueous phase was extracted with EtOAc (3 x 250 mL). The combined organic phases were washed with water (500 mL) and saturated aqueous NaCl (500 mL). The organic phase was dried over sodium sulfate, filtered and concentrated in vacuo. This material was dissolved into DCM (200 mL) and triethylamine (31.0 mL, 222 mmol) was added. Then 6-aminopyridine-3-sulfonyl chloride (9.40 g, 48.8 mmol, published PCT patent application no. WO

2009/140309) was added in potions over 10 min period. The brown mixture was stirred at room temperature for 10 min. The reaction mixture was washed with water (300 mL) and saturated aqueous NaCl (300 mL). The organic phase was dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography (780 g of total silica, 30 to 90% EtOAc in hexanes) to afford 2-(4-(4-((6-amino-3-pyridinyl)sulfonyl)-2-(l-propyn-l-yl)-l-piperazinyl)phenyl)-l,l,l,3,3,3-hexafluoro-2-propanol (19.4 g) as a mixture of two enantiomers.

Paper

Nonracemic Synthesis of GK–GKRP Disruptor AMG-3969

Therapeutic Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
Amgen Inc. 360 Binney Street, Cambridge, Massachusetts 02142, United States
J. Org. Chem., 2014, 79 (8), pp 3684–3687

Abstract Image

A nonracemic synthesis of the glucokinase–glucokinase regulatory protein disruptor AMG-3969 (5) is reported. Key features of the synthetic approach are an asymmetric synthesis of the 2-alkynyl piperazine core via a base-promoted isomerization and a revised approach to the synthesis of the aminopyridinesulfonamide with an improved safety profile.

(S)-2-(4-(4-((6-Aminopyridin-3-yl)sulfonyl)-2-(prop-1-yn-1-yl)piperazin-1-yl)phenyl)-1,1,1,3,3,3-hexafluoropropan-2-ol, AMG-3969 (5)

(S)-2-(4-(4-((6-aminopyridin-3-yl)sulfonyl)-2-(prop-1-yn-1-yl)piperazin-1-yl)phenyl)-1,1,1,3,3,3-hexafluoropropan-2-ol (5) (64.0 g, 49% yield) as white solid. The enanatiomeric excess was found to be >99.5% by chiral SFC (see Supporting Information):
1H NMR (400 MHz, CDCl3) δ 8.47 (s, 1 H), 7.79 (d, J = 8.6 Hz, 1 H), 7.59 (d, J = 8.2 Hz, 2 H), 6.97 (d, J = 8.6 Hz, 2 H), 6.55 (d, J = 8.8 Hz, 1 H), 5.06 (br s, 2 H), 4.45 (br s, 1 H), 3.96 (br s, 1 H), 3.77 (t, J = 12.1 Hz, 2 H), 3.50–3.35 (m, 2 H), 2.82 (d, J = 11.0 Hz, 1 H), 2.68 (t, J = 10.9 Hz, 1 H), 1.79 (s, 3 H);
13C NMR (101 MHz, CD3OD) δ 163.8, 152.0, 150.1, 138.2, 129.0, 124.7 (q), 123.9, 121.1, 117.5, 109.3, 82.8, 78.3 (m), 75.5, 52.0, 47.2, 44.9, 3.2;
 
HRMS (ESI-TOF) m/z [M + H]+calcd for C21H21F6N4O3S 523.1239, found 523.1229;
 
mp 113–123 °C;
 
[α]D20 = +75.1 (c = 2.2, MeOH).
 

Clip

AMG-3969 is a disruptor of the glucokinase (GK)–glucokinase regulatory protein (GKRP) protein–protein interaction. Bourbeau and co-workers at Amgen describe their efforts towards an asymmetric synthesis of this compound ( J. Org. Chem. 2014, 79, 3684). The discovery route to this compound involved seven steps (14% overall yield), had certain safety concerns and relied upon SFC separation of the API enantiomers. The new route requires five steps (26% overall yield) and delivers the API in excellent enantiomeric excess (99% ee). A key feature of the synthetic approach was an asymmetric synthesis of the 2-alkynylpiperazine core via a base-promoted isomerization. It was found that the strongly basic conditions employed for the “alkyne-walk” did not erode the previously established stereocenter. Also, safety concerns around a late-stage amination of a 2-chloropyridine intermediate in the discovery route were alleviated by starting with a Boc-protected diaminopyridine instead.
PATENT

INTERMEDIATE A: TERT-EUTYL (5-(CHLOROSULFONYL)-2-PYRIDINYL)CARBAMATE

0,N

STEP 1 : TERT-BUTY (5-NITRO-2-PYRIDINYL)CARBAMATE

A 3-L round-bottomed flask was charged with 5-nitro-2-pyridinamine (75.0 g, 539 mmol, Alfa Aesar, Ward Hill, MA) and 500 mL of DCM. To this was added triethylamine (82 g, 810 mmol), di-tert-butyl dicarbonate (129 g, 593 mmol, Sigma-Aldrich, St. Louis, MO), and N,N-dimethylpyridin-4-amine (32.9 g, 270 mmol, Sigma-Aldrich, St. Louis, MO). After stirring at rt for 18 h, the mixture was diluted with water and the solid was collected by filtration. The yellow solid was washed with MeOH to give tert-butyl (5-nitro-2-pyridinyl)carbamate (94.6 g) as a light yellow solid.

STEP 2: TERT-BUTY (5 – AMINO-2-P YRIDINYL)C ARB AM ATE

A 3-L round-bottomed flask was charged with tert-butyl (5-nitro-2-pyridinyl)carbamate (96.4 g, 403 mmol), 500 mL of MeOH, 500 mL of THF, and 100 mL of sat aq NH4Cl. Zinc (105 g, 1610 mmol, Strem Chemical Inc, Newburyport, MA) was slowly added (over 10 min) to this solution. The mixture was stirred at room temperature for 12 h, then filtered. The filtrate was concentrated and then diluted with EtOAc and washed with water. The organic extracts were dried over MgS04, filtered, and concentrated. The resulting solid was recrystallized from MeOH to give tert-butyl(5-amino-2-pyridinyl)carbamate (38.6 g) as a light-yellow solid.

