Quantcast
Channel: New Drug Approvals
Viewing all 2925 articles
Browse latest View live

RO 5114436……..The chemokine receptor CCR5 is a clinically validated target for Human Immunodeficiency Virus (HIV) disease and a potentially interesting target for the inflammation therapy area

$
0
0

 

RO 5114436

1220514-67-9  CAS OF FREE BASE

1220514-58-8 0F HCL SALT

Hoffmann La Roche,

 

3-​Furancarboxamide, N-​[(1S)​-​3-​[(3aR,​6aS)​-​5-​[(4,​6-​dimethyl-​5-​pyrimidinyl)​carbonyl]​hexahydropyrrolo[3,​4-​c]​pyrrol-​2(1H)​-​yl]​-​1-​(3-​fluorophenyl)​propyl]​tetrahydro-​, (3R)​-

(R)-Tetrahydrofuran-3-carboxylic acid [(S)-3-[5-(4,6-dimethylpyrimidine-5-carbonyl)hexahydropyrrolo[3,4-c]pyrrol-2-yl]-1-(3-fluorophenyl)propyl]amide

The chemokine receptor CCR5 is a clinically validated target for Human Immunodeficiency Virus (HIV) disease and a potentially interesting target for the inflammation therapy area. The first small-molecule CCR5 antagonist on the market, maraviroc (Selzentry), was approved by the FDA for treatment of HIV-1 infection.(1) Medicinal chemistry research at Roche led to the discovery of a series of 3,7-diazabicyclo[3.3.0]octane compounds,(2) represented by RO5114436 (1), that are potent CCR5 antagonists. Compound 1 also showed high potency in functional assays for inflammation. The PK properties of 1 were superior to those of maraviroc in preclinical species, including rat, dog, and monkey.

 

octahydro-pyrrolo[3,4-c]pyrrole derivatives useful in the treatment of a variety of disorders, including those in which the modulation of CCR5 receptors is implicated. More particularly, the present invention relates to 3-(hexahydro- pyrrolo[3,4-c]pyτrol-2-yl)-l-phenyl-propylamine and [3-(hexahydro-pyrrolo[3,4- c]pyτrol-2-yl)-propyl]-phenyl-amine compounds and related derivatives, to compositions containing, to uses of such derivatives and to processes for preparing said compoundsz. Disorders that may be treated or prevented by the present derivatives include HIV and genetically related retroviral infections (and the resulting acquired immune deficiency syndrome, AIDS), diseases of the immune system and inflammatory diseases.

A-M. Vandamme et al. (Antiviral Chemistry & Chemotherapy, 1998 9:187-203) disclose current HAART clinical treatments of HIV- 1 infections in man including at least triple drug combinations. Highly active anti-retroviral therapy (HAART) has traditionally consisted of combination therapy with nucleoside reverse transcriptase inhibitors (NRTI), non-nucleoside reverse transcriptase inhibitors (NNRTI) and protease inhibitors (PI). These compounds inhibit biochemical processes required for viral replication. In compliant drug-naive patients, HAART is effective in reducing mortality and progression of HIV- 1 to AIDS. While HAART has dramatically altered the prognosis for HIV infected persons, there remain many drawbacks to the current therapy including highly complex dosing regimes and side effects which can be very severe (A. Carr and D. A. Cooper, Lancet 2000356(9239):1423-1430). Moreover, these multidrug therapies do not eliminate HIV-1 and long-term treatment usually results in multidrug resistance, thus limiting their utility in long term therapy. Development of new drug therapies to provide better HIV-1 treatment remains a priority. Compounds of the present invention modulate the activity of the chemokine CCR5 receptors. The chemokines are a large family of pro-inflammatory peptides that exert their pharmacological effect through G-protein-coupled receptors. The name “chemokine”, is a contraction of “chemotactic cytokines”. The chemokines are a family of leukocyte chemotactic proteins capable of attracting leukocytes to various tissues, which is an essential response to inflammation and infection. Human chemokines include approximately 50 small proteins of 50-120 amino acids that are structurally homologous. (M. Baggiolini etal, Annu. Rev. Immunol. 1997 15:675-705)

Modulators of the CCR5 receptor may be useful in the treatment of various inflammatory diseases and conditions, and in the treatment of infection by HIV-1 and genetically related retroviruses. As leukocyte chemotactic factors, chemokines play an indispensable role in the attraction of leukocytes to various tissues of the body, a process which is essential for both inflammation and the body’s response to infection. Because chemokines and their receptors are central to the pathophysiology of inflammatory and infectious diseases, agents which are active in modulating, preferably antagonizing, the activity of chemokines and their receptors, are useful in the therapeutic treatment of such inflammatory and infectious diseases. The chemokine receptor CCR5 is of particular importance in the context of treating inflammatory and infectious diseases. CCR5 is a receptor for chemokines, especially for the macrophage inflammatory proteins (MIP) designated MIP- la and MIP- lb, and for a protein which is regulated upon activation and is normal T-cell expressed and secreted (RANTES).

……………..

OPRD [PAPER
Figure

http://pubs.acs.org/doi/abs/10.1021/op100020z

Org. Process Res. Dev., 2010, 14 (3), pp 592–599
DOI: 10.1021/op100020z

A practical asymmetric synthesis of a 3,7-diazabicyclo[3.3.0]octane derivative (1), a representative of a new class of potent CCR5 receptor antagonists, is described. The benzylamine stereogenic center of 1 was introduced by a ruthenium-catalyzed asymmetric reductive amination using (R)-MeOBIPHEP as ligand. Aldehyde 4, prepared by Parikh−Doering oxidation, was used without workup in the reductive amination reaction, which not only simplified the process but also overcame the instability of 4. The 3,7-diazabicyclo[3.3.0]octane core was obtained by a [3 + 2] cycloaddition.

(R)-Tetrahydrofuran-3-carboxylic acid [(S)-3-[5-(4,6-dimethylpyrimidine-5-carbonyl)hexahydropyrrolo[3,4-c]pyrrol-2-yl]-1-(3-fluorophenyl)propyl]amide Hydrochloride Salt (1·HCl).
Oxalyl chloride (46.0 g, 36.2 mmol) was added over 1 h to a solution of (R)-tetrahydrofuran-3-carboxylic acid 3(16) (40.1 g, 34.5 mmol) in toluene (310 mL) containing DMF (0.5 mL) with stirring, while maintaining the temperature at 10 °C with an ice bath. …………DELETED…………………….The aqueous phase was extracted with Me-THF (20 mL), and the combined organic phase was evaporated at 40 °C to give the free base of 1 (11.44 g, 23.08 mmol) as a slightly tacky dry foam.
A solution of the free base of 1 (11.44 g, 23.08 mmol) in acetone (57.4 mL) and water (1.45 mL) was acidified with 12 N HCl (2.42 mL, 29.0 mmol). The clear solution was seeded with authentic product and stirred for 3.5 h, after which the resulting slurry was filtered and washed with ice-cold acetone (12 mL in two portions). Drying in a vacuum oven at 68 °C, 30−50 Torr gave 1·HCl as a dry, white powder (10.57 g, 86.1% theory, 97.8% purity by HPLC area).
Analytical data for 1·HCl salt:
mp 149−150 °C.
1H NMR (300 MHz, D2O) 1.73−1.91 (m, 1 H), 1.98−2.26 (m, 3 H), 2.34 (s, 3 H), 2.37 (s, 3 H), 2.82−3.37 (m, 8 H), 3.40−4.10 (m, 9 H), 4.81 (t, J = 7.54 Hz, 1 H), 6.93−7.13 (m, 3 H), 7.33 (td, J = 7.82, 5.84 Hz, 1 H), 8.83 (s, 1 H).
MS m/z 496.2 [M + H]+.
……………………..
WO/2005/121145
CLOSEST EXAMPLE

Example 14

(S)-4,4-Difluoro-cyclohexanecarboxylic acid [3- [5-(4,6-dimethyl-pyrimidine-5- carbonyl)-hexahydro-pyrrolo[3,4-c]pyrrol-2-yl]-l-(3-fluoro-phenyl)-propyl]-amide (I- 485)

 

56 step 2 I ^ 57a: R = Boc 57b: R = H

step 1 – To a solution of 56 (562 mg, 2.1 mmol, prepared as described in WO2004/018425) and 44 (518 mg, 2.1 mmol) in DCM (20mL) containing HOAc (0.31 mL) was added NaBH(OAc)3 (579 mg, 2.73 mmol) in 1 portion and the reaction mixture was stirred for 18 hrs at RT. The reaction was quenched by the addition of 10% K2CO3 (20 mL) and stirring continued for 30 min. The product was twice extracted with DCM (25 mL). The combined extracts were dried (MgSO ) and concentrated in vacuo. The crude product was purified by flash chromatography on silica eiuting with DCM/ 5% MeOH (containing 2% NH4OH) to afford 821 mg (79% theory) of 57a as a white foam: ms (ES+) m/z 498 (M+H)+. step 2 – A solution of 57a (821 mg, 1.65 mmol) dissolved in 10 M HCl in MeOH(40 mL) was heated at 65° C for 2 h. The MeOH was evaporated under reduced pressure and the residue cautiously partitioned between DCM (35 mL) and 20% K2CO3 solution. The aqueous layer was extracted with DCM (2 x 35 mL). The combined organic extracts were dried (Na2SO4) and concentrated in vacuo to afford 641 mg (98%) of 57b as a viscous liquid: ms (ES+) m/z 398 (M+H)+. step 3 – To a solution of 57b (98 mg, 0.25 mmol) in DCM (4 mL) at RT was added 4,4-difluorocyclohexanecarboxyιic acid (49 mg, 0.30mmol). To the resulting solution was added sequentially EDCI (61.4 mg, 0.32 mmol), HOBt (43 mg. 0.32 mmol) and DIPEA (0.13 mL, 0.74 mmol). The mixture was stirred for 4 h. The reaction mixture washed with brine and dried (Na2SO4), then concentrated in vacuo. The crude product was flash chromatographed on silica eiuting with DCM/ 7.5% MeOH (containing 2% NH OH) to afford 113 mg (84%) of 1-485 a white foam: ms (ES+) m/z 544 (M+H)+.

Example 13

Cyclopentanecarboxylic acid {3- [5-(4,6-dimethyl-pyrimidine-5-carbonyl)- hexahydro-pyrrolo[3,4-c]pyrrol-2-yl]-l-phenyl-propyl}-amide (1-29)

 

To a solution of 12 (0.24 g, 0.70 mmol) in DCM (10 mL) were added 4,6-dimethyl- pyrimidine-5-carboxylic acid (55, 0.12 g, 0.84 mmol) , EDCI (0.17 g, 0.91 mmol), HOBt (0.12 g, 0.91 mmol) and DIPEA (0.36 mL, 2.10 mmol). The mixture was stirred at RT for 3 h. The reaction mixture was washed with saturated NaHCO3 and the organic layer was dried (Na2SO4). The crude product was purified by SiO2 column chromatography eiuting with DCM:MeOH:NH4OH (150:10:1) to afford 0.27 g (81 %) of 1-29:: mp 48.0- 49.0 °C; ms (ES+) m/z 476 (M + H); Anal. (C28H37N5O2.0.2M CH2C12) C; calcd, 68.76; found, 68.61; H; calcd, 7.65; found, 7.51; N; calcd, 14.22; found, 14.28

 

……………..
REF
  • 1   Haycock-Lewandowski, S. J.; Wilder, A.; Åhman, J. Org. Process Res. Dev. 2008, 12, 10941103
    (b) Åhman, J.; Birch, M.; Haycock-Lewandowski, S. J.; Long, J.; Wilder, A. Org. Process Res. Dev. 2008, 12, 11041113

    and references therein

  • 2.
    Lee, E. K.; Melville, C. R.; Rotstein, D. M. Chem. Abstr. 2005, 144, 69821
    PCT Int. Publication Number WO/2005/121145 A2, 2005

Filed under: Uncategorized Tagged: RO 5114436, RO5114436

Engineered Virus With Dual Protease Key System Opens to Release Gene Therapy

$
0
0

Originally posted on lyranara.me:

engineered virus 1 Engineered Virus With Dual Protease Key System Opens to Release Gene Therapy

Viruses cause many diseases but can also serve as vectors for delivery of genetic cargo for therapeutic purposes. Rice University researchers have now modified the adeno-associated virus, commonly used to deliver gene therapy, to work like a lock box that opens itself up only in the presence of two different chemical “keys”.

The virus responds to proteases, enzymes that break down other proteins, opening up and releasing the cargo only when both of the markers are present. By selecting which proteases unlock the virus, a new form of therapy may develop that allows doctors to precisely tune where gene delivery happens.

More from Rice:

“We were looking for other types of biomarkers beyond cellular receptors present at disease sites,” [Junghae Suh, bioengineer at Rice] said. “In breast cancer, for example, it’s known the tumor cells oversecrete extracellular proteases, but perhaps more important are the infiltrating immune cells that migrate into the tumor…

View original 262 more words


Filed under: Uncategorized

Regadenoson……..Adenosine A2a receptor agonist, for Coronary artery disease; Sickle cell anemia

$
0
0

Regadenoson structure.svg

2-{4-[(methylamino)carbonyl]- 1H-pyrazol-1-yl}adenosine

(1-{9-[(2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-6-aminopurin-2-yl}pyrazol-4-yl)-N-methylcarboxamide.