STEP 3: TERT-BUTYL (5-(CHLOROSULFONYL)-2-PYRIDINYL)CARBAMATE

A 3-L round-bottomed flask was charged with sodium nitrite (15.3 g, 221 mmol, J. T. Baker, Philipsburg, NJ), 100 mL of water and 500 mL of MeCN. After cooling to 0 °C, cone, hydrochloric acid (231 mL, 2770 mmol) was slowly added keeping the internal temperature below 10 °C. After stirring at 0 °C for 10 min, tert-butyl (5-amino-2-pyridinyl)carbamate (38.6 g, 184 mmol) was added as a suspension in MeCN (200 mL). The mixture was stirred for 30 min, then 150 mL of AcOH, copper(ii) chloride (12.4 g, 92.2 mmol, Sigma-Aldrich, St. Louis, MO), and copper(i) chloride (0.183 g, 1.85 mmol, Strem Chemical Inc,

Newburyport, MA) were added. S02 gas (Sigma-Aldrich, St. Louis, MO) was bubbled through the solution for 15 min. The mixture was stirred at 0 °C for 30 min, then about 500 mL of ice-cold water was added. The resulting precipitate was collected by filtration and dried over MgS04 to give tert-butyl (5-(chlorosulfonyl)-2-pyridinyl)carbamate (15.5 g) as a white solid.

1H NMR (400MHz, CDC13) δ ppm 8.93 (br s, 1 H), 8.63 – 8.42 (m, 1 H), 8.35 -7.94 (m, 2 H), 1.58 (s, 9 H).

INTERMEDIATE B: (3S)-l-BENZYL-3-(l-PROPYN-l-YL)PIPERAZINE

STEP 1 : (3S)-l-BENZYL-3-(2-PROPYN-l-YL)-2,5-PIPERAZINEDIONE

A 1-L round-bottoemd flask was charged with (S)-2-((tert-butoxycarbonyl)amino)pent-4-ynoic acid (42.0 g, 197 mmol, AK Scientific, Union City, CA), ethyl 2-(benzylamino)acetate (40.0 g, 207 mmol, Sigma-Aldrich, St. Louis, MO), HATU (90 g, 240 mmol, Oakwood Products, West Columbia, SC) and 200 mL of DMF. To this was added N-ethyl-N-isopropylpropan-2-amine (51.5 ml, 296 mmol, Sigma-Aldrich, St. Louis, MO). After 15 min of stirring at rt, the mixture was diluted with water 300 mL and extracted with 1 L of 20% EtOAc in diethyl ether. The layers were separated and the organic was washed with 2 M HCl, water, sat. aq. NaHC03 and brine. The extracts were dried and concentrated to give an off-white solid. To this was added 200 mL of DCM and TFA (152 ml, 1970 mmol, Sigma-Aldrich, St. Louis, MO). After stirring at rt for 30 min, the mixture was concentrated and then azetroped with 100 mL toluene (twice). To the brown oil obtained was added ammonia (2 M in MeOH, 394 ml, 789 mmol, Sigma-Aldrich, St. Louis, MO). The mixture was stirred at rt for 30 min. The mixture was concentrated, dissolved in EtOAc, and washed with water. The organics were dried (MgS04), filtered, and concentrated to give a white solid that was triturated with diethyl ether to give (S)-l-benzyl-3-(prop-2-yn-l-yl)piperazine-2,5-dione (37.3 g) as a white solid.

STEP 2: (3S)-l-BENZYL-3-(2-PROPYN-l-YL)PIPERAZINE

A 1-L round-bottomed flask was charged with (S)-l-benzyl-3-(prop-2-yn-l-yl)piperazine-2,5-dione (37.3 g, 154 mmol) and 150 mL of THF. To this was slowly added aluminum (III) lithium hydride (1M in THF, 539 ml, 539 mmol, Sigma-Aldrich, St. Louis, MO). After the addition was complete the mixture was heated at 80 °C for 12 h. The mixture was then cooled to 0 °C and solid sodium sulfate decahydrate was added until bubbling ceased. The mixture was filtered and the filtrate was concentrated to give (S)-l-benzyl-3-(prop-2-yn-l-yl)piperazine (18.1 g) as a yellow oil.

STEP 3: (35)-l-BENZYL-3-(l-PROPYN-l-YL)PIPERAZINE

To a solution of (35)-l-benzyl-3-(2-propyn-l-yl)piperazine (2.3 g, 11 mmol) in THF (50 mL) was added potassium t-butoxide (2.41 g, 21.5 mmol, Sigma-Aldrich, St. Louis, MO). The reaction mixture was stirred at rt for 30 min, then quenched with water (200 mL) and EtOAc (300 mL) was added. The organic phase was dried over sodium sulfate, filtered and concentrated under a vacuum to give a solid that was purified by silica gel column chromatography (0 to 10% MeOH in CH2CI2) and then recrystallized from hexanes to afford (35)- 1-benzyl-3-(l-propyn-l-yl)piperazine (2.16 g) as an off-white solid.

1H NMR (400MHz, CD3OD) δ ppm 7.42 – 7.21 (m, 5 H), 3.59 – 3.49 (m, 3 H), 2.93 (td, J= 2.9, 12.4 Hz, 1 H), 2.86 – 2.73 (m, 2 H), 2.68 (d, J= 11.3 Hz, 1 H), 2.22 – 2.04 (m, 2 H), 1.80 (d, J= 2.3 Hz, 3 H).

INTERMEDIATE C: N,N-BIS(4-METHOXYBENZYL)-5-(((35)-3-(l-PROPYN- 1 – YL)- 1 -PIPERAZINYL)SULFONYL)-2-PYRIDIN AMINE

STEP 1 : (35)-l-((6-CHLORO-3-PYRIDINYL)SULFONYL)-3-(l-PROPYN-l-YL)PIPERAZINE

To a stirred solution of benzyl (35)-3-(l-propyn-l-yl)-l-piperazinecarboxylate (2.51 g, 9.71 mmol, Intermediate E) in TFA (20 mL) in 250-mL round-bottomed flask, trifluoromethanesulfonic acid (2.59 mL, 29.1 mmol, Alfa Aesar, Ward Hill, MA) was added slowly at rt. After stirring at room temperature for 3 min, the reaction mixture was concentrated to dryness under a vacuum. DCM (20 mL) was added to the residue followed by triethylamine (13.5 mL, 97 mmol). After the material went into solution, the mixture was cooled to 0 °C and 6-chloro-3-pyridinesulfonyl chloride (2.06 g, 9.73 mmol, Organic Process Research & Development 2009, 13, 875) was added portion-wise. After 5 min of stirring at 0 °C, water (40 mL) was added at that temperature and the layers were separated. The aqueous phase was extracted with DCM (2 x 50 mL). The combined organic phases were washed with saturated aqueous sodium chloride (60 mL). The organic phase was dried over sodium sulfate, filtered and concentrated under a vacuum. The crude product was purified by column chromatography (100 g of silica, 30 to 90% EtOAc in hexanes) to afford (35)- 1-((6-chloro-3-pyridinyl)sulfonyl)-3-(l-propyn-l-yl)piperazine (2.61 g) as an off-white solid.