US FDA:link

313348-27-5  875148-45-1

Lexiscan, CVT-3146, UNII-7AXV542LZ4, 313348-27-5, CVT 3146, 2-[4-(methylcarbamoyl)-1h-pyrazol-1-yl]adenosine, Rapiscan, Lexiscan (TN)
Molecular Formula: C15H18N8O5
Molecular Weight: 390.35402
Regadenoson is an A2A adenosine receptor agonist that causes coronary vasodilation and used for myocardial perfusion imagining. Manufactured by Astellas and FDA approved April 10, 2008
Codeveloped by Gilead Palo Alto and Astellas, and now marketed by Astellas and Rapidscan, regadenoson is a selective A2a adenosine receptor agonist. The product is indicated as a coronary vasodilator to allow radionuclide myocardial perfusion imaging (MPI) in patients unable to undergo adequate exercise stress. Family members of the product case, WO0078779 start expiring from 2019 to 2025.
CV Therapeutics (Originator), Fujisawa (Licensee)

Regadenoson (INN, code named CVT-3146) is an A2A adenosine receptor agonist that is a coronary vasodilator. It produces hyperemia quickly and maintains it for a duration that is useful for radionuclide myocardial perfusion imaging.[1]

It was approved by the United States Food and Drug Administration on April 10, 2008 and is marketed by Astellas Pharma under the tradename Lexiscan.[2] It is approved for use in the European Union and under the name of Rapiscan. It is currently being marketed by GE Healthcare and is being sold in both the United Kingdom and Germany.

Regadenoson has a 2- to 3-minute biological half-life, as compared with adenosine‘s 30-second half-life. Regadenoson stress protocols using a single bolus have been developed, obviating the need for an intravenous line. Regadenoson stress tests are not affected by the presence of beta blockers, as regadenoson vasodilates but does not stimulate beta adrenergic receptors.

 

Regadenoson is an A2A adenosine receptor agonist that is a coronary vasodilator [see CLINICAL PHARMACOLOGY]. Regadenoson is chemically described as adenosine, 2-[4-[(methylamino)carbonyl]-1H-pyrazol-1-yl]-, monohydrate. Its structural formula is:

 

LEXISCAN® (regadenoson) Structural Formula Illustration

 

The molecular formula for regadenoson is C15H18N8O5 • H2O and its molecular weight is 408.37. Lexiscan is a sterile, nonpyrogenic solution for intravenous injection. The solution is clear and colorless. Each 1 mL in the 5 mL pre-filled syringe contains 0.084 mg of regadenoson monohydrate, corresponding to 0.08 mg regadenoson on an anhydrous basis, 10.9 mg dibasic sodium phosphate dihydrate or 8.7 mg dibasic sodium phosphate anhydrous, 5.4 mg monobasic sodium phosphate monohydrate, 150 mg propylene glycol, 1 mg edetate disodium dihydrate, and Water for Injection, with pH between 6.3 and 7.7.

Regadenoson is also referred to in the literature as CVT- 3146 or (1-{9-[(4S,2R,3R,5R)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-6- aminopurin-2-yl}pyrazol-4-yl)-N-methylcarboxamide and has the formula:

 

Methods for synthesizing regadenoson and related compounds are set forth in U.S. Patent No. 6,403,567, the specification of which is incorporated herein by reference in its entirety.

Regadenoson may be administered by pharmaceutical administration methods that are known in the art. It is preferred that regadenoson is dosed i.v. It is more preferred that regadenoson is administered in a single dose i.v. The term “single dose” refers generally to a single quickly administered dose of a therapeutic amount of regadenoson. The term “single dose” does not encompass a dose or doses administered over an extended period of time by, for example continuous i.v. infusion.

Regadenoson will typically be incorporated into a pharmaceutical composition prior to use. The term “pharmaceutical composition” refers to the combination of regadenoson with at least one liquid carrier that together form a solution or a suspension. Lyophilized powders including compositions of this invention fall within the scope of “pharmaceutical compositions” so long as the powders are intended to be reconstituted by the addition of a suitable liquid carrier prior to use. Examples of suitable liquid carriers include, but are not limited to water, distilled water, de-ionized water, saline, buffer solutions, normal isotonic saline solution, dextrose in water, and combinations thereof.

Regadenoson [(l-{9-[(4S, 2R, 3R, 5R)-3,4-dihydroxy-5-(hydroxymethyl)oxalan-2-yl]-6- aminopurin-2-yl}pyrazol-4-yl)-N-methylcarboxamine] is a selective A2A-adenosine receptor agonist that is a coronary vasodilator. It is currently marketed in the form of a monohydrate as a pharmacologic stress agent indicated for radionuclide myocardial perfusion imaging (MPI) in patients unable to undergo adequate exercise stress.

U.S. Patent No. 8,106,183 describes amorphous regadenoson, and three forms of regadenoson, referred to as Form A (a monohydrate), Form B and Form C.

The synthesis of regadenoson is described, for example, in U.S. Patent Nos. 6,403,567 and 7,183,264. The syntheses disclosed are multi-step processes that proceed via 2- hydrazinoadenosine, which is prepared from the corresponding iodo-derivative (2- iodoadenosine).

 

……………………………

http://www.google.com/patents/WO2012149196A1?cl=en

 

EXAMPLE 1

Synthesis of N-Methyl-4-carboxamide

20 g (143 mmol, 1 equiv) of ethyl pyrazole-4-carboxylate and 200 mL (2310 mmol, 16.2 equiv) of a 40 % aqueous solution of methylamine were added to a three-necked flask equipped with a condenser and a heating mantle. The mixture was stirred to aid dissolution, and heated to 65 °C for 2 hours. The reaction was monitored using HPLC at 220 nm with a C18 column. The reaction mixture was then concentrated in vacuo to obtain a syrup / solid. The crude product was co-evaporated with acetonitrile (3 x 200 mL). 100 mL of acetonitrile was then added to the solids and the mixture was stirred for several hours until the solids were well suspended. The solids were then isolated by filtration, washed with 100 mL acetonitrile, and dried in an oven at 40°C to afford 14.4 g (80 % yield) of N-methyl-4-carboxamide with a purity of 93.5% by HPLC.

EXAMPLE 2

Synthesis of IDAAR-Cu+2

This preparation has reported in the literature. See, e.g., Chinese Chemical Letters, (21(1), 51-54, 2010.

An Erlenmeyer flask was charged with 350 mL of water and 75 g of Chelex 100 resin. With stirring, an aqueous solution of copper sulfate pentahydrate (59 g in 350 mL of water) was slowly added over a period of 15 minutes. The resulting slurry was stirred for 2 hours, then filtered. The resulting solids were washed with 100 – 200 mL of water and dried in a vacuum oven at 50 °C for 16 hours to afford 18 g of IDAAR-Cu+2. The copper content of the product was determined to be 11 wt % using Inductively Coupled Plasma Optical Emission Spectroscopy (ICP-OES).

EXAMPLE 3

Synthesis of Regadenoson Monohydrate

5 g (17.5 mmol, 1 equiv) of 2-fluoroadenosine, 3.07g (24.5 mmol, 1.4 equiv) of N- methylpyrazole-4-carboxamide, and 32 mL of dimethylsulfoxide were added under a nitrogen atmosphere to a dry 3-necked reaction flask equipped with a condenser and a heating mantle.. The mixture was stirred to afford a solution. 100 mL of acetonitrile was then added followed by the addition of 2.2 g of IDAAR-Cu2+ and 5.34 g (5.24 mL, 35.1 mmol, 2 equiv) of

diazabicycloundecene (DBU). The reaction mixture was heated to 70 – 80 °C overnight and monitored by HPLC at 260 nm with a C18 column until the reaction was complete. Then, the reaction mixture was evaporated in vacuo to remove most of the acetonitrile. The remaining dimethylsufoxide solution was purified by reverse phase chromatography using methanol and water. The product was dried in vacuo at a temperature that did not exceed 40° C to afford 3 g (44% yield) of regadenoson monohydrate.

EXAMPLE 4

Synthesis of 2-Hydazineadenosine

2-fluoroadenosine (4g, 14 mmol) was dissolved in 100 mL ethanol in a 300 mL three- necked flask. Hydrazine hydrate (4.1 mL, 6 equivalents, 84 mmol) was added and the mixture was heated to reflux for 1 hour. The reaction mixture was allowed to cool to room temperature and stirred overnight (16 hours). The resulting white precipitate was isolated by filtration and dried in oven at 40°C overnight to afford 2-hydrazinoadenosine (yield: 94%, 3.5g, 96% purity).

EXAMPLE 5

Synthesis of Regadenoson Form D

2-Fluoroadenosine (45 g, 0.158 moL, 1 eq.), 4-(N-methylcarboxamido)pyrazole (27.64 g, 0.221 moL, 1.4 eq.), dimethylsulfoxide (DMSO) (320 mL) and acetonitrile (960 mL) were added to a dry 3000 ml 3-neck reaction flask equipped with a condenser and heating mantle. After stirring for 10 minutes, IDAAR-Cu (20.07 g, 0.032 moL, 0.2 eq.) and DBU (48.0 g, 0.316 moL, 2 eq.) were added. The resulting mixture was then heated to 65°C overnight (18 hours).

The reaction mixture was then filtered and the filtrate was evaporated followed by 2 x 500 mL co-evaporation with xylene. The residue was diluted with 5 L acetonitrile, transferred to a 10 L flask and kept in a cold room (4°C) overnight. The resulting white precipitate was isolated by filtration and stirred in 1.8 L of water. The mixture was heated to 80° C for 2 hours, then allowed to cool in a cold room (4°C) overnight.

The white precipitate was isolated by filtration, then dissolved in 200 ml of 1 : 1 mixture of DMSO and methanol. The clear and slightly yellow solution was loaded to a reverse phase column (10 L) and eluted with water/methanol (gradient with a 5% increase of MeOH every 10 L).

The fractions with HPLC purity of more than 99.9% were combined and concentrated to a paste. The supernatant liquid was decanted and the flask heated in an oil-bath at 150° C under reduced pressure of 20mmHg for 6 hours to afford 6.2 g of Regadenoson Form D as white solid (99.94% HPLC, KF analysis 0.8%).

The fractions with HPLC purity between 50 and 99.8% (~ 23g of product as indicated by HPLC) were combined and subjected to a second purification stage.

 

 

………………………………

WO 0078779

https://www.google.com/patents/WO2000078779A2?cl=en

Example 5

 

(l-{9-[(4S,2R,3R,5R)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-6-aminopurin-2- yl}pyrazol-4-yl)-N-methylcarboxamide (16)

Compound 12 (0.05 mg, 0.12 mmol) was added to 4 mL methylamine (40% sol. In water). The mixture heated at 65 °C in for 24 h. After concentration in vacuo, the residue was purified using prep. TLC (10% MeOH:DCM). ‘HNMR (CD3OD) 62.90 (s, 3 H), 3.78 (m, 1

H), 3.91 (m, 1 H), 4.13 (d, 1 H), 4.34 (d, 1 H), 4.64 (m, 1 H), 6.06 (d, 1 H), 8.11 (s, 1 H), 8.38

(s, 1 H), 9.05 (s, 1 H).

 

…………………..

https://www.google.com/patents/US6403567

U.S. Patent Nos. 6,403,567

Scheme 1.

 

Compound I can be prepared by reacting compound 1 with appropriately substituted 1,3 -dicarbonyl in a mixture of AcOH and MeOH at 80° C. (Holzer et al., J. Heterocycl. Chem. (1993) 30, 865). Compound II, which can be obtained by reacting compound I with 2,2-dimethoxypropane in the presence of an acid, can be oxidized to the carboxylic acid III, based on structurally similar compounds using potassium permanganate or pyridinium chlorochromate (M. Hudlicky, (1990) Oxidations in Organic Chemistry, ACS Monographs, American Chemical Society, Washington D.C.). Reaction of a primary or secondary amine having the formula HNR6R7, and compound III using DCC (M. Fujino et al., Chem. Pharm. Bull. (1974), 22, 1857), PyBOP (J. Martinez et al., J. Med. Chem. (1988) 28, 1874) or PyBrop (J. Caste et al. Tetrahedron, (1991), 32, 1967) coupling conditions can afford compound IV.

 

Compound V can be prepared as shown in Scheme 2. The Tri TBDMS derivative 4 can be obtained by treating compound 2 with TBDMSCl and imidazole in DMF followed by hydrolysis of the ethyl ester using NaOH. Reaction of a primary or secondary amine with the formula HNR6R7, and compound 4 using DCC (M. Fujino et al., Chem. Pharm. Bull. (1974), 22, 1857), PyBOP (J. Martinez et al., J. Med. Chem. (1988) 28, 1874) or PyBrop (J. Caste et al. Tetrahedron, (1991), 32, 1967) coupling conditions can afford compound V.

 

A specific synthesis of compound 11 is illustrated in Scheme 3. Commercially available guanosine 5 was converted to the triacetate 6 as previously described (M. J. Robins and B. Uznanski, Can. J. Chem. (1981), 59, 2601-2607). Compound 7, prepared by following the literature procedure of Cerster et al. (J. F. Cerster, A. F. Lewis, and R. K. Robins, Org. Synthesis, 242-243), was converted to compound 9 in two steps as previously described (V. Nair et al., J. Org. Chem., (1988), 53, 3051-3057). Compound 1 was obtained by reacting hydrazine hydrate with compound 9 in ethanol at 80° C. Condensation of compound 1 with ethoxycarbonylmalondialdehyde in a mixture of AcOH and MeOH at 80° C. produced compound 10. Heating compound 10 in excess methylamine afforded compound 11.

 

The synthesis of 1,3-dialdehyde VII is described in Scheme 4. Reaction of 3,3-diethoxypropionate or 3,3-diethoxypropionitrile or 1,1-diethoxy-2-nitroethane VI (R3=CO2R, CN or NO2) with ethyl or methyl formate in the presence of NaH can afford the dialdehyde VII (Y. Yamamoto et al., J. Org. Chem. (1989) 54, 4734).