STEP 2: N,N-BIS(4-METHOXYBENZYL)-5-(((35)-3-(l-PROPYN-l-YL)-l-PIPERAZINYL)SULFONYL)-2-PYRIDIN AMINE

A mixture of (35)-l-((6-chloro-3-pyridinyl)sulfonyl)-3-(l-propyn-l-yl)piperazine (2.6 g, 8.7 mmol), N-(4-methoxybenzyl)-l-(4-methoxyphenyl)methanamine (2.40 g, 9.33 mmol, WO2007/109810A2), and DIPEA (2.4 mL, 14 mmol) in z-BuOH (8.0 mL) was heated at 132 °C using a microwave reactor for 3 h. This reaction was run three times (total starting material amount was 7.2 g). The mixtures from the three runs were combined and partitioned between EtOAc (200 mL) and aqueous NaHC03 (half saturated, 50 mL). The organic layer was washed with aqueous NaHC03 (3 x 50 mL), dried over Na2S04, filtered, and concentrated. The residue was purified (5-times total) by chromatography on silica using MeOH:DCM:EtOAc:hexane

(4:20:20:60) as eluent to give N,N-bis(4-methoxybenzyl)-5-(((3S)-3-(l-propyn-i-yl)-l-piperazinyl)sulfonyl)-2-pyridinamine (6.6 g) as a white foam.

1H NMR (400MHz ,CDC13) δ ppm 8.55 (d, J= 2.3 Hz, 1 H), 7.64 (dd, J= 2.5, 9.0 Hz, 1 H), 7.13 (d, J= 8.6 Hz, 4 H), 6.91 – 6.81 (m, 4 H), 6.47 (d, J= 9.0 Hz, 1 H), 4.75 (s, 4 H), 3.80 (s, 6 H), 3.68 – 3.61 (m, 1 H), 3.57 (d, J= 11.2 Hz, 1 H), 3.41 (d, J= 11.3 Hz, 1 H), 3.07 (td, J= 3.3, 12.1 Hz, 1 H), 2.87 (ddd, J= 2.9, 9.7, 12.2 Hz, 1 H), 2.63 – 2.47 (m, 2 H), 1.80 (d, J= 2.2 Hz, 3 H). One exchangeable proton was not observed, m/z (ESI, +ve ion) 521.2 (M+H)+.

INTERMEDIATE D: rEi?r-BUTYL(5-(((35)-3-(l-PROPYN-l-YL)-4-(4-(2-(TRIFLUOROMETHYL)-2-OXIRANYL)PHENYL)- 1 -PIPERAZINYL)SULFONYL)-2-PYRIDINYL)CARBAMATE

step 1 step 2

STEP 1 : l-BR0M0-4-(l-(TRIFLU0R0METHYL)ETHENYL)BENZENE

To a 1-L round-bottomed flask was added methyl phenylphosphonium bromide (25.4 g, 71.1 mmol, Sigma- Aldrich, St. Louis, MO) and toluene (75 mL). The resulting mixture was stirred for 5 min then concentrated and dried under high vacuum for 30 min. To this residue was added THF (300 mL) followed by n-butyllithium (2.5 M in hexanes, 29.0 mL, 71.1 mmol, Aldrich, St. Louis, MO) dropwise via an addition funnel. After being stirred for 1 h at rt, a solution of l-(4-bromophenyl)-2,2,2-trifluoroethanone (15.0 g, 59.3 mmol, Matrix Scientific, Columbia, SC) in THF (20 mL) was added to the reaction mixture dropwise via an addition funnel. The reaction mixture was stirred at rt for 2 h. The reaction was quenched with saturated aqueous NH4C1 and the mixture was concentrated. The residue was partitioned between diethyl ether (150 mL) and saturated aqueous NH4C1 (80 mL). The organic layer was washed with water and brine, dried over MgS04, filtered, and concentrated. The resulting crude product was purified by column chromatography (330 g of silica gel, 2 to 5% EtOAc in hexanes) to afford l-bromo-4-(l-(trifluoromethyl)ethenyl)benzene (14.0 g) as a brown liquid.

STEP 2: 2-(4-BROMOPHENYL)-3,3,3-TRIFLUORO-l,2-PROPANEDIOL

To a solution of l-bromo-4-(l-(trifluoromethyl)ethenyl)benzene (13.5 g, 53.8 mmol) in acetone (100 mL) and water (100 mL) was added NMO (6.90 g, 59.2 mmol, Sigma- Aldrich, St. Louis, MO) and osmium tetroxide (0.140 mL, 2.70 mmol, Sigma-Aldrich, St. Louis, MO). The resulting mixture was stirred at rt for 6 h. The reaction mixture was filtered and the filtrate was concentrated. The residue was partitioned between EtOAc (100 mL) and water (30 mL). The aqueous layer was extracted with EtOAc (2 x 75 mL). The combined organic layers were dried over MgS04, filtered, and concentrated. The resulting product was purified by column chromatography (330 g of silica gel, 0 to 8% MeOH in DCM) to afford 2-(4-bromophenyl)-3,3,3-trifluoro-l,2-propanediol (14.5 g) as an off-white solid.

STEP 3: 4-(4-BROMOPHENYL)-2,2-DIMETHYL-4-(TRIFLUOROMETHYL)-1,3-DIOXOLANE

To a solution of 2-(4-bromophenyl)-3,3,3-trifluoro-l,2-propanediol (14.5 g, 51.0 mmol) in acetone (200 mL) was added 2,2-dimethoxypropane (19.0 mL, 153 mmol, Sigma-Aldrich, St. Louis, MO) and /?-toluenesulfonic acid (0.485 g, 2.54 mmol, Sigma-Aldrich, St. Louis, MO). The resulting mixture was stirred at rt for 20 h. Additional 2,2-dimethoxypropane (19.0 mL, 153 mmol, Sigma-Aldrich, St. Louis, MO) and /?-toluenesulfonic acid (0.485 g, 2.54 mmol, Sigma-Aldrich, St. Louis, MO) were added and the reaction was stirred for another 20 h. The reaction was quenched with saturated aqueous NaHC03 (10 mL). The reaction mixture was concentrated and the residue was partitioned between

EtOAc (100 mL) and saturated aqueous NaHC03 (60 mL). The aqueous layer was extracted with EtOAc (2 x 50 mL). The combined organic layers were dried over MgS04, filtered, and concentrated. The resulting product was purified by column chromatography (330 g of silica gel, 0 to 8% EtOAc in hexanes) to afford 4-(4-bromophenyl)-2,2-dimethyl-4-(trifluoromethyl)-l,3-dioxolane (15.7 g) as a colorless liquid.

STEP 4: BENZYL (3S)-4-(4-(2,2-DIMETHYL-4-(TRIFLUOROMETHYL)-l,3-DIOXOLAN-4-YL)PHENYL)-3-(l -PROPYN- 1 -YL)- 1 -PIPERAZINECAPvBOXYLATE

To a 20-mL vial was added benzyl (3S)-3-(l -propyn- l-yl)-l-piperazinecarboxylate (1.0 g, 3.87 mmol, Intermediate E), RuPhos Palladacycle (0.250 g, 0.310 mmol, Strem Chemical, Newburyport, MA), 4-(4-bromophenyl)-2,2-dimethyl-4-(trifluoromethyl)-l,3-dioxolane (2.50 g, 7.74 mmol), dioxane (15.0 mL), and sodium t-butoxide (0.740 g, 7.74 mmol, Sigma-Aldrich, St.

Louis, MO). The reaction mixture was degassed by bubbling N2 through the solution for 5 min, then the vial was capped. The reaction mixture was heated at 80 °C for 30 min then allowed to cool to rt and partitioned between EtOAc (70 mL) and water (40 mL). The aqueous layer was extracted with EtOAc (1 x 50 mL). The combined organic layers were dried over MgS04, filtered, and concentrated. The crude product was purified by column chromatography (80 g of silica, 5% to 30% EtOAc in hexanes) to afford benzyl (35)-4-(4-(2,2-dimethyl-4-(trifluoromethyl)- 1 ,3-dioxolan-4-yl)phenyl)-3-(l -propyn- 1 -yl)- 1 -piperazinecarboxylate (1.6 g) as a yellow foam.

STEP 5: rEi?r-BUTYL(5-(((35)-3-(l-PROPYN-l-YL)-4-(4-(2,2,2-TRIFLUORO- 1 -HYDROXY- 1 -(HYDROXYMETH YL)ETHYL)PHENYL)- 1 -PIPERAZINYL)SULFONYL)-2-PYRIDINYL)CARBAMATE

To a 150-mL round-bottomed flask was added benzyl (3S)-4-(4-(2,2-dimethyl-4-(trifluoromethyl)- 1 ,3 -dioxolan-4-yl)phenyl)-3 -( 1 -propyn- 1 -yl)- 1 -piperazinecarboxylate (1.60 g, 3.18 mmol) and TFA (20 mL, Sigma-Aldrich, St. Louis, MO). After the substrate was completely dissolved in TFA,

trifluoromethanesulfonic acid (0.850 mL, 9.55 mmol, Alfa Aesar, Ward Hill,

MA) was added and the resulting mixture was stirred at rt for 1.5 h. The reaction mixture was slowly poured into a 300-mL beaker which contained 100 mL ice water. The resulting mixture was stirred while NaOH pellets (11.0 g) were slowly added to adjust the pH to 7. The solution was extracted with EtOAc (2 x 70 mL) and 10% IPA in CHCI3 (2 x 40 mL). The combined organic layers were dried over MgS04, filtered, and concentrated. The resulting intermediate was redissolved in DCM (60 mL). Triethylamine (2.20 mL, 16.0 mmol, Sigma-Aldrich, St. Louis, MO) and tert-butyl (5-(chlorosulfonyl)-2-pyridinyl)carbamate (1.04 g, 3.60 mmol, Intermediate A) were added. The reaction mixture was stirred at rt for 1 h then partitioned between DCM (70 mL) and water (30 mL). The aqueous layer was extracted with DCM (2 x 40 mL). The combined organic layers were dried over MgS04, filtered, and concentrated. The crude product was purified by column chromatography (120 g of silica, 10% to 40% acetone in hexanes) to afford tert-butyl (5-(((35)-3-(l-propyn-l-yl)-4-(4-(2,2,2-trifiuoro-l-hydroxy- 1 -(hydroxymethyl)ethyl)phenyl)- 1 -piperazinyl)sulfonyl)-2-pyridinyl)carbamate (1.0 g) as a yellow foam.

STEP 6: rEi?r-BUTYL(5-(((35)-3-(l-PROPYN-l-YL)-4-(4-(2-(TRIFLUOROMETHYL)-2-OXIRANYL)PHENYL)- 1 -PIPERAZINYL)SULFONYL)-2-PYRIDINYL)CARBAMATE

To a solution of tert-butyl (5-(((35)-3-(l-propyn-l-yl)-4-(4-(2,2,2-trifiuoro- 1 -hydroxy- 1 -(hydroxymethyl)ethyl)phenyl)- 1 -piperazinyl)sulfonyl)-2-pyridinyl)carbamate (0.300 g, 0.513 mmol) in DCM (5 mL) was added triethylamine (0.400 mL, 2.88 mmol, Sigma-Aldrich, St. Louis, MO) and p-toluenesulfonyl chloride (0.108 g, 0.564 mmol, Sigma-Aldrich, St. Louis, MO). The resulting mixture was heated at reflux (50 °C) under N2 for 2 h. The reaction mixture was cooled to rt and partitioned between sat. NaHCOs (30 mL) and DCM (70 mL). The aqueous layer was extracted with DCM (2 x 40 mL). The combined organic layers were dried over MgS04, filtered, and concentrated. The crude product was purified by column chromatography (40 g of silica, 10 to 40%> acetone in hexanes) to afford tert-butyl (5-(((35)-3-(l-propyn-l-yl)-4-(4-(2-(trifluoromethyl)-2-oxiranyl)phenyl)- 1 -piperazinyl)sulfonyl)-2-pyridinyl)carbamate (0.240 g) as an off-white solid.

1H NMR (400MHz, CDC13) δ ppm 8.66 (dd, J= 0.6, 2.3 Hz, 1 H), 8.20 – 8.10 (m, 1 H), 8.04 (dd, J= 2.2, 8.9 Hz, 1 H), 7.63 (s, 1 H), 7.41 (d, J= 8.6 Hz, 2 H), 6.94 (d, J= 8.8 Hz, 2 H), 4.42 (d, J= 2.2 Hz, 1 H), 3.89 – 3.67 (m, 2 H), 3.38 (d, J = 5.3 Hz, 3 H), 2.97 – 2.83 (m, 2 H), 2.80 – 2.60 (m, 1 H), 1.78 (dd, J= 0.8, 2.0 Hz, 3 H), 1.55 (s, 9 H). m/z (ESI, +ve ion) 567.2 (M+H)+.