EXAMPLE 5

 

 

(1-{9-[(4S,2R,3R,5R)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-6-aminopurin-2 -yl}pyrazol-4N-methylcarboxamide which can also be identified as 2-(4-methylaminocarbonylpyrazol-1-yl)adenosine (16)

The mixture heated at 65° C. in for 24 h. After concentration in vacuo, the residue was purified using prep. TLC (10% MeOH:DCM). 1HNMR (CD3OD) δ2.90 (s, 3 H), 3.78 (m, 1 H), 3.91 (m, 1 H), 4.13 (d, 1 H), 4.34 (d, 1 H), 4.64 (m, 1 H), 6.06 (d, 1 H), 8.11 (s, 1 H), 8.38 (s, 1 H), 9.05 (s, 1 H).

………………………….

US 7,183,264

http://www.google.com/patents/US7183264

EXAMPLE 5

 

(1-{9-[(4S,2R,3R,5R)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-6-aminopurin-2-yl}pyrazol-4-yl)-N-methylcarboxamide (16)

 

Compound 12 (0.05 mg, 0.12 mmol) was added to 4 mL methylamine (40% sol. In water). The mixture heated at 65° C. in for 24 h. After concentration in vacuo, the residue was purified using prep. TLC (10% MeOH:DCM). 1HNMR (CD3OD) δ2.90 (s, 3 H), 3.78 (m, 1 H), 3.91 (m, 1 H), 4.13 (d, 1 H), 4.34 (d, 1 H), 4.64 (m, 1 H), 6.06 (d, 1 H), 8.11 (s, 1 H), 8.38 (s, 1 H), 9.05 (s, 1 H).

 

References

  1. Cerqueira MD (July 2004). “The future of pharmacologic stress: selective A2A adenosine receptor agonists”. Am. J. Cardiol. 94 (2A): 33D–40D; discussion 40D–42D. doi:10.1016/j.amjcard.2004.04.017. PMID 15261132.
  2. CV Therapeutics and Astellas Announce FDA Approval for Lexiscan(TM)

 

12-28-2007
Use of A2A Adenosine Receptor Agonists in the Treatment of Ischemia
2-28-2007
N-pyrazole A2A receptor agonists
1-10-2007
Polymer coating for medical devices
5-5-2006
Polymer coating for medical devices

 

1-32-2012
USE OF A2A ADENOSINE RECEPTOR AGONISTS
1-32-2012
PROCESS FOR PREPARING AN A2A-ADENOSINE RECEPTOR AGONIST AND ITS POLYMORPHS
10-21-2011
PROCESS FOR PREPARING AN A2A-ADENOSINE RECEPTOR AGONIST AND ITS POLYMORPHS
6-8-2011
PROCESS FOR PREPARING AN A2A-ADENOSINE RECEPTOR AGONIST AND ITS POLYMORPHS
6-9-2010
Process for preparing an A2A-adenosine receptor agonist and its polymorphs
3-3-2010
PROCESS FOR PREPARING AN A2A-ADENOSINE RECEPTOR AGONIST AND ITS POLYMORPHS
2-3-2010
Use of A2A adenosine receptor agonists
9-3-2008
Polymer coating for medical devices
7-4-2008
POLYMER COATING FOR MEDICAL DEVICES
4-30-2008
Polymer coating for medical devices

 

US6403567 Jun 22, 1999 Jun 11, 2002 Cv Therapeutics, Inc. To stimulate mammalian coronary vasodilatation and for imaging the heart; regadenoson
US7183264 Aug 29, 2003 Feb 27, 2007 Cv Therapeutics, Inc. Such as ethyl-1-(9-((4S,2R,3R,5R)-3,4-dihydroxy-5-(hydroxy-methyl)oxolan-2-yl)-6 -aminopurin-2-yl)pyrazole-4-carboxylate; adenosine receptors (A2A); for stimulating mammalian coronary vasodilatation for therapy and imaging the heart
US7732595 Feb 2, 2007 Jun 8, 2010 Gilead Palo Alto, Inc. Process for preparing an A2A-adenosine receptor agonist and its polymorphs
US8106183 Apr 22, 2010 Jan 31, 2012 Gilead Sciences, Inc. Process for preparing an A2A-adenosine receptor agonist and its polymorphs

 

NEW PATENT

Novel process for the preparation of (1-{9-[(4S,2R,3R,5R)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl)-6-aminopurin-2-yl}pyrazole-4-yl)-N-methylcarboxamide

WO-2014068589

Biophore India Pharmaceuticals Pvt Ltd


Filed under: Uncategorized Tagged: regadenoson

New technique uses ATP as trigger for targeted anti-cancer drug delivery

$
0
0

Originally posted on lyranara.me:

New technique uses ATP as trigger for targeted anti-cancer drug delivery

Biomedical engineering researchers from North Carolina State University and the University of North Carolina have developed a new technique that uses adenosine-5′-triphosphate (the so-called ‘energy molecule’) to trigger the release of anti-cancer drugs directly into cancer cells. The spherical nanoparticles are coated with a shell that incorporates hyaluronic acid, which interacts with proteins found on the surface of some cancer cells. The nanoparticle is filled with DNA molecules that release anti-cancer drug doxorubicin when they come into contact with the adenosine-5′-triphosphate inside a cancer cell. Credit: Ran Mo

Biomedical engineering researchers have developed a new technique that uses adenosine-5′-triphosphate (ATP), the so-called “energy molecule,” to trigger the release of anti-cancer drugs directly into cancer cells. Early laboratory tests show it increases the effectiveness of drugs targeting breast cancer. The technique was developed by researchers at North Carolina State University and the University of North Carolina at Chapel Hill.

“This is…

View original 339 more words


Filed under: Uncategorized

New method sneaks drugs into cancer cells before triggering release

$
0
0

Originally posted on lyranara.me:

A liposome-based co-delivery system composed of a fusogenic liposome encapsulating ATP-responsive elements with chemotherapeutics and a liposome containing ATP was developed for ATP-mediated drug release triggered by liposomal fusion. The fusogenic liposome had a protein–DNA complex core containing an ATP-responsive DNA scaffold with doxorubicin (DOX) and could release DOX through a conformational change from the duplex to the aptamer/ATP complex in the presence of ATP. A cell-penetrating peptide-modified fusogenic liposomal membrane was coated on the core, which had an acid-triggered fusogenic potential with the ATP-loaded liposomes or endosomes/lysosomes. Directly delivering extrinsic liposomal ATP promoted the drug release from the fusogenic liposome in the acidic intracellular compartments upon a pH-sensitive membrane fusion and anticancer efficacy was enhanced both in vitro and in vivo.
New method sneaks drugs into cancer cells before triggering release

Biomedical engineering researchers have developed an anti-cancer drug delivery method that essentially smuggles the drug into a cancer cell before triggering its release. Credit: Ran Mo

Biomedical engineering…

View original 456 more words


Filed under: Uncategorized

For National Women’s Health Week, FDA Resources Help Women Make Informed Health Choices

$
0
0


For National Women’s Health Week, FDA Resources Help Women Make Informed Health Choices

By: Marsha B. Henderson, M.C.R.P. “Ask your mother.” In households throughout the country, women often make decisions about foods and medical products for themselves and their loved ones. As we celebrate National Women’s Health Week (May 11-17), I want to … Continue reading →http://blogs.fda.gov/fdavoice/index.php/2014/05/for-national-womens-health-week-fda-resources-help-women-make-informed-health-choices/?source=govdelivery&utm_medium=email&utm_source=govdelivery


Filed under: FAST TRACK FDA Tagged: fda

Amgen-AstraZeneca Psoriasis Drug Brodalumab (AMG 827) Hits Phase 3 Endpoints

$
0
0

 

 

AstraZeneca and Amgen announced that the Phase 3 AMAGINE-1TM study evaluating brodalumab in patients with moderate-to-severe plaque psoriasis met all primary and secondary endpoints for both evaluated doses.

Read more… http://www.dddmag.com/news/2014/05/amgen-astrazeneca-psoriasis-drug-hits-phase-3-endpoints?et_cid=3935059&et_rid=523035093&type=cta

Brodalumab is a human monoclonal antibody designed for the treatment of inflammatory diseases.[1] It is being tested for the treatment of moderate to severe psoriasis[2] in Phase III clinical trials as of November 2013.[3][4]

Brodalumab was developed by Amgen, Inc.

Mechanism of action

Brodalumab binds to the interleukin-17 receptor and so prevents interleukin 17 (IL-17) from activating the receptor. This mechanism is similar to that of another anti-psoriasis antibody, ixekizumab, which however binds to IL-17 itself.[2]
At present, brodalumab is the only experimental drug in development that inhibits the IL-17 receptor, thus inhibiting several of the IL-17 ligands at once from transmitting signals to the body. Other agents currently in development seek to target the individual IL-17 ligands. By inhibiting the attachment of these ligands with the receptor, brodalumab stops the body from receiving signals that may otherwise cause inflammation and other ailments.

Researchers are currently investigating brodalumab for the treatment of psoriasis (Phase II and planned Phase III), asthma (Phase II), and psoriatic arthritis (Phase II).

Psoriasis is a chronic disease of the immune system that causes the skin cells to grow at a faster rate. Worldwide, the condition affects around 125 million individuals. Even though several types of psoriasis exist, around 80% of sufferers have plaque psoriasis. Plaque psoriasis can cause painful and itchy red, scaly patches to appear on the skin.

Brodalumab

(AMG 827)

Monoclonal antibody
Type Whole antibody
Source Human
Target Interleukin 17 receptor A
Clinical data
Legal status Investigational
Identifiers
CAS number 1174395-19-7
ATC code None
KEGG D10061 
Chemical data
Formula C6372H9840N1712O1988S52 
Mol. mass 144.06 kDa

 

About Brodalumab (AMG 827)

Brodalumab is a novel human monoclonal antibody that binds to the interleukin-17 (IL-17) receptor and inhibits inflammatory signaling by blocking the binding of several IL-17 ligands to the receptor. By stopping IL-17 ligands from activating the receptor, brodalumab prevents the body from receiving signals that may lead to inflammation. The IL-17 pathway plays a central role in inducing and promoting inflammatory disease processes. In addition to moderate-to-severe plaque psoriasis (Phase 3), brodalumab is currently being investigated for the treatment of psoriatic arthritis (Phase 3) and asthma (Phase 2).

About the Amgen and AstraZeneca Collaboration

In April 2012, Amgen and AstraZeneca formed a collaboration to jointly develop and commercialize five monoclonal antibodies from Amgen’s clinical inflammation portfolio. With oversight from joint governing bodies, Amgen leads clinical development and commercialization for brodalumab (Phase 3 for moderate-to-severe plaque psoriasis and psoriatic arthritis, Phase 2 for asthma) and AMG 557/MEDI5872 (Phase 1b for autoimmune diseases such as systemic lupus erythematosus). AstraZeneca, through its biologics arm MedImmune, leads clinical development and commercialization for MEDI7183/AMG 181 (Phase 2 for ulcerative colitis and Crohn’s disease), MEDI2070/AMG 139 (Phase 2 for Crohn’s disease) and MEDI9929/AMG 157 (Phase 2 for asthma).

About Amgen

Amgen is committed to unlocking the potential of biology for patients suffering from serious illnesses by discovering, developing, manufacturing and delivering innovative human therapeutics. This approach begins by using tools like advanced human genetics to unravel the complexities of disease and understand the fundamentals of human biology.

Amgen focuses on areas of high unmet medical need and leverages its biologics manufacturing expertise to strive for solutions that improve health outcomes and dramatically improve people’s lives. A biotechnology pioneer since 1980, Amgen has grown to be the world’s largest independent biotechnology company, has reached millions of patients around the world and is developing a pipeline of medicines with breakaway potential.

For more information, visit www.amgen.com and follow us on www.twitter.com/amgen.

 

About AstraZeneca

AstraZeneca is a global, innovation-driven biopharmaceutical business that focuses on the discovery, development and commercialisation of prescription medicines, primarily for the treatment of cardiovascular, metabolic, respiratory, inflammation, autoimmune, oncology, infection and neuroscience diseases. AstraZeneca operates in over 100 countries and its innovative medicines are used by millions of patients worldwide. For more information please visit: www.astrazeneca.com.

References

  1. “Statement On A Nonproprietary Name Adopted By The USAN Council: Brodalumab”. American Medical Association.
  2. “Neue Antikörper in der Pipeline”. Pharmazeutische Zeitung (in German) (12). 2012.
  3. ClinicalTrials.gov NCT01708590 Study of Efficacy, Safety, and Withdrawal and Retreatment With Brodalumab in Moderate to Severe Plaque Psoriasis Subjects (AMAGINE-1)
  4. ClinicalTrials.gov NCT01708629 Study of Efficacy and Safety of Brodalumab Compared With Placebo and Ustekinumab in Moderate to Severe Plaque Psoriasis Subjects (AMAGINE-3)

http://ksclinic.exblog.jp/18270693/

学術面で最初の講演は、米国のJames Krueger教授による「Th1細胞,Th17細胞,Th22細胞が複雑なサイトカインネットワークによって、細胞レベル、分子レベルで乾癬を引き起こす」でした。その要約を示すスライドを幾枚か失敬します(Krueger先生、ごめんなさい)。

乾 癬の原因究明、病態(病気の起こり方)解明の主役となった免疫学的研究の最先端を行くKrueger先生の、最新情報がコンパクトにまとまった素晴らしい 講演でした。生物学的製剤の治療根拠となるサイトカインネットワークは、現在TipDC – Th17経路によって、きわめて明快に説明されるようになり、Th17細胞が放出するIL17が表皮細胞(ケラチノサイト)の乾癬化を起こします。現在使 用されている抗TNFα製剤、抗IL12/23製剤が、より上流(免疫反応の根っこ)で免疫反応を抑制するのに比べ、IL17はより末梢における乾癬の原 因サイトカインであることから、IL17の抑制は、より乾癬をピンポイントで、そして副作用もミニマムにすることが期待される。

現在、3種類のIL17抑制薬剤が開発され、治療研究が進められている。
①IL17A抗体(Secukinumab Novartis社)
②IL17A抗体(Ixekizumab Lilly社)
③IL17A受容体抗体(Brodalumab Amgen社)


その一つ、Secukinumabの効果(PASI75)=すごく乾癬がよくなる)では、たった3回の注射で90%以上の患者がPASI75を達成する。


PASI90(=乾癬がほとんどなくなる)でみても、60%の患者で達成されている。

Secukinumabの臨床効果。上の段は「プラセボ(偽薬)」、下の段がSecukinumab。

Ixekizumabの効果(PASI90)。約80%の患者で達成されている。驚異的である。

Brodalumabの臨床効果

印象深かった講演をもう一つ、詳細に紹介いたします。
米 国のAnne Bowcock教授の”The genetics of psoriasis: Old risks, novel loci (乾癬の遺伝子研究:昔から言われていた異常、新しく見つかった場所)です。Bowcock教授は、乾癬の原因遺伝子について世界で最初に報告した研究者 です。ここでも少し講演スライドを拝借(Bowcock先生、ごめんなさい)。

Bowcock 教授は1999年、乾癬家系の詳細な遺伝子調査から第17染色体に乾癬と関わり深い遺伝子異常があることをみつけ、科学雑誌Scienceに報告した。 21世紀を迎える直前のことであり、遠からず乾癬の原因遺伝子が確定し、完治治療を開発することも夢ではないと、当時期待したものでした。
ところが、次々と関連遺伝子はみつけられるものの(現在は30種類以上)、肝心の原因遺伝子、特定のタンパク、メカニズムは不明のままでした。

Bowcock 教授の息の長い研究は、第17染色体上にあるCARD14と呼ばれるタンパクの、その異常が直接乾癬を起こすことを説き明かしました。CARD14は細胞 膜上にあるタンパクで、細胞外で起こる炎症から生じる様々な刺激物質を、細胞の膜から細胞の中へ伝える役割を果たしています。その伝達経路はNFκBを介 しています(乾癬ではこの経路が活発に動いていることが、高知大学の佐野教授により解明されました)。

遺伝性膿疱性乾癬患者では、このCARD14遺伝子に点突然変異が起こっていることを発見しました。この点突然変異だけで、特殊タイプではありますが、乾癬の原因が特定されたのです。

点突然変異だけではなく、CARD14遺伝子に起こりやすい変異も、ほかの遺伝子異常(PSORS1、MHC遺伝子)、あるいは環境変化が加わると乾癬を引き起こすことも証明しました。

大変感銘深い講演でした。
会議の模様、IFPA代表者会議の報告は、また後日掲載いたします(『2012年9月教室抄録』をご覧ください)。
ブログ「PHOTO & ESSAY」もご覧ください。


Filed under: ANTIBODIES, Monoclonal antibody, Phase3 drugs Tagged: AMG 827, Brodalumab, IL-17 receptor, Monoclonal antibody, Psoriasis

La cantera de los biológicos

$
0
0

Originally posted on biologicos et al.:

Los medicamentos biotecnológicos están en plena ebullición, de forma continuada aparecen nuevas moléculas. Muchas de ellas se encuentran en fases preliminares de desarrollo y se espera que en los próximos años se vayan aprobando. ¿Se mantendrá este ritmo en el tiempo?

View original 320 more words


Filed under: Uncategorized

Very First Human Trials Using Cannbis To Treat Brain Cancer Are Under Way

$
0
0

hemp

 

 

 

 

 

 

 

 

The picture to your left is showing immunofluorescence of the human glioma cell line. (View more pictures here)

A European based pharmaceutical company called GW Pharmaceuticals is set to commence its first phase of clinical trials for the treatment of Glioblastoma Multiforme (GBM). It’s a bio-pharmaceutical company focused on discovering, developing and commercializing novel therapeutics from its proprietary cannabinoid product platform.

According to the New England Journal of Medicine, GBM accounts for approximately 50% of the 22,500 new cases of brain cancer diagnosed in the United States alone each year.(1) Treatment with regards to brain cancer are very limited which makes the study of cannabis and its effect on brain tumors crucial.

http://www.hempforfuture.com/2014/03/26/very-first-human-trials-using-cannbis-to-treat-brain-cancer-are-under-way/?utm_content=buffer8c9a9&utm_medium=social&utm_source=facebook.com&utm_campaign=buffer

 


The picture to your left is showing immunofluorescence of the human glioma cell line. (View more pictures here) A European…
WWW.HEMPFORFUTURE.COM

Filed under: cancer, Uncategorized Tagged: CANCER, cannabis

BIOSIMILARS MARKET IS NOT EQUIVALENT TO GENERICS MARKET

Pharma Trends: Global Medicine Spending to Pass $1 Trillion in 2014

$
0
0

This year will mark a milestone in the pharmaceutical industry’s growth as drug sales will exceed $1 Trillion. This was the finding of IMS Health in their new report on the spending on pharmaceuticals and trends that will affect it between now and 2017.
Two factors will be the major drivers of this growth, increased spending on medicines in emerging markets and increased spending on new drugs for cancer and orphan diseases.

http://msg-latam-sfb.blogspot.in/2013/12/pharma-trends-global-medicine-spending.html


Filed under: COMPANIES Tagged: medicine, PHARMA

Ezatiostat……….designed to stimulate the production of blood cells in the bone marrow

$
0
0

Ezatiostat structure

Ezatiostat

168682-53-9 (Ezatiostat); 286942-97-0 (Ezatiostat HCl salt)

gamma-Glu-S-BzCys-PhGly diethyl ester

 HCl; salt, D08917, 

Ezatiostat hydrochloride

Target: glutathione S-transferase P1-1 (GSTP1-1) inhibitor
Pathway: hsa00480 Glutathione metabolism
Activity: Treatment of disorders of bone marrow cellular growth and differentiation

see http://www.ncbi.nlm.nih.gov/pubmed?term=TLK-199&cmd=search

Telik, Inc.

innovator

TLK199; TLK-199; TLK 199; Brand name: TELINTRA®ethyl (2R)-[(4S)-4-amino-5-ethoxy-5-oxopentanoyl]-S-benzyl-L-cysteinyl-2- phenylglycinate. 

ethyl (2S)-2-amino-5-[[(2R)-3-benzylsulfanyl-1-[[(1R)-2-ethoxy-2-oxo-1-phenylethyl]amino]-1-oxopropan-2-yl]amino]-5-oxopentanoate.

IUPAC/Chemical name: 

(S)-ethyl 2-amino-5-(((R)-3-(benzylthio)-1-(((S)-2-ethoxy-2-oxo-1-phenylethyl)amino)-1-oxopropan-2-yl)amino)-5-oxopentanoate

C27H35N3O6S
Exact Mass: 529.2246

nmr.http://www.medkoo.com/Product-Data/Ezatiostat/ezatiostat-QC-CRB40225web.pdf

Telintra is a small molecule product candidate designed to stimulate the production of blood cells in the bone marrow. Many conditions are characterized by depleted bone marrow, including myelodysplastic syndrome, a form of pre-leukemia in which the bone marrow produces insufficient levels of one or more of the 3 major blood elements (white blood cells, red blood cells and platelets). A reduction in blood cell levels is also a common, toxic effect of many standard chemotherapeutic drugs.

Ezatiostat is a liposomal small-molecule glutathione analog inhibitor of glutathione S-transferase (GST) P1-1 with hematopoiesis-stimulating activity. After intracellular de-esterification, the active form of ezatiostat binds to and inhibits GST P1-1, thereby restoring Jun kinase and MAPK pathway activities and promoting MAPK-mediated cellular proliferation and differentiation pathways. This agent promotes the proliferation and maturation of hematopoietic precursor cells, granulocytes, monocytes, erythrocytes and platelets

Phase II trial myelodysplastic syndrome (MDS): Cancer. 2012 Apr 15;118(8):2138-47.

Phase I trial myelodysplastic syndrome (MDS): J Hematol Oncol. 2012 Apr 30;5:18. doi: 10.1186/1756-8722-5-18; Blood. 2009 Jun 25;113(26):6533-40; J Hematol Oncol. 2009 May 13;2:20.

Ezatiostat hydrochloride is the hydrochloride acid addition salt of ezatiostat. Ezatiostat, also known as TLK199 or TER 199, is a compound of the formula:

 

Ezatiostat has been shown to induce the differentiation of HL-60 promyelocyte leukemia cells in vitro, to potentiate the activity of cytotoxic agents both in vitro and in vivo, and to stimulate colony formation of all three lineages of hematopoietic progenitor cells in normal human peripheral blood.

In preclinical testing, ezatiostat has been shown to increase white blood cell production in normal animals, as well as in animals in which white blood cells were depleted by treatment with cisplatin or fluorouracil. Similar effects may provide a new approach to treating myelodysplastic syndrome (MDS).

Many conditions, including MDS, a form of pre-leukemia in which the bone marrow produces insufficient levels of one or more of the three major blood elements (white blood cells, red blood cells, and platelets), are characterized by depleted bone marrow. Myelosuppression, which is characterized by a reduction in blood cell levels and in a reduction of new blood cell generation in the bone marrow, is also a common, toxic effect of many standard chemotherapeutic drugs.

Ezatiostat hydrochloride in a liposomal injectable formulation was studied in a clinical trial for the treatment of MDS, and results from this trial, reported by Raza et al., J Hem. One, 2:20 (published online 13 May 2009), demonstrated that administration of TLK199 was well tolerated and resulted in multi-lineage hematologic improvement.

Ezatiostat hydrochloride in a tablet formulation has been evaluated in a clinical trial for the treatment of MDS, as reported by Raza et al., Blood, 113:6533-6540 (prepublished online 27 April 2009) and a single-patient report by Quddus et al., J Hem. One, 3:16 (published online 23 April 2010), and is currently being evaluated in clinical trials for the treatment of MDS and for severe chronic idiopathic neutropenia.

When used for treating humans, it is important that a crystalline therapeutic agent like ezatiostat hydrochloride retains its polymorphic and chemical stability, solubility, and other physicochemical properties over time and among various manufactured batches of the agent. If the physicochemical properties vary with time and among batches, the administration of a therapeutically effective dose becomes problematic and may lead to toxic side effects or to ineffective therapy, particularly if a given polymorph decomposes prior to use, to a less active, inactive, or toxic compound.

Therefore, it is important to choose a form of the crystalline agent that is stable, is manufactured reproducibly, and has physicochemical properties favorable for its use as a therapeutic agent.

Ezatiostat hydrochloride (USAN) has the molecular weight of 566.1, the trademark of Telintra®, and the CAS registry number of 286942-97-0. Ezatiostat hydrochloride has been evaluated for the treatment of myelodysplastic syndrome (MDS), in a Phase I-IIa study using a liposomal formulation (U.S. Pat. No. 7,029,695), as reported at the 2005 Annual Meeting of the American Society for Hematology (Abstract #2250) and by Raza et al. in Journal of Hematology & Oncology, 2:20 (published online on 13 May 2009); and in a Phase I study using a tablet formulation, as reported at the 2007 Annual Meeting of the American Society for Hematology (Abstract #1454) and by Raza et al. in Blood, 113:6533-6540 (prepublished online on 27 Apr. 2009), and in a single patient case report by Quddus et al. in Journal of Hematology & Oncology, 3:16 (published online on 23 Apr. 2010).

…………………………………………………………………

http://www.google.com/patents/US20110301376

 

Preparation of Ezatiostat Hydrochloride

In another aspect, this invention provides a process comprising the steps of contacting a compound of formula:

 

 

or a salt thereof with a compound of formula:

 

 

or a salt thereof and an activating agent under conditions which provide a compound of formula:

 

 

In one embodiment, the process further comprises deprotecting the compound of formula:

 

 

under conditions which provide a compound of formula:

 

 

or a salt thereof. In another embodiment, the compound provided is ezatiostat hydrochloride.

In another aspect, this invention provides a process comprising contacting a compound of formula:

 

 

or a salt thereof with an ethylating agent under conditions which provide a compound of formula:

 

 

In another embodiment, the process further comprises debenzylating the compound of formula:

 

 

under conditions which provide a compound of formula:

 

 

or a salt thereof.

In another aspect, this invention provides a process comprising the steps of contacting a compound of formula:

 

 

or a salt thereof having a t-butoxycarbonyl group with an activating agent and a compound of formula:

 

 

or a salt thereof under conditions which provide a compound of formula:

 

 

In another embodiment, the process further comprises deprotecting the tertiarybutyloxycarboyl (Boc) group under conditions to provide a compound of formula:

 

 

or a salt thereof.

Certain preferred embodiments of this invention are illustrated in the reaction scheme and described below. In the peptide coupling the amino acid reagents are used generally at a 1:1 molar ratio, and the activating reagent (isobutyl chloroformate) and the base (NMM) are used in slight excess over the amino acid reagents; while in the esterification of the N-BOC-L-glutamic acid γ-benzyl ester the esterifying agent (diethyl sulfate) and base are used in about 1.4-fold excess.