ALTERNATIVE ROUTE TO 2-(4-BROMOPHENYL)-3,3,3-TRIFLUORO-l,2-PROPANEDIOL (INTERMEDIATE D STEP 2):

F3

step 1

STEP 1 : 2-(4-BROMOPHENYL)-2-(TRIFLUOROMETHYL)OXIRANE

To a flame-dried, 50-mL, round-bottomed flask was added potassium t-butoxide (0.450 g, 4.01 mmol, Sigma- Aldrich, St. Louis, MO), DMSO (5.0 mL) and trimethylsulfoxonium iodide (1.00 g, 4.54 mmol, Sigma- Aldrich, St. Louis, MO). The resulting mixture was stirred at rt for 40 min. To this reaction mixture was added l-(4-bromophenyl)-2,2,2-trifluoroethanone (1.0 g, 4.0 mmol, Matrix Scientific, Columbia, SC) in DMSO (5.0 mL) dropwise via an addition funnel. The reaction mixture was stirred at rt for 30 min then quenched with water (1 mL) and partitioned between EtOAc (70 mL) and water (30 mL). The organic layer was washed with water (4 x 30 mL), dried over MgS04, filtered, and concentrated. The crude product was purified by column chromatography (40 g of silica, 10 to 20% acetone in hexanes) to afford 2-(4-bromophenyl)-2-(trifluoromethyl)oxirane (0.610 g) as a pale-yellow liquid.

STEP 2: 2-(4-BROMOPHENYL)-3,3,3-TRIFLUORO-l,2-PROPANEDIOL

To a 20-mL vial was added 2-(4-bromophenyl)-2-(trifluoromethyl)oxirane (0.200 g, 0.750 mmol), dioxane (2.0 mL), and water (3.0 mL). The resulting mixture was heated at 85 °C for 24 h. The reaction mixture was cooled to rt and extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over MgS04, filtered and concentrated. The crude product was purified by column chromatography (40 g of silica, 10 to 30% acetone in hexanes) to afford 2-(4-bromophenyl)-3,3,3-trifluoro-l,2-propanediol (2.0 g) as a white solid.

INTERMEDIATE E: BENZYL (3S)-3-(l-PROPYN-l-YL)-l-PIPERAZINECARBOXYLATE

-Cbz

STEP 1 : 4-BENZYL 1 – TER Γ-BUT YL 2-0X0-1,4-PIPERAZINEDICARBOXYLATE

A 2-L Erlenmeyer flask was charged with 2-piperazinone (36.5 g, 364 mmol, Sigma-Aldrich, St. Louis, MO), sodium carbonate (116 g, 1090 mmol, J. T. Baker, Philipsburg, NJ), 600 mL of dioxane, and 150 mL of water. To this was slowly added benzyl chloroformate (62.1 g, 364 mmol, Sigma-Aldrich, St. Louis, MO) at rt over 20 min. After the addition was complete, the mixture was stirred for 2 h and then diluted with water and extracted with EtOAc (2 L). The combined organic extracts were dried (MgS04), filtered, and concentrated to give a white solid. To this solid was added 500 mL of DCM, triethylamine (128 mL, 911 mmol, Sigma-Aldrich, St. Louis, MO), DMAP (4.45 g, 36.4 mmol, Sigma-Aldrich, St. Louis, MO), and di-tert-butyl dicarbonate (119 g, 546 mmol, Sigma-Aldrich, St. Louis, MO). After stirring at room temperature for 1 h, the mixture was diluted with water and the organics were separated. The organics were dried (MgS04), filtered, and concentrated to give a brown oil. To this oil was added 100 mL of DCM followed by 1 L of hexane. The resulting white solid was collected by filtration to give 4-benzyl 1-tert-butyl 2-oxo-l,4-piperazinedicarboxylate (101 g).

STEP 2: BENZYL (2-((7¾’i?J,-BUTOXYCARBONYL)AMINO)ETHYL)(2-OXO-3 -PENT YN- 1 – YL)C ARB AMATE

A 150-mL round-bottomed flask was charged with 4-benzyl 1-tert-butyl 2-oxo- 1 ,4-piperazinedicarboxylate (1.41 g, 4.22 mmol) and THF (5 mL). 1-Propynylmagnesium bromide (0.5 M in THF, 20.0 mL, 10.0 mmol, Sigma-Aldrich, St. Louis, MO) was added at 0 °C slowly. The mixture was stirred at 0 °C for 2 h. Saturated aqueous NH4C1 (40 mL) was added and the aqueous phase was extracted with EtOAc (200 mL, then 2 x 100 mL). The combined organic phases were dried over sodium sulfate, filtered and concentrated under a vacuum. The crude product was purified by column chromatography (50 g of silica, 0 to 50% EtOAc in hexanes) to afford benzyl (2- tert-butoxycarbonyl)amino)ethyl)(2-oxo-3-pentyn-l-yl)carbamate (1.55 g) as a clear oil.

STEP 3: BENZYL 3-(l-PROPYN-l-YL)-l-PIPERAZINECARBOXYLATE

A 3-L round-bottomed flask was charged with 2-((tert-butoxycarbonyl)amino)ethyl)(2-oxo-3-pentyn-l-yl)carbamate (82.17 g, 219 mmol) and 300 mL of DCM. After cooling to -10 °C, TFA (169 mL, 2200

mmol) was added and the resulting dark solution was stirred at rt for 15 min.

Sodium triacetoxyborohydride (186 g, 878 mmol, Sigma- Aldrich, St. Louis, MO) was then added portion- wise over 10 min. After 2 h, the mixture was

concentrated, diluted with EtOAc (1 L), and neutralized with 5 N NaOH. The layers were separated and the organic extracts were washed with brine, dried (MgS04), filtered and concentrated. The resulting orange oil was purified via column chromatography (750 g of silica gel, 0 to 4.5 % MeOH/DCM) to give benzyl 3 -(l-propyn-l-yl)-l -piperazmecarboxylate (43.67 g) as a brown foam.

STEP 4: 4-BENZYL 1 – TER Γ-BUT YL 2-(l -PROP YN-l-YL)- 1,4-PIPERAZINEDICARBOXYLATE

A 20-mL vial was charged with benzyl 3-(l-propyn-l-yl)-l-piperazinecarboxylate (0.616 g, 2.38 mmol), di-tert-butyl dicarbonate (0.979 g, 4.49 mmol, Sigma-Aldrich, St. Louis, MO), DMAP (0.0287 g, 0.235 mmol, Sigma-Aldrich, St. Louis, MO), TEA (0.90 mL, 6.5 mmol) and DCM (8 mL). The mixture was stirred at rt for 30 min. The reaction mixture was partitioned between water (20 mL) and EtOAc (20 mL). The aqueous phase was extracted with EtOAc (20 mL). The organic phase was washed with saturated aqueous sodium chloride (40 mL), dried over sodium sulfate, filtered, and concentrated under a vacuum. The crude product was purified by column chromatography (25 g of silica, 0 to 50% EtOAc in hexanes) to afford 4-benzyl 1-tert-butyl 2-(l-propyn-l-yl)-l,4-piperazinedicarboxylate (0.488 g) as a colorless oil.