 

EXAMPLESAs relevant and unless otherwise noted, all operations were conducted under nitrogen purge and with stirring. Water was osmosis purified, and solvents were filtered. Unless otherwise stated, all temperatures are in degrees Celcius (° C.) and the following abbreviations have the following definitions:Et EthylHCl(g) HCl gasN-BOC or N-Boc N-tertiarybutyloxycarbonylL LiterKg KilogramNMM N-methylmorpholineMol Molew/w weight by weightExample 1Preparation of S-benzyl-L-cysteinyl-D-phenylglycine ethyl ester hydrochloride (3)Without stirring, 45.1 Kg N-BOC-S-benzyl-L-cysteine (1) was added to a 600 L jacketed glass-lined reactor, followed by 45 L ethyl acetate. Stirring was started and the temperature was reduced to 13° C. NMM, 15.3 Kg, was added over 50 minutes, and rinsed in with 6 L ethyl acetate, and stirring stopped. Ethyl acetate, 315 L, was added to an 800 L cooled jacketed glass-lined reactor, followed by 20.7 Kg isobutyl chloroformate, rinsed in with 11 L ethyl acetate, and the mixture cooled to −10° C. The N-BOC-S-benzyl-L-cysteine NMM salt solution was added to the 800 L reactor over 5 hours, its reactor rinsed with 11 L ethyl acetate, and the rinse solution added to the 800 L reactor, while maintaining the temperature at (−10˜−7)° C. D-Phenylglycine ethyl ester hydrochloride, 31.2 Kg, was added in 8 portions over 50 minutes, followed by 15.3 Kg NMM in 8 portions over 1.3 hours, rinsed in with 2×5 L portions of ethyl acetate, allowing the mixture to warm to −1° C. by the end of the addition. The mixture was gradually warmed to 1° C. for 30 minutes, then to 20° C. over 2 hours, and maintained at (20˜25)° C. for 5 hours. The reaction mixture was washed twice with water: the first time adding 66 L water, stirring at room temperature for 40 minutes, allowing the phases to separate for 30 minutes, then removing the aqueous phase; the second time adding 68 L water, bringing the pH to 1.9 with the addition of 0.45 L 36% hydrochloric acid, stirring at room temperature for 35 minutes, allowing the phases to separate for 1 hour, then removing the aqueous phase. The organic phase was then heated to 38° C., and the pressure reduced to about 0.25 bar until no further gas was released, then to about (0.07-0.1) bar and solvents removed by distillation until 266 L of distillate had been removed. Four cycles of addition of 45 L ethyl acetate and removal of 45 L solvent by distillation were performed, and the water content of the remaining mixture was checked to ensure that it was below 0.1%. With the mixture at 36° C., 194 L heptanes was added, maintaining the temperature about 36° C., and held at that temperature for 2.3 hours. A further 194 L heptanes was added, allowing the temperature to cool to 30° C., and the temperature then reduced to −1° C. over 2.3 hours and then to −5° C. over 1 hour, and N-BOC-S-benzyl-L-cysteinyl-D-phenylglycine ethyl ester recovered by filtration, washing twice with 30 L each of heptanes at −5° C., giving 85 Kg (63 Kg dry basis) N-BOC-S-benzyl-L-cysteinyl-D-phenylglycine ethyl ester. Without stirring, the damp N-BOC-S-benzyl-L-cysteinyl-D-phenylglycine ethyl ester was loaded into an 800 L jacketed glass-lined reactor, followed by 257 L ethyl acetate. Stirring was started and the temperature brought to 22° C., then the nitrogen purge stopped and 12.2 Kg hydrogen chloride gas was added through an immersion tube over 1.8 hours, allowing the temperature to increase to 38° C. The temperature was increased to 41° C., and the mixture held at that temperature for 9 hours. About 280 L of solvents were removed by distillation at that temperature and a pressure of (0.2˜0.1) bar over about 2 hours. Two cycles of addition of ethyl acetate and removal of solvent by distillation were performed, using 52 L in the first cycle and 77 L in the second cycle, and the viscous solution of S-benzyl-L-cysteinyl-D-phenylglycine ethyl ester hydrochloride (3) in ethyl acetate, 148 Kg, was cooled to room temperature and filtered into a storage drum.Example 2Preparation of N-BOC-L-glutamic acid α-ethyl ester (6)Without stirring, 41 Kg N-BOC-L-glutamic acid γ-benzyl ester (4) was added to an 800 L jacketed glass-lined reactor, followed by 2.5 L water and 123 L ethyl acetate. The mixture was then stirred until the N-BOC-L-glutamic acid γ-benzyl ester completely dissolved, keeping the temperature below 15° C. Potassium carbonate fine powder, 23.4 Kg, was added in five batches, and the mixture then heated to 55° C. and maintained at that temperature for 40 minutes, giving a heterogeneous and completely fluid mixture. Diethyl sulfate, 26.2 Kg, was added over 2 hours, and rinsed in with 5 L ethyl acetate, with the temperature remaining at about 52° C. The nitrogen purge was stopped and a solution of 20 Kg ammonium chloride in 73 L water at room temperature added over 2 hours to the mixture, maintaining the temperature near 50° C., then rinsing in with 10 L water. Nitrogen purging was resumed, and the mixture was maintained at about 50° C. for 3 hours, then lowered to about 45° C., the stirring stopped, the phases allowed to separate for 30 minutes, and the lower, aqueous, phase removed. The organic phase, containing N-BOC-L-glutamic acid γ-benzyl α-ethyl ester (5), was washed three times with water, each time adding 41 L water, stirring at room temperature for 30 minutes, allowing the phases to separate for 30 minutes, then removing the aqueous phase. The organic phase was heated to 35° C., and the pressure reduced, starting at about 0.2 bar and reducing as necessary until 82 Kg solvent had been removed by distillation, leaving about (70˜80) L of slightly opalescent solution. This solution was heated to 53° C., and 102 L heptanes was added, maintaining the same temperature. The solution was then filtered, rinsing with a further 13 L heptanes, then cooled to 32° C. to cause crystallization and maintained at that temperature for 1 hour. A further 66 L heptanes was added, and the mixture cooled to 22° C. and held for 1 hour, then cooled to −5° C. and held for another 1 hour. The mixture was then filtered to isolate the N-BOC-L-glutamic acid γ-benzyl α-ethyl ester (5), which was washed twice, each time with 25 L heptanes cooled to (−5˜0)° C., and dried under vacuum at 40° C., giving 39.3 Kg N-BOC-L-glutamic acid γ-benzyl α-ethyl ester (5).A 4000 L hydrogenator was purged with nitrogen, then under nitrogen sweep and no stirring loaded with 39.2 Kg N-BOC-L-glutamic acid γ-benzyl α-ethyl ester (5), 2.0 Kg 5% palladium on carbon, and 432 L ethyl acetate, and purged (3 bar) and decompressed (0.2 bar) twice with nitrogen and twice with hydrogen. Stirring was begun and the mixture heated to (37±2)° C., hydrogenated at that temperature under 2.8 bar hydrogen pressure until no further hydrogen absorption occurred, then held under 2.8 bar hydrogen pressure for 12 hours. Completion of hydrogenation was confirmed by thin-layer chromatography of a sample. The mixture was cooled to 28° C., the hydrogen purged from the hydrogenator, and the hydrogenator purged (2 bar) and decompressed (0.2 bar) twice with nitrogen. The mixture was filtered through a filter precoated with 10 Kg powdered cellulose in 200 L ethyl acetate, then the filter washed with the ethyl acetate used to form the precoat, giving a total of 626 Kg of a dilute ethyl acetate solution containing 29.5 Kg N-BOC-L-glutamic acid α-ethyl ester (6). This was distilled at (35˜40)° C. and (0.16˜0.18) bar to give 67 L of concentrated solution, then 29 L of ethyl acetate added and the solution redistilled to again give 67 L of concentrated solution.Example 3Preparation of Ezatiostat HydrochlorideThe concentrated solution of N-BOC-L-glutamic acid α-ethyl ester (6), 61.2 Kg (containing 27.8 Kg N-BOC-L-glutamic acid α-ethyl ester), was added to a 600 L jacketed glass-lined reactor, rinsed in with 5 L ethyl acetate, then cooled to 14° C. NMM, 10.8 Kg, was added over 50 minutes and rinsed in with 5 L ethyl acetate, then stirring stopped, giving an ethyl acetate solution of N-BOC-L-glutamic acid α-ethyl ester NMM salt. Ethyl acetate, 475 L, was added to a 1300 L cooled jacketed glass-lined reactor, followed by 14.5 Kg isobutyl chloroformate, rinsed in with 2×10 L ethyl acetate, and the mixture cooled to −11° C. The N-BOC-L-glutamic acid α-ethyl ester NMM salt solution was added to the 1300 L reactor over 1.3 hours, its reactor rinsed with 10 L ethyl acetate, and the rinse solution added to the 1300 L reactor, then stirred for an additional 30 minutes, while maintaining the temperature at about −13° C.S-benzyl-L-cysteinyl-D-phenylglycine ethyl ester hydrochloride (3) in ethyl acetate, 112 Kg (containing 41.3 Kg S-benzyl-L-cysteinyl-D-phenylglycine ethyl ester hydrochloride) was added in 4 portions over 45 minutes, and rinsed in with 5 L ethyl acetate, followed by 10.8 Kg NMM in 8 portions over 1.3 hours, rinsed in with 2×5 L portions of ethyl acetate, allowing the mixture to warm to −4° C. by the end of the addition. The mixture was gradually warmed to 30° C. over 2 hours, and maintained at (30˜35)° C. for 2 hours. The reaction mixture was washed twice with water: the first time adding 100 L water, heating to 41° C., allowing the phases to separate for 30 minutes, then removing the aqueous phase; the second time adding 100 L water, bringing the pH to 2.0 with the addition of 0.8 L 36% hydrochloric acid, stirring at 43° C. for 30 minutes, allowing the phases to separate for 1 hour, then removing the aqueous phase. The organic phase was then heated to 42° C., and the pressure reduced to about 0.25 bar until no further gas was released and solvents removed by distillation until 495 L of distillate had been removed. Four cycles of addition of 120 L ethyl acetate and removal of 120 L solvent by distillation were performed, and the water content of the remaining mixture was checked to ensure that it was below 0.1%. With the mixture at 42° C., 610 L of ethyl acetate was added, maintaining the temperature about 41° C., then heating to 58° C. to ensure dissolution. The solution was filtered, rinsing the filter with 18 L ethyl acetate, and the solution allowed to cool to 22° C. The nitrogen purge was stopped and 22.2 Kg hydrogen chloride gas was added through an immersion tube over 2 hours, then the mixture held at that temperature for 2 hours. The mixture was heated to 31° C. over 1.5 hours, and held at about that temperature for 15.5 hours. Solvents were removed by distillation at 33° C. and a pressure of about 0.13 bar over about 1.5 hours to give a volume of concentrated solution of about 630 L. Ethyl acetate, 100 L, was added, and the mixture cooled to 25° C. and held at that temperature for 30 minutes. The crude ezatiostat hydrochloride was recovered by filtration and washed with 30 L ethyl acetate, giving 113 Kg damp crude ezatiostat hydrochloride, which was dried at 40° C. under vacuum for 24 hours to give 52.8 Kg dry crude ezatiostat hydrochloride.Example 4Crystallization of Ezatiostat Hydrochloride to Form Pure Crystalline Ezatiostat Hydrochloride Ansolvate Form D61.5 Kg crude ezatiostat hydrochloride was added to a reactor at room temperature, followed by 399 liter (L) ethanol, and this mixture was heated to 68° C. to completely dissolve the ezatiostat hydrochloride, filtered, then allowed to cool to 65° C. and checked for clarity and the absence of crystallization. About 1.3 Kg of ezatiostat hydrochloride ansolvate form D was suspended in 9 L of ethyl acetate, and about one-half of this suspension was added to the ethanol solution. The mixture was cooled to 63° C. and the second half of the suspension added to the mixture. The resulting mixture was cooled gradually to 45° C., 928 L ethyl acetate was added, and the mixture was cooled to 26° C. and held at about that temperature for about 5 hours, then cooled to −2° C. The mixture, containing crystalline ezatiostat hydrochloride ansolvate, was filtered, and the residue washed twice with 65 L of chilled (0-5° C.) ethyl acetate. The crystalline ezatiostat hydrochloride ansolvate was dried at 30° C. for 48 hours, then cooled to room temperature and sieved. Analysis of the material by DSC and XRPD confirmed its identity as crystalline ezatiostat hydrochloride ansolvate, and Karl Fischer analysis showed a water content of 0.1%.Example 5Purifying Ezatiostat Hydrochloride Crystals to Form Pure Crystalline Ezatiostat Hydrochloride Ansolvate Form DCrude ezatiostat hydrochloride, 51.4 Kg, was added to a 600 L jacketed glass-lined reactor at room temperature, followed by 334 L of ethanol. The mixture was heated to 68° C. to completely dissolve the ezatiostat hydrochloride. The resulting solution was filtered into a 1300 L jacketed glass-lined reactor, and an additional 27 L ethanol warmed to 66° C. used to rinse the first reactor into the second reactor through the filter. The resulting solution in the second reactor was cooled to 63° C. and checked for complete dissolution; then 4 L of a seeding suspension of crystalline ezatiostat hydrochloride ansolvate in ethyl acetate was added, and the mixture cooled to 60° C. The remaining 4 L of the seeding suspension was added, and the mixture cooled to 47° C. over 2 hours. The solids in the mixture were shown by DSC to contain more than one form of ezatiostat hydrochloride, so the stages of heating to dissolution, cooling, and adding seeding suspension (this time 2×2 L), were repeated, then the mixture cooled to 41° C. This time the solids in the mixture were confirmed by DSC to be crystalline ezatiostat hydrochloride ansolvate. Ethyl acetate, 776 L, was added, and the mixture was cooled to 25° C. over 1.3 hours and further to 20° C. over an additional 5 hours, then cooled to −3° C. The mixture, containing crystalline ezatiostat hydrochloride ansolvate, was filtered and the solids washed twice with 54 L each of chilled (−5˜0)° C. ethyl acetate. The damp solids of crystalline ezatiostat hydrochloride ansolvate, 70 Kg, were dried in a vacuum oven at 25° C. for 16 hours, 35° C. for 7 hours, then at room temperature for 1 hour, then sieved. The crystalline ezatiostat hydrochloride ansolvate, 44.2 Kg, had a loss on drying at 40° C. under vacuum for 2 hours of 0.09%, and a water content by Karl Fischer analysis of 0.09%.

……………………………………………………………

U.S. Pat. No. 5,763,570

 https://www.google.com/patents/US5763570

……………………………..

http://www.google.com/patents/WO2011156025A1?cl=en

Example 1. Preparation Of Ezatiostat Hydrochloride Ansolvate By Slurrying

[0082] Ezatiostat hydrochloride monohydrate was added to methyl tert-butyl ether at room temperature in excess, so that undissolved solids were present. The mixture was then agitated in a sealed vial at room temperature for 4 days, and the solids were then isolated by suction filtration. XRPD analysis of the solids established that the isolated solids were ezatiostat hydrochloride ansolvate.

[0083] Ezatiostat hydrochloride monohydrate was added to hexanes at 60 °C in excess, so that undissolved solids were present. The mixture was then agitated in a sealed vial at 60 °C for 4 days, and the solids were then isolated by suction filtration. XRPD analysis of the solids established that the isolated solids were ezatiostat hydrochloride ansolvate.

Example 2. Preparation Of Crystalline Ezatiostat Hydrochloride Ansolvate By Heating

[0084] DSC of crystalline ezatiostat hydrochloride monohydrate showed the pattern in FIG. 1, as discussed in paragraph above. Hot stage microscopy showed an initial melt followed by a recrystallization at 153 °C and a final melt at 166 °C. VT-XRPD, where XRPD patterns were obtained at 28 °C, 90 °C, and 160 °C during heating, and 28 °C after cooling of the formerly heated material, showed the presence of ezatiostat hydrochloride monohydrate at 28 °C and 90 °C during heating and of crystalline ezatiostat hydrochloride ansolvate at 160 °C and 28 °C after cooling of the formerly heated material. This confirmed that the transition at around 153/156 °C was a conversion of ezatiostat hydrochloride monohydrate form A to crystalline ezatiostat hydrochloride ansolvate form D and that the final DSC endothermic peak at about 177 °C (166 °C in the hot stage microscopy) was due to the melting of crystalline ezatiostat hydrochloride ansolvate. This was further confirmed by XRPD of the TG-IR material, where XRPD patterns obtained at room temperature both before and after heating to about 160 °C showed that the material before heating was form A and that the material after heating was form D ansolvate. DSC of crystalline ezatiostat hydrochloride ansolvate prepared by recrystallization showed the pattern in FIG. 5, with only the endothermic peak at about

177 °C followed by a broad endotherm at about (205 – 215) °C. Accordingly, the presence of the DSC endothermic peak at about 177 °C, for example at (177±2) °C, when measured under the conditions described above, is considered characteristic of crystalline ezatiostat hydrochloride ansolvate, and the substantial absence of thermal events at temperatures below this is considered indicative of the absence of other forms of ezatiostat hydrochloride

………………………………..

http://www.google.com/patents/WO2013082462A1?cl=en

Ezatiostat, also known as TLK199 or TER 199, is a compound of the formula:

 

[0003] Ezatiostat has been shown to induce the differentiation of HL-60 promyelocytic leukemia cells in vitro, to potentiate the activity of cytotoxic agents both in vitro and in vivo, and to stimulate colony formation of all three lineages of hematopoietic progenitor cells in normal human peripheral blood. In preclinical testing, ezatiostat has been shown to increase white blood cell production in normal animals, as well as in animals in which white blood cells were depleted by treatment with cisplatin or fluorouracil. Similar effects may provide a new approach to treating myelodysplasia syndrome (MDS).

[0004] Many conditions, including MDS, a form of pre-leukemia in which the bone marrow produces insufficient levels of one or more of the three major blood elements (white blood cells, red blood cells, and platelets), are characterized by depleted bone marrow.

Myelosuppression, which is characterized by a reduction in blood cell levels and in a reduction of new blood cell generation in the bone marrow, is also a common, toxic effect of many standard chemotherapeutic drugs.

[0005] Ezatiostat hydrochloride is the hydrochloride acid addition salt of ezatiostat.

Ezatiostat hydrochloride in a liposomal injectable formulation was studied in a clinical trial for the treatment of MDS, and results from this trial, reported by Raza et al., J. Hem. One, 2:20 (published online 13 May 2009), demonstrated that administration of TLK199 was well tolerated and resulted in multi-lineage hematologic improvement. Ezatiostat hydrochloride in a tablet formulation has been evaluated in a clinical trial for the treatment of MDS, as reported by Raza et al, Blood, 113:6533-6540 (prepublished online 27 April 2009) and a single-patient report by Quddus et al, J. Hem. One, 3:16 (published online 23 April 2010), and is currently being evaluated in clinical trials for the treatment of MDS and for severe chronic idiopathic neutropenia.

…………………………………..

http://www.google.com/patents/WO2013082462A1?cl=en

Example 1

[0048] 80 mg of crystalline ezatiostat hydrochloride was placed in a round bottom flask and dissolved in 25 mL of methanol. The solvent was then evaporated on a rotary evaporation apparatus under reduced pressure at 30 °C. After 30 minutes, the solid sample was removed from the round bottom flask and stored in a sealed vial at 2 °C in a refrigerator. Analysis of this sample was carried out within 24 hours of removing it from the rotary evaporation apparatus.

[0049] The resulting amorphous material was analyzed by 1H NMR, 13C NMR, DSC, and X-Ray powder diffraction experiments. The DSC conditions were 30 to 300 °C at 10 °C/ min using 7 mg of the amorphous material. The X-Ray powder diffraction was taken at 0-60 of 2theta. The crystalline ezatiostat hydrochloride was also analyzed.

Phase II trials: Ezatiostat is the first GSTP1-1 inhibitor shown to cause clinically significant and sustained reduction in RBC transfusions, transfusion independence, and multilineage responses in MDS patients. The tolerability and activity profile of ezatiostat may offer a new treatment option for patients with MDS. (source:  Cancer. 2012 Apr 15;118(8):2138-47.)

 

References

1: Galili N, Tamayo P, Botvinnik OB, Mesirov JP, Brooks MR, Brown G, Raza A. Prediction of response to therapy with ezatiostat in lower risk myelodysplastic syndrome. J Hematol Oncol. 2012 May 6;5:20. doi: 10.1186/1756-8722-5-20. PubMed PMID: 22559819; PubMed Central PMCID: PMC3407785.

2: Raza A, Galili N, Mulford D, Smith SE, Brown GL, Steensma DP, Lyons RM, Boccia R, Sekeres MA, Garcia-Manero G, Mesa RA. Phase 1 dose-ranging study of ezatiostat hydrochloride in combination with lenalidomide in patients with non-deletion (5q) low to intermediate-1 risk myelodysplastic syndrome (MDS). J Hematol Oncol. 2012 Apr 30;5:18. doi: 10.1186/1756-8722-5-18. PubMed PMID: 22546242; PubMed Central PMCID: PMC3416694.

3: Lyons RM, Wilks ST, Young S, Brown GL. Oral ezatiostat HCl (Telintra®, TLK199) and idiopathic chronic neutropenia (ICN): a case report of complete response of a patient with G-CSF resistant ICN following treatment with ezatiostat, a glutathione S-transferase P1-1 (GSTP1-1) inhibitor. J Hematol Oncol. 2011 Nov 2;4:43. doi: 10.1186/1756-8722-4-43. PubMed PMID: 22047626; PubMed Central PMCID: PMC3235963.

4: Raza A, Galili N, Smith SE, Godwin J, Boccia RV, Myint H, Mahadevan D, Mulford D, Rarick M, Brown GL, Schaar D, Faderl S, Komrokji RS, List AF, Sekeres M. A phase 2 randomized multicenter study of 2 extended dosing schedules of oral ezatiostat in low to intermediate-1 risk myelodysplastic syndrome. Cancer. 2012 Apr 15;118(8):2138-47. doi: 10.1002/cncr.26469. Epub 2011 Sep 1. PubMed PMID: 21887679.

5: Quddus F, Clima J, Seedham H, Sajjad G, Galili N, Raza A. Oral Ezatiostat HCl (TLK199) and Myelodysplastic syndrome: a case report of sustained hematologic response following an abbreviated exposure. J Hematol Oncol. 2010 Apr 23;3:16. doi: 10.1186/1756-8722-3-16. PubMed PMID: 20416051; PubMed Central PMCID: PMC2873355.

6: Steensma DP. Novel therapies for myelodysplastic syndromes. Hematol Oncol Clin North Am. 2010 Apr;24(2):423-41. doi: 10.1016/j.hoc.2010.02.010. Review. PubMed PMID: 20359635.

7: D’Alò F, Greco M, Criscuolo M, Voso MT. New treatments for myelodysplastic syndromes. Mediterr J Hematol Infect Dis. 2010 Aug 11;2(2):e2010021. doi: 10.4084/MJHID.2010.021. PubMed PMID: 21415972; PubMed Central PMCID: PMC3033133.

8: Raza A, Galili N, Callander N, Ochoa L, Piro L, Emanuel P, Williams S, Burris H 3rd, Faderl S, Estrov Z, Curtin P, Larson RA, Keck JG, Jones M, Meng L, Brown GL. Phase 1-2a multicenter dose-escalation study of ezatiostat hydrochloride liposomes for injection (Telintra, TLK199), a novel glutathione analog prodrug in patients with myelodysplastic syndrome. J Hematol Oncol. 2009 May 13;2:20. doi: 10.1186/1756-8722-2-20. PubMed PMID: 19439093; PubMed Central PMCID: PMC2694211.

9: Raza A, Galili N, Smith S, Godwin J, Lancet J, Melchert M, Jones M, Keck JG, Meng L, Brown GL, List A. Phase 1 multicenter dose-escalation study of ezatiostat hydrochloride (TLK199 tablets), a novel glutathione analog prodrug, in patients with myelodysplastic syndrome. Blood. 2009 Jun 25;113(26):6533-40. doi: 10.1182/blood-2009-01-176032. Epub 2009 Apr 27. PubMed PMID: 19398716.