STEP 5: 4-BENZYL 1 – TER Γ-BUT YL (2S)-2-( 1 -PROP YN-l-YL)- 1,4-PIPERAZINEDICARBOXYLATE

The individual enantiomers of 4-benzyl 1-tert-butyl 2-(l-propyn-l-yl)-1 ,4-piperazinedicarboxylate were isolated using chiral SFC. The method used was as follows: Chiralpak® ADH column (Daicel Inc., Fort Lee, NJ) (30 x 250 mm, 5 μιη) using 12% ethanol in supercritical C02 (total flow was 170 mL/min).

This separated the two enantiomers with enantiomeric excesses greater than 98%. The first eluting peak was subsequently identified as 4-benzyl 1-tert-butyl (2S)-2-(l-propyn-l-yl)-l,4-piperazinedicarboxylate and used in the next step.

STEP 6: BENZYL (3S)-3-(l-PROPY -l-YL)-l-PIPERAZINECAPvBOXYLATE

A 100-mL round-bottomed flask was charged with 4-benzyl 1-tert-butyl (25)-2-(l-propyn-l-yl)-l,4-piperazinedicarboxylate (0.145 g, 0.405 mmol), TFA (1.0 mL, 13 mmol) and DCM (2 mL). The mixture was stirred at rt for 40 min. The mixture was concentrated and solid NaHC03 was added followed by saturated aqueous NaHC03. The aqueous phase was extracted with EtOAc (2 x 20 mL). The combined organic phases were washed with IN NaOH (40 mL), saturated aqueous NaHC03 (40 mL), water (40 mL) and saturated aqueous sodium chloride (40 mL). The organic phase was dried over sodium sulfate, filtered, and concentrated under a vacuum to afford benzyl (35)-3-(l-propyn-l-yl)-l-piperazinecarboxylate (0.100 g) as a pale yellow clear oil which solidified upon standing to give a pale yellow solid.

1H NMR (400MHz, MeOD) δ ppm 7.47 – 7.13 (m, 5 H), 5.27 – 5.00 (m, 2 H), 3.88 – 3.58 (m, 3 H), 3.48 – 3.33 (m, 2 H), 3.22 – 3.02 (m, 1 H), 2.89 – 2.63 (m, 1 H), 1.80 (s, 3 H). m/z (ESI, +ve ion) 259.1 (M+H)+.

XAMPLE 23: 5-(((3S)-3-(l-PROPYN-l-YL)-4-(4-(l,2,2,2-TETRAFLUORO-1 -(TRIFLUOROMETHYL)ETHYL)PHENYL)- 1 -PIPERAZINYL)SULFONYL)-2-PYRIDIN AMINE

STEP 1 : 2-(4-((2S)-4-BENZYL-2-(l-PROPYN-l-YL)-l-PIPERAZINYL)PHENYL)-1 , 1 ,1 ,3,3,3-HEXAFLUORO-2-PROPANOL

A 20-mL vial was charged with (3S)-l-benzyl-3-(l-propyn-l-yl)piperazine (2.143 g, 10 mmol, Intermediate B), 2-(4-bromophenyl)-1,1,1, 3,3, 3-hexafluoropropan-2-ol (3.09 g, 11.5 mmol, Bioorg. Med. Chem. Lett. 2002, 12, 3009), sodium 2-methylpropan-2-olate (1.92 g, 20.0 mmol, Sigma-Aldrich, St. Louis, MO), dioxane (5 mL), RuPhos palladacycle (0.364 g, 0.500 mmol, Strem Chemical Inc., Newburyport, MA), and RuPhos (0.233 g, 0.500 mmol, Strem Chemical Inc., Newburyport, MA). The vial was sealed and heated at 100 °C for 1 h. The mixture was allowed to cool to rt, and diluted with water and extracted with EtOAc. The combined organic phases were dried over sodium sulfate, filtered and concentrated under a vacuum to give a solid that was purified by silica gel column chromatography (0 to 40% EtOAc in hexanes) to afford 2-(4-((2S)-4-benzyl-2-( 1 -propyn- 1 -yl)- 1 -piperazinyl)phenyl)- 1,1,1,3,3,3-hexafluoro-2-propanol (1.75 g) as a slightly yellow oil.

STEP 2: l,l,l,3,3,3-HEXAFLUORO-2-(4-((2S)-2-(l-PROPYN-l-YL)-l-PIPERAZINYL)PHENYL)-2-PROPANOL

A 250 mL round-bottomed flask was charged with 2-(4-((2S)-4-benzyl-2-( 1 -propyn- 1 -yl)- 1 -piperazinyl)phenyl)- 1,1,1 ,3 ,3 ,3-hexafluoro-2-propanol (1.75 g, 4.35 mmol), potassium carbonate (2.40 g, 17.4 mmol, Sigma-Aldrich, St. Louis, MO), CH2CI2 (25 mL), and 1-chloroethyl chlorocarbonate (1.88 mL, 17.4 mmol, Sigma-Aldrich, St. Louis, MO). After 30 min at rt, the reaction was filtered and the filtrate was concentrated. To the resulting oil was added MeOH (25 mL). This mixture was heated at 75 °C for 1.5 h then concentrated. The residue was triturated with diethyl ether to give l,l,l,3,3,3-hexafluoro-2-(4-((2S)-2-(l-propyn-l-yl)-l-piperazinyl)phenyl)-2-propanol (1.44 g) as a white solid.

STEP 3: TERT-BUTYL (5-(((3S)-3-(l-PROPYN-l-YL)-4-(4-(2,2,2-TRIFLUORO- 1 -HYDROXY- 1 -(TRIFLUOROMETHYL)ETHYL)PHENYL)- 1 -PIPERAZINYL)SULFONYL)-2-PYRIDINYL)CARBAMATE

A 250-mL round-bottomed flask was charged with 1,1,1,3,3,3-hexaf uoro-2-(4-((2S)-2-( 1 -propyn- 1 -yl)- 1 -piperazinyl)phenyl)-2-propanol (18.9 g, 51.6 mmol) and DCM (150 mL) and cooled to 0 °C. TEA was added (14.4 mL, 103 mmol, Sigma-Aldrich, St. Louis, MO) followed by tert-butyl (5- (chlorosulfonyl)pyridin-2-yl)carbamate (15.9 g, 54.2 mmol, Intermediate A) portionwise. After 10 min, the reaction mixture was diluted with water (100 mL) and the organic layer was separated, dried over Na2S04, filtered and concentrated under a vacuum to give a solid that was purified by silica gel column

chromatography (0 to 50% EtO Ac in hexanes) to afford tert-butyl (5 -(((3 S)-3 -( 1 -propyn- 1 -yl)-4-(4-(2,2,2-trifluoro- 1 -hydroxy- 1 -(trifluoromethyl)ethyl)phenyl)- 1 -piperazinyl)sulfonyl)-2-pyridinyl)carbamate (19.9 g) as a tan foam.

STEP 4: 5-(((3S)-3-(l-PROPYN-l-YL)-4-(4-(l,2,2,2-TETRAFLUORO-l- (TRIFLUOROMETHYL)ETHYL)PHENYL)- 1 -PIPERAZINYL)SULFONYL)-2-PYRIDIN AMINE

A 500-mL round-bottomed flask was charged with tert-butyl (5-(((3S)-3-(1 -propyn- 1 -yl)-4-(4-(2,2,2-trifluoro- 1 -hydroxy- 1 – (trifluoromethyl)ethyl)phenyl)-l-piperazinyl)sulfonyl)-2-pyridinyl)carbamate (19.7 g, 31.6 mmol) and DCM (300 mL) and cooled to 0 °C.

(Diethylamino)sulfur trifluoride (4.18 mL, 31.6 mmol, Matrix Scientific, Columbia, SC) was added, and after 10 min, the reaction was diluted with water (250 mL) and DCM (200 mL). The organic layer was separated, dried over

Na2S04, filtered and concentrated under a vacuum. The resultant foam was taken up in DCM (200 mL) and cooled to 0 °C. TFA (100 mL, 1298 mmol) was added and the reaction mixture was warmed to rt for 1.5 h. The reaction was then re-cooled to 0 °C and solid sodium bicarbonate was added slowly until gas evolution ceased. The mixture was diluted with water (250 mL) and DCM (300 mL) and the organic layer was separated, dried over Na2S04, filtered and concentrated under a vacuum to give a solid that was purified by silica gel column chromatography (0 to 100% EtOAc in hexanes) to afford 5-(((3S)-3-(l-propyn- 1 -yl)-4-(4-( 1 ,2,2,2-tetrafluoro- 1 -(trifluoromethyl)ethyl)phenyl)- 1 -piperazinyl)sulfonyl)-2-pyridinamine (11.05 g) as a single enantiomer.

1H NMR (400MHz, CD3OD) δ ppm 8.31 (d, J= 2.2 Hz, 1 H), 7.74 (dd, J= 2.4, 8.9 Hz, 1 H), 7.47 (d, J = 8.8 Hz, 2 H), 7.12 (d, J = 9.0 Hz, 2 H), 6.63 (d, J= 8.8 Hz, 1 H), 4.76-4.70 (m, 1 H), 3.76 (dd, J= 1.9, 11.2 Hz, 2 H), 3.66 – 3.52 (m, 1 H), 3.29 – 3.20 (m, 1 H), 2.79 – 2.72 (m, 1 H), 2.66 – 2.53 (m, 1 H), 1.76 (d, J = 2.2 Hz, 3 H). m/z (ESI, +ve ion) 525.2 (M+H)+. GK-GKRP IC50 (Binding) = 0.187 μΜ.

PAPER

Small Molecule Disruptors of the Glucokinase–Glucokinase Regulatory Protein Interaction: 2. Leveraging Structure-Based Drug Design to Identify Analogues with Improved Pharmacokinetic Profiles

Department of Therapeutic Discovery—Medicinal Chemistry, Department of Therapeutic Discovery—Molecular Structure and Characterization, §Department of Metabolic Disorders, Department of Pharmacokinetics and Drug Metabolism, Department of Pathology, #Department of Pharmaceutics Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California, 91320 and 360 Binney Street, Cambridge, Massachusetts, 02142, United States
J. Med. Chem., 2014, 57 (2), pp 325–338
DOI: 10.1021/jm4016747
Abstract Image

In the previous report, we described the discovery and optimization of novel small molecule disruptors of the GK-GKRP interaction culminating in the identification of 1 (AMG-1694). Although this analogue possessed excellent in vitro potency and was a useful tool compound in initial proof-of-concept experiments, high metabolic turnover limited its advancement. Guided by a combination of metabolite identification and structure-based design, we have successfully discovered a potent and metabolically stable GK-GKRP disruptor (27, AMG-3969). When administered to db/db mice, this compound demonstrated a robust pharmacodynamic response (GK translocation) as well as statistically significant dose-dependent reductions in fed blood glucose levels.

2-(4-((2S)-4-((6-Amino-3-pyridinyl)sulfonyl)-2-(1-propyn-1-yl)-1-piperazinyl)phenyl)-1,1,1,3,3,3-hexafluoro-2-propanol (27)

1H NMR (400 MHz, CDCl3) δ 8.48 (d, J = 2.3 Hz, 1 H), 7.77 (dd, J = 2.5, 8.8 Hz, 1 H), 7.57 (d, J = 8.8 Hz, 2 H), 6.95 (d, J = 9.2 Hz, 2 H), 6.52 (d, J = 8.8 Hz, 1 H), 4.94 (s, 2 H), 4.44 (br s, 1 H), 3.82–3.71 (m, 2 H), 3.58–3.33 (m, 3 H), 2.81 (dd, J = 3.2, 11.1 Hz, 1 H), 2.67 (dt, J = 3.9, 11.0 Hz, 1 H), 1.78 (d, J = 2.2 Hz, 3 H).
m/z (ESI, +ve ion) 523.2 (M + H)+.
REFERENCES
St Jean, D.J. Jr.; Ashton, K.; Andrews, K.; et al.
Small molecule disruptors of the glucokinase-glucokinase regulatory protein (GK-GKRP) interaction
34th Natl Med Chem Symp (May 18-21, Charleston) 2014, Abst 4
Small molecule disruptors of the GK-GKRP interaction as potential antidiabetics
247th Am Chem Soc (ACS) Natl Meet (March 16-20, Dallas) 2014, Abst MEDI 214
Use of non-traditional conformational restriction in the design of a novel, potent, and metabolically stable series of GK-GKRP inhibitors
248th Am Chem Soc (ACS) Natl Meet (August 10-14, San Francisco) 2014, Abst MEDI 267
Small molecule inhibitors for glucokinase-glucokinase regulatory protein (GK-GKRP) binding: Optimization for in vivo target assessment of type II diabetes
248th Am Chem Soc (ACS) Natl Meet (August 10-14, San Francisco) 2014, Abst MEDI 268