Author Unknown, “Dose-Ranging Study of Telintra® Tablets + Revlimid® in Patients with Non-Deletion (5q) Low to Intermediate-1 Risk Myelodysplastic Syndrome (MDS)“, Clinical Trials, 2010, Retrieved from the Internet: URL:http://clinicaltrials.gov/ct2/show/NCT01062152?term=ezatiostat&rank=2.
2 Author Unknown, “Phase 2 Study Comparing Two Dose Schedules of Telintra(TM) in Myelodysplastic Syndrome (MDS)“, 2008, Retrieved from the Internet: URL: http://clinicaltrials.gov/ct2/show/NCT00700206?term=ezatiostat&rank=3.
3 Author Unknown, “Telik initiates phase I trial of ezatiostat in patients with myelodysplastic syndrome“, Thomson Reuters Integrity, 2010, Retrieved from the Internet: URL:https://integrity.thomson-pharma.com/integrity/xmlxsl/pk-ref-list.xml-show-ficha-ref? p-re-id=1444034.
4 Author Unknown, “Telik initiates Telintra Phase 2 trial in Revlimid refractory or resistant, del 5q MDS“, 2011, Abstract, retrieved from Internet: URL:http://www.new-medical.net/new/20110608/Telik-initiates-Telintra-Phase-2-trial-in-Revlimid-refractory-or-resistant-del-5q-MDS.aspx.
5 Author Unknown, “Telik reports phase II data on ezatiostat in MDS“, Thomson Reuters Integrity, 2010, Retrieved from the Internet: URL:https://integrity.thomson-pharma.com/integrity/xmlxsl/pk-ref -list.xml-show-ficha-ref?p-ref-id=1513842.
6 Author Unknown, “Phase 2 Study Comparing Two Dose Schedules of Telintra™ in Myelodysplastic Syndrome (MDS)“, 2008, Retrieved from the Internet: URL: http://clinicaltrials.gov/ct2/show/NCT00700206?term=ezatiostat&rank=3.
7 Author Unknown, “Telik initiates phase I trial of ezatiostat in patients with myelodysplastic syndrome“, Thomson Reuters Integrity, 2010, Retrieved from the Internet: URL:https://integrity.thomson-pharma.com/integrity/xmlxsl/pk—ref—list.xml—show—ficha—ref? p—re—id=1444034.
8 Author Unknown, “Telik reports phase II data on ezatiostat in MDS“, Thomson Reuters Integrity, 2010, Retrieved from the Internet: URL:https://integrity.thomson-pharma.com/integrity/xmlxsl/pk—ref —list.xml—show—ficha—ref?p—ref—id=1513842.
9 * Beckmann (Eng. Life Sci. 2003, 3, 113-120).
10 International Search Report dated Apr. 19, 2012 for PCT/US2011/030376 filed Mar. 29, 2011.
11 Kibbe, A. Croscarmellose Sodium. Handbook for Pharmaceutical Excipients, American Pharmaceutical Association, Third Edition, 2000, pp. 160-162.
12 Lyttle et al. “Isozyme-specific Glutathione-S-Transferase Inhibitors: Design and Synthesis,” Journal of Medicinal Chemistry, American Chemical Society, 1994, 37:189-194.
13 Quddus et al. “Oral Ezatiostat HCI (TLK199) and Myelodysplastic syndrome: A case report of sustained hematologic response following an abbreviated exposure“, Journal of Hematology & Oncology, 2010, 3:16.
14 Raza et al. “Multilineage Hematologic Improvement (HI) by TLK199 (TELINTRA(TM)), A Novel Glutathione Analog, in Myelodysplastic Syndrome: Phase 2 Study Results.” Poster Presentation, 2005, American Society of Hematology.
15 Raza et al. “Phase 1 Dose Escalation Study of TLK199 Tablets (Ezatiostat HCI, TELINTRA®), a Novel Glutathione Analog, in Myelodysplastic Syndrome.” Poster Presentation, 2007, American Society of Hematology.
16 Raza et al. “Phase 1 Dose Escalation Study of TLK199 Tablets (Telintra), a Novel Glutathione Analog, in Myelodysplastic Syndrome,” Abstract #1454 appears in Blood, vol. 100, issue 11, Nov. 16, 2007.
17 Raza et al. “Phase 1 multicenter dose-escalation study of ezatiostat hydrochloride (TLKI99 tablets), a novel glutathione analog prodrug, in patients with myelodysplastic syndrome“, BLOOD, 2009, 113(26):6533-6540.
18 Raza et al. “Phase 1-2a multicenter dose-escalation study of ezatiostat hydrochloride liposomes for injection (Telintra®, TLKI99), a novel glutathione analog prodrug in patients with myelodysplastic syndrome,” Journal of Hematology & Oncology, 2009, 2:20.
19 Raza et al. “Phase 2 Randomized Multicenter Study of Extended Dosing Schedules of Oral Ezatiostat HCI (Telintra), a Glutathione Analog Prodrug GSTP1-1 Inhibitor, In Low to Intermediate-1 Risk Myelodysplastic Syndrome (MDS)“, Myelodysplastic Syndromes: Poster II, Abstract 2910, Blood 2010; 116:2910a.
20 Raza et al. “Multilineage Hematologic Improvement (HI) by TLK199 (TELINTRA™), A Novel Glutathione Analog, in Myelodysplastic Syndrome: Phase 2 Study Results.” Poster Presentation, 2005, American Society of Hematology.
21 Rowe, et al. Hypromellose. Handbook of Pharmaceutical Excipients, Pharmaceutical Press, 2003, pp. 297-300.
22 Rowe, et al. Mannitol. Handbook of Pharmaceutical Excipients, Pharmaceutical Press, 2009, pp. 1-3.
23 Rowe, et al. Sucrose; Magnesium Stearate. Handbook of Pharmaceutical Excipients, Pharmaceutical Press, 2006, 744, pp. 430-431.
24 U.S. Appl. No. 13/041,136, filed Mar. 4, 2011, Parent et al.
25 U.S. Appl. No. 13/094,693, filed Apr. 26, 2011, Leclerc et al.
26 U.S. Appl. No. 13/108,752, filed May 16, 2011, Brown et al.
27 U.S. Appl. No. 13/108,754, filed May 16, 2011, Brown et al.
28 U.S. Appl. No. 13/108,756, filed May 16, 2011, Brown et al.
29 Yoshioka et al . “Crystalline State and Polymorphism in Solid Drugs,” In: “Stability of drugs and dosage forms,” Kluwer Academic, 2000, ISBN: 0-306-46404-7, Chaper 2.2.11, pp. 107-108.

 

WO2013082462A1 * Nov 30, 2012 Jun 6, 2013 Telik, Inc. Amorphous ezatiostat ansolvate
US20120251496 * Mar 20, 2012 Oct 4, 2012 Telik, Inc. Ezatiostat for treating multiple myeloma

……….

Full-size image (11 K)

Figure 1.

Known Yes1 kinase inhibitors, dasatinib and saracatinib.

 

Table 1. Select Yes1 kinase inhibitors from a HTS and their corresponding clinical phase, known targets, and IC50 values
Compound name and NCGC ID Structure Clinical phase Known targets Yes1 IC50 (nM)
Dasatinib (1)
NCGC00181129
Approved Lyn, PDGFR, KIT, Lck, BTK, Bcr–Abl,
Fyn, Yes1, c-Src
0.5 (<1.0)a
Saracatinib (2)
NCGC00241099
Phase II/III c-Src, Bcr–Abl, Yes1, Lck 6.2 (0.70)a
AEE-788 (3)
NCGC00263149
Phase I/II EGFR, HER-2, VEGFR-2 17.5 (13.1)a
Dovitinib (4)
NCGC00249685
Phase III FGFR, EGFR, PDGFR, VEGFR-1,2 31 (1.4)a
DCC-2036 (5)
NCGC00263172
Phase I/II Bcr–Abl, Tie-2, Lyn, FLT3, VEGFR-2 2.5 (1.5)a
SGI-1776 (6)
NCGC00263186
Discontinued Pim-1, FLT3 2670 (240)a
AMG-Tie-2-1 (7)
NCGC00263199
Preclinical Tie-2 8.7 (22.0)a
AZ-23 (8)
NCGC00250381
Preclinical Trk 39.1 (3.0)a
Dorsomorphin (9)
NCGC00165869
Preclinical AMPK, BMPR, TGFβ Receptor 195.9 (29.8)a
AZ-628 (10)
NCGC00250380
Preclinical Raf Kinase B,C 348.3 (51.2)a
http://www.sciencedirect.com/science/article/pii/S0960894X13006677
Data in parentheses were gathered by Reaction Biology Corp. using a [γ-33P]-ATP radiolabeled enzyme activity assay at an ATP concentration of 10 μM (www.reactionbiology.com).

 

 

 


Filed under: Phase3 drugs, Uncategorized Tagged: Ezatiostat, TELINTRA®, TLK-199, TLK199

Qingqi Chen’s Book……”Anticancer Drug Research Guide.” New drugs in development for cancers

Dovitinib in phase 3 for Cancer, bladder (urothelial carcinoma)

$
0
0

 

Dovitinib
(3E)-4-amino-5-fluoro-3-[5-(4-methylpiperazin-1-yl)-1,3-dihydrobenzimidazol-2-ylidene]quinolin-2-one, is one kind of white crystalline powder, odorless, little bitter taste.

4-Amino-5-fluoro-3-[6-(4-methyl-1-piperazinyl)-1H-benzimidazol-2-yl]-2(1H)-quinolinone 2-hydroxypropanoate

4-Amino-5-fluoro-3-[6-(4-methyl-1-piperazinyl)-1H-benzimidazol-2-yl]-2(1H)-quinolinone 2-hydroxypropanoate hydrate (1:1:1);

CAS No. 405169-16-6 (free base), 915769-50-5,  804551-71-1 of lactate
 TKI-258; CHIR-258.
Formula C21H21FN6O.C3H6O3.H2O
Molecular Weight 500.53

for treatment of cancer

Novartis Ag, innovator

Dovitinib lactate is the orally bioavailable lactate salt of a benzimidazole-quinolinone compound with potential antineoplastic activity. Dovitinib strongly binds to fibroblast growth factor receptor 3 (FGFR3) and inhibits its phosphorylation, which may result in the inhibition of tumor cell proliferation and the induction of tumor cell death. In addition, this agent may inhibit other members of the RTK superfamily, including the vascular endothelial growth factor receptor; fibroblast growth factor receptor 1; platelet-derived growth factor receptor type 3; FMS-like tyrosine kinase 3; stem cell factor receptor (c-KIT); and colony-stimulating factor receptor 1; this may result in an additional reduction in cellular proliferation and angiogenesis, and the induction of tumor cell apoptosis. The activation of FGFR3 is associated with cell proliferation and survival in certain cancer cell types

Dovitinib (TKI258) is a highly potent, novel multitargeted growth factor receptor kinase inhibitor with IC50 of 1, 2, 10, 8, 27, 36 nM for FLT3, c-KIT, VEGFR1/2/3, PDGFRß and CSF-1R, respectively. It shows both antitumor and antiangiogenic activities in vivo. [1] It potently inhibits FGFR3 with an inhibitory concentration of 50% (IC50) of 5 nM in in vitro kinase assays and selectively inhibited the growth of B9 cells and human myeloma cell lines expressing wild-type (WT) or activated mutant FGFR3. Antiproliferative activity of Dovitinib (TKI258) against MV4;11 was ~24-fold greater compared with RS4;11, indicating more potent inhibition against cells with constitutively activated FLT3 ITD. [2][3]

References on Dovitinib (TKI258)
  • [1] Clin Cancer Res 2005;11:3633-3641
  • [2] Blood 2005;105: 2941-2948
  • [3] Clin Cancer Res 2005;11:5281-5291

Dovitinib lactate is an angiogenesis inhibitor in phase III clinical trials at Novartis for the treatment of refractory advanced/metastatic renal cell cancer. Early clinical trials are also under way at the company for the oral treatment of several types of solid tumors, multiple myeloma and glioblastoma multiforme. Phase II trials are ongoing for the treatment of castration-resistant prostate cancer and for the treatment of Von-Hippel Lindau disease, for the treatment of non-small cell lung cancer (NSCLC) and for the treatment of colorectal cancer. Novartis and Seoul National University Hospital are conducting phase II clinical studies for the treatment of adenoid cystic carcinoma. Additional phase II clinical trials are ongoing at Asan Medical Center for the treatment of metastatic or advanced gastrointestinal stromal tumors (GIST). The University of Pennsylvania is conducting phase II clinical trials for the treatment of advanced malignant pheochromocytoma or paraganglioma. Phase II clinical studies are ongoing by Novartis for the treatment of advanced malignant pleural mesothelioma which has progressed following prior platinum-antifolate chemotherapy (DOVE-M) and for the oral treatment of hepatocellular carcinoma.

In 2009, Novartis discontinued development of dovitinib lactate for the treatment of acute myeloid leukemia (AML) based on the observation of time dependent drug accumulation. A phase I trial was also stopped for the same reason.

The drug candidate has been shown to inhibit multiple growth factor tyrosine kinases, including vascular endothelial growth factor receptor (VEGFR) tyrosine kinases VEGFR1 and VEGFR2, fibroblast growth factor receptor (FGFR) and platelet-derived growth factor receptor (PDGFR) tyrosine kinases. In previous studies, the benzimidazole-quinoline inhibited VEGF-mediated human microvascular endothelial cell (HMVEC) proliferation and demonstrated concentration-dependent antiangiogenic activity in in vitro assays, as well as potent antiproliferative activity against a subset of cancer cell lines.

In 2013, an orphan drug designation was assigned in the U.S. for the treatment of adenoid cystic carcinoma.

“Molecularly Targeted Agents for Renal Cell Carcinoma: The Next Generation”, C. Lance Cowey and Thomas E. Hutson -Clinical Advances in Hematology & Oncology, 2010, 8, 357.

 

Lee S. H.; Lopes de Menezes, D. Vora, J. Harris, A.; Ye, H. Nordahl, L.; Garrett, E.; Samara, E.; Aukerman, S. L.; Gelb, A. B. Heise, C. In Vivo Target Modulation and Biological Activity of CHIR-258, a Multitargeted Growth Factor Receptor Kinase Inhibitor, in Colon Cancer Models. Clin. Cancer Res. 2005, 11 (10), 3633–3641.
Lopes de Menezes, D. E.; Peng, J.; Garrett, E. N.; Louie, S. G.; Lee, S. H.; Wiesmann, M.; Tang, Y.; Shephard, L.; Goldbeck, C.; Oei, Y.; Ye, H.; Aukerman, S. L.; Heise, C. CHIR-258: A Potent Inhibitor of FLT3 Kinase in Experimental Tumor Xenograft Models of Human Acute Myelogenous Leukemia. Clin. Cancer Res. 2005, 11 (14), 5281–5291.
Trudel, S.; Li, Z. H.; Wei, E.; Wiesmann, M.; Chang, H.; Chen, C.; Reece, D.; Heise, C.; Stewart, A. K. CHIR-258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t(4;14) multiple myeloma. Blood 2005, 105 (7), 2941–2948.

Synthesis of Dovitinib

Tetrahedron Letters 47 (2006) 657–660
LHMDS mediated tandem acylation–cyclization of 2-aminobenzenecarbonitriles with 2-benzymidazol-2-yl acetates: a short and efficient route to the synthesis of 4-amino-3-benzimidazol-2-ylhydroquinolin-2-ones
William R. Antonios-McCrea, Kelly A. Frazier, Elisa M. Jazan, Timothy D. Machajewski, Christopher M. McBride, Sabina Pecchi, Paul A. Renhowe, Cynthia M. Shafer and Clarke Taylor

 

 

 

cas 852433-84-2

 

…………………………..

 

 

852433-84-2

 

…………………………..

WO 2002022598 or https://www.google.com/patents/EP1317442A1?cl=en

Figure imgf000090_0001

Figure imgf000090_0002

………………………………

WO 2003087095 or http://www.google.fm/patents/US20030028018?cl=un

…………………..