MAKING CONNECTIONS Aleksandra Baranczak (right), a fourth-year grad student in Gary A. Sulikowski’s lab at Vanderbilt University, discusses her efforts to synthesize the core of the diazo-containing natural product lomaiviticin A with Kate Ashton, a medicinal chemist at Amgen
Dr. Kate Ashton

Mark Norman

Mark Norman

Michael Bartberger

Michael Bartberger

Chris Fotsch

Chris Fotsch

David St. Jean

David St. Jean

Klaus Michelsen

Klaus Michelsen

///////////1361224-53-4, AMGEN, AMG 3969, Type 2 Diabetes,  PRECLINICAL
O=S(=O)(c1ccc(N)nc1)N2C[C@H](C#CC)N(CC2)c3ccc(cc3)C(O)(C(F)(F)F)C(F)(F)F

Filed under: Preclinical drugs, Uncategorized Tagged: 1361224-53-4, amg 3969, amgen, preclinical, TYPE 2 DIABETES

Astellas Pharma Inc. new Glucokinase Activator, ASP ? for Type 2 Diabetes

$
0
0

str1

ASP ?

(2R)-2-(4-cyclopropanesulfonyl-3-cyclopropylphenyl)-N-[5-(hydroxymethyl)pyrazin-2-yl]-3-[(R)-3-oxocyclopentyl]propanamide

CAS 1174229-89-0
MW C25 H29 N3 O5 S
Benzeneacetamide, 3-cyclopropyl-4-(cyclopropylsulfonyl)-N-[5-(hydroxymethyl)-2-pyrazinyl]-α-[[(1R)-3-oxocyclopentyl]methyl]-, (αR)-
Molecular Weight, 483.58
[α]D20 −128.7 (c 1.00, MeOH);
1H NMR (DMSO-d6, 400 MHz) δ 11.07 (s, 1H), 9.20 (d, J = 1.4 Hz, 1H), 8.41 (d, J = 1.4 Hz, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.41 (dd, J = 8.2, 1.8 Hz, 1H), 7.15 (d, J = 1.8 Hz, 1H), 5.52 (t, J = 5.7 Hz, 1H), 4.56 (d, J = 6.0 Hz, 2H), 4.04 (t, J = 7.6 Hz, 1H), 3.03–2.97 (m, 1H), 2.79 (tt, J = 8.4, 5.1 Hz, 1H), 2.25–1.81 (m, 8H), 1.53–1.47 (m, 1H), 1.17–1.12 (m, 2H), 1.08–1.02 (m, 4H), 0.89–0.84 (m, 2H);
13C NMR (DMSO-d6, 101 MHz) δ 218.5, 171.8, 152.1, 147.3, 145.7, 143.2, 140.3, 138.2, 134.8, 129.0, 125.3, 125.1, 62.5, 49.9, 44.4, 38.4, 38.2, 34.8, 32.1, 29.1, 12.4, 10.8, 10.7, 5.8;
FTIR (ATR, cm–1) 3544, 3257, 1727, 1692, 1546, 1507, 1363, 1285, 1149, 719;
HRMS (ESI) m/z [M + Na]+ calcd for C25H29N3O5S 506.1726, found 506.1747.
Anal. Calcd for C25H29N3O5S: C, 62.09; H, 6.04; N, 8.69. Found: C, 61.79; H, 6.19; N, 8.62.

To Astellas Pharma,Inc.

Inventors Masahiko Hayakawa, Yoshiyuki Kido, Takahiro Nigawara, Mitsuaki Okumura, Akira Kanai, Keisuke Maki, Nobuaki Amino
Applicant Astellas Pharma Inc.

Image result for Process Chemistry Labs., Astellas Pharma Inc., 160-2 Akahama, Takahagi-shi, Ibaraki 318-0001, Japan

Synthesis

contd…………………………..

PATENT

WO2009091014

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=56E9927692EF5105140FE1CD1FD14A5D.wapp1nC?docId=WO2009091014&recNum=114&maxRec=374&office=&prevFilter=&sortOption=&queryString=FP%3A%28astellas+pharma%29&tab=FullText

str1

PAPER

A Practical and Scalable Synthesis of a Glucokinase Activator via Diastereomeric Resolution and Palladium-Catalyzed C–N Coupling Reaction

Process Chemistry Labs., Astellas Pharma Inc., 160-2 Akahama, Takahagi-shi, Ibaraki 318-0001, Japan
Astellas Research Technologies Co., Ltd., 21 Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
§ Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoicho, Inageku, Chiba 263-8522, Japan
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00415
 Abstract Image

Here we describe the research and development of a process for the practical synthesis of glucokinase activator (R)-1 as a potential drug for treating type-2 diabetes. The key intermediate, chiral α-arylpropionic acid (R)-2, was synthesized in high diastereomeric excess through the diasteromeric resolution of 7 without the need for a chiral resolving agent. The counterpart 2-aminopyrazine derivative 3 was synthesized using a palladium-catalyzed C–N coupling reaction. This efficient process was demonstrated at the pilot scale and yielded 19.0 kg of (R)-1. Moreover, an epimerization process to obtain (R)-7 from the undesired (S)-7 was developed.

Hayakawa, M.; Kido, Y.; Nigawara, T.; Okumura, M.; Kanai, A.; Maki, K.; Amino, N. PCT Int. Appl. WO/2009/091014 A1 20090723,2009.

https://www.astellas.com/en/ir/library/pdf/3q2017_rd_en.pdf

///////////1174229-89-0, ASTELLAS, Glucokinase Activator, TYPE 2 DIABETES, PRECLINICAL, ASP ?, WO 2009091014Masahiko Hayakawa, Yoshiyuki Kido, Takahiro Nigawara, Mitsuaki Okumura, Akira Kanai, Keisuke Maki, Nobuaki AminoWO2009091014,

O=C(Nc1cnc(cn1)CO)[C@H](C[C@@H]2CC(=O)CC2)c3ccc(c(c3)C4CC4)S(=O)(=O)C5CC5


Filed under: DIABETES, Preclinical drugs Tagged: 1174229-89-0, Akira Kanai, ASP ?, astellas, Glucokinase Activator, Keisuke Maki, Masahiko Hayakawa, Mitsuaki Okumura, Nobuaki Amino, preclinical, Takahiro Nigawara, TYPE 2 DIABETES, WO 2009091014, WO2009091014, Yoshiyuki Kido
Viewing all 87 articles
Browse latest View live