WO 2005046589 or http://www.google.com/patents/EP1692085A2?cl=en

lactate salt of the compound of

Structure I or the tautomer thereof is administered to the subject and/or is used to prepare the medicament. [0062] In some embodiments, the compound of Structure I has the following formula

Figure imgf000032_0001

……………………

WO 2006125130

http://www.google.com/patents/WO2006125130A1?cl=en

formula IHB

Figure imgf000013_0001

HIB

Scheme 1

 

Figure imgf000056_0001

 

Figure imgf000056_0002

 

Figure imgf000056_0003

Example 4

Synthesis of 4-Amino-5-fluoro-3-[6-(4-rnethyl-piperazin-1 -yl)-1 H-benzimidazol- 2-yl]-1 H-quinolin-2-one Procedure A

Figure imgf000068_0001

[0149] [6-(4-Methyl-piperazin-1-yl)-1 H-benzimidazol-2-yl]-acetic acid ethyl ester (250 g, 820 mmol) (dried with ethanol as described above) was dissolved in THF (3800 ml_) in a 5000 ml_ flask fitted with a condenser, mechanical stirrer, temperature probe, and purged with argon. 2-Amino-6-fluoro-benzonitrile (95.3 g, 700 mmol) was added to the solution, and the internal temperature was raised to 40°C. When all the solids had dissolved and the solution temperature had reached 4O0C, solid KHMDS (376.2 g, 1890 mmol) was added over a period of 5 minutes. When addition of the potassium base was complete, a heterogeneous yellow solution was obtained, and the internal temperature had risen to 62°C. After a period of 60 minutes, the internal temperature decreased back to 40°C, and the reaction was determined to be complete by HPLC (no starting material or uncyclized intermediate was present). The thick reaction mixture was then quenched by pouring it into H2O (6000 ml_) and stirring the resulting mixture until it had reached room temperature. The mixture was then filtered, and the filter pad was washed with water (1000 ml_ 2X). The bright yellow solid was placed in a drying tray and dried in a vacuum oven at 50°C overnight providing 155.3 g (47.9%) of the desired 4-amino-5-fluoro-3-[6-(4-methyl-piperazin-1-yl)-1 H-benzimidazol-2- yl]-1 H-quinolin-2-one.

Procedure B

[0150] A 5000 mL 4-neck jacketed flask was equipped with a distillation apparatus, a temperature probe, a N2 gas inlet, an addition funnel, and a mechanical stirrer. [6-(4-Methyl-piperazin-1-yl)-1 H-benzimidazol-2-yl]-acetic acid ethyl ester (173.0 g, 570 mmol) was charged into the reactor, and the reactor was purged with N2 for 15 minutes. Dry THF (2600 mL) was then charged into the flask with stirring. After all the solid had dissolved, solvent was removed by distillation (vacuum or atmospheric (the higher temperature helps to remove the water) using heat as necessary. After 1000 mL of solvent had been removed, distillation was stopped and the reaction was purged with N2. 1000 mL of dry THF was then added to the reaction vessel, and when all solid was dissolved, distillation (vacuum or atmospheric) was again conducted until another 1000 mL of solvent had been removed. This process of adding dry THF and solvent removal was repeated at least 4 times (on the 4thdistillation, 60% of the solvent is removed instead of just 40% as in the first 3 distillations) after which a 1 mL sample was removed for Karl Fischer analysis to determine water content. If the analysis showed that the sample contained less than 0.20% water, then reaction was continued as described in the next paragraph. However, if the analysis showed more than 0.20% water, then the drying process described above was continued until a water content of less than 0.20% was achieved.

[0151] After a water content of less than or about 0.20% was achieved using the procedure described in the previous paragraph, the distillation apparatus was replaced with a reflux condenser, and the reaction was charged with 2-amino-6- fluoro-benzonitrile (66.2 g, 470 mmol)( in some procedures 0.95 equivalents is used). The reaction was then heated to an internal temperature of 38-420C. When the internal temperature had reached 38-420C, KHMDS solution (1313 g, 1.32 mol, 20% KHMDS in THF) was added to the reaction via the addition funnel over a period of 5 minutes maintaining the internal temperature at about 38-50°C during the addition. When addition of the potassium base was complete, the reaction was stirred for 3.5 to 4.5 hours (in some examples it was stirred for 30 to 60 minutes and the reaction may be complete within that time) while maintaining the internal temperature at from 38-420C. A sample of the reaction was then removed and analyzed by HPLC. If the reaction was not complete, additional KHMDS solution was added to the flask over a period of 5 minutes and the reaction was stirred at 38-420C for 45-60 minutes (the amount of KHMDS solution added was determined by the following: If the IPC ratio is < 3.50, then 125 ml_ was added; if 10.0 >IPC ratio >3.50, then 56 mL was added; if 20.0 ≥IPC ratio >10, then 30 mL was added. The IPC ratio is equal to the area corresponding to 4-amino-5-fluoro-3-[6- (4-methyl-piperazin-1 -yl)-1 H-benzimidazol-2-yl]-1 H-quinolin-2-one) divided by the area corresponding to the uncyclized intermediate). Once the reaction was complete (IPC ratio > 20), the reactor was cooled to an internal temperature of 25- 300C, and water (350 mL) was charged into the reactor over a period of 15 minutes while maintaining the internal temperature at 25-35°C (in one alternative, the reaction is conducted at 400C and water is added within 5 minutes. The quicker quench reduces the amount of impurity that forms over time). The reflux condenser was then replaced with a distillation apparatus and solvent was removed by distillation (vacuum or atmospheric) using heat as required. After 1500 mL of solvent had been removed, distillation was discontinued and the reaction was purged with N2. Water (1660 mL) was then added to the reaction flask while maintaining the internal temperature at 20-300C. The reaction mixture was then stirred at 20-300C for 30 minutes before cooling it to an internal temperature of 5- 100C and then stirring for 1 hour. The resulting suspension was filtered, and the flask and filter cake were washed with water (3 x 650 mL). The solid thus obtained was dried to a constant weight under vacuum at 5O0C in a vacuum oven to provide 103.9 g (42.6% yield) of 4-amino-5-fluoro-3-[6-(4-methyl-piperazin-1-yl)-1H- benzimidazol-2-yl]-1H-quinolin-2-one as a yellow powder.

Procedure C

Figure imgf000071_0001

[0152] [6-(4-Methyl-piperazin-1-yl)-1 H-benzimidazol-2-yl]-acetic acid ethyl ester (608 g, 2.01 mol) (dried) and 2-amino-6-fluoro-benzonitrile (274 g, 2.01 mol) were charged into a 4-neck 12 L flask seated on a heating mantle and fitted with a condenser, mechanical stirrer, gas inlet, and temperature probe. The reaction vessel was purged with N2, and toluene (7.7 L) was charged into the reaction mixture while it was stirred. The reaction vessel was again purged with N2 and maintained under N2. The internal temperature of the mixture was raised until a temperature of 630C (+/- 3°C) was achieved. The internal temperature of the mixture was maintained at 63°C (+/- 30C) while approximately 2.6 L of toluene was distilled from the flask under reduced pressure (380 +/- 10 torr, distilling head t = 40°C (+/- 1O0C) (Karl Fischer analysis was used to check the water content in the mixture. If the water content was greater than 0.03%, then another 2.6 L of toluene was added and distillation was repeated. This process was repeated until a water content of less than 0.03% was achieved). After a water content of less than 0.03% was reached, heating was discontinued, and the reaction was cooled under N2 to an internal temperature of 17-19°C. Potassium t-butoxide in THF (20% in THF; 3.39 kg, 6.04 moles potassium t-butoxide) was then added to the reaction under N2 at a rate such that the internal temperature of the reaction was kept below 20°C. After addition of the potassium t-butoxide was complete, the reaction was stirred at an internal temperature of less than 2O0C for 30 minutes. The temperature was then raised to 25°C, and the reaction was stirred for at least 1 hour. The temperature was then raised to 30°C, and the reaction was stirred for at least 30 minutes. The reaction was then monitored for completion using HPLC to check for consumption of the starting materials (typically in 2-3 hours, both starting materials were consumed (less than 0.5% by area % HPLC)). If the reaction was not complete after 2 hours, another 0.05 equivalents of potassium t-butoxide was added at a time, and the process was completed until HPLC showed that the reaction was complete. After the reaction was complete, 650 mL of water was added to the stirred reaction mixture. The reaction was then warmed to an internal temperature of 50°C and the THF was distilled away (about 3 L by volume) under reduced pressure from the reaction mixture. Water (2.6 L) was then added drop wise to the reaction mixture using an addition funnel. The mixture was then cooled to room temperature and stirred for at least 1 hour. The mixture was then filtered, and the filter cake was washed with water (1.2 L), with 70% ethanol (1.2 L), and with 95% ethanol (1.2 L). The bright yellow solid was placed in a drying tray and dried in a vacuum oven at 50°C until a constant weight was obtained providing 674 g (85.4%) of the desired 4-amino-5-fluoro-3-[6-(4-methyl-piperazin-1-yl)-1 H- benzimidazol-2-yl]-1 H-quinolin-2-one.

Preparation of Lactic Acid salt of 4-Amino-5-fluoro-3-[6-(4-methyl-piperazin-1- yl)-1 H-benzimidazol-2-yl]-1 H-quinolin-2-one

Figure imgf000073_0001

D,L-Lactic Acid

Figure imgf000073_0002

[0154] A 3000 ml_ 4-necked jacketed flask was fitted with a condenser, a temperature probe, a N2 gas inlet, and a mechanical stirrer. The reaction vessel was purged with N2 for at least 15 minutes and then charged with 4-amino-5-fluoro- 3-[6-(4-methyl-piperazin-1-yl)-1 H-benzimidazol-2-yl]-1 H-quinoiin-2-one (484 g, 1.23 mol). A solution of D,L-Lactic acid (243.3 g, 1.72 mol of monomer-see the following paragraph), water (339 mL), and ethanol (1211 mL) was prepared and then charged to the reaction flask. Stirring was initiated at a medium rate, and the reaction was heated to an internal temperature of 68-720C. The internal temperature of the reaction was maintained at 68-72°C for 15-45 minutes and then heating was discontinued. The resulting mixture was filtered through a 10-20 micron frit collecting the filtrate in a 12 L flask. The 12 L flask was equipped with an internal temperature probe, a reflux condenser, an addition funnel, a gas inlet an outlet, and an overhead stirrer. The filtrate was then stirred at a medium rate and heated to reflux (internal temperature of about 780C). While maintaining a gentle reflux, ethanol (3,596 mL) was charged to the flask over a period of about 20 minutes. The reaction flask was then cooled to an internal temperature ranging from about 64-700C within 15-25 minutes and this temperature was maintained for a period of about 30 minutes. The reactor was inspected for crystals. If no crystals were present, then crystals of the lactic acid salt of 4-amino-5-fluoro-3-[6-(4-methyl- piperazin-1-yl)-1 H-benzimidazol-2-yl]-1 H-quinolin-2-one (484 mg, 0.1 mole %) were added to the flask, and the reaction was stirred at 64-7O0C for 30 minutes before again inspecting the flask for crystals.

[0155] Once crystals were present, stirring was reduced to a low rate and the reaction was stirred at 64-700C for an additional 90 minutes. The reaction was then cooled to about 00C over a period of about 2 hours, and the resulting mixture was filtered through a 25-50 micron fritted filter. The reactor was washed with ethanol (484 ml_) and stirred until the internal temperature was about 00C. The cold ethanol was used to wash the filter cake, and this procedure was repeated 2 more times. The collected solid was dried to a constant weight at 500C under vacuum in a vacuum oven yielding 510.7 g (85.7%) of the crystalline yellow lactic acid salt of 4-amino-5-fluoro-3-[6-(4-methyl-piperazin-1 -yl)-1 H-benzimidazol-2-yl]-1 H-quinolin- 2-one. A rubber dam or inert conditions were typically used during the filtration process. While the dry solid did not appear to be very hygroscopic, the wet filter cake tends to pick up water and become sticky. Precautions were taken to avoid prolonged exposure of the wet filter cake to the atmosphere.

[0156] Commercial lactic acid generally contains about 8-12% w/w water, and contains dimers and trimers in addition to the monomeric lactic acid. The mole ratio of lactic acid dimer to monomer is generally about 1.0:4.7. Commercial grade lactic acid may be used in the process described in the preceding paragraph as the monolactate salt preferentially precipitates from the reaction mixture.

[0157] It should be understood that the organic compounds according to the invention may exhibit the phenomenon of tautomerism. As the chemical structures within this specification can only represent one of the possible tautomeric forms at a time, it should be understood that the invention encompasses any tautomeric form of the drawn structure. For example, the compound having the formula NIB is shown below with one tautomer, Tautomer INBa:

Figure imgf000075_0001

HIB

Figure imgf000075_0002

Tautomer HIBa

Other tautomers of the compound having the formula NIB, Tautomer INlBb and Tautomer IHBc, are shown below:

Figure imgf000075_0003

Tautomer IIIBb

Figure imgf000075_0004

Tautomer IIIBc

……………………….

WO 2006127926

……………………..

Design, structure-activity relationships and in vivo characterization of 4-amino-3-benzimidazol-2-ylhydroquinolin-2-ones: A novel class of receptor tyrosine kinase inhibitors
J Med Chem 2009, 52(2): 278

………………………………

WO 2003087095

…………………..

WO 2005046589

……………………

WO 2006125130

 

……………………….

WO 2006127926

……………………..

Design, structure-activity relationships and in vivo characterization of 4-amino-3-benzimidazol-2-ylhydroquinolin-2-ones: A novel class of receptor tyrosine kinase inhibitors
J Med Chem 2009, 52(2): 278


Filed under: 0rphan drug status, cancer, Phase3 drugs, Uncategorized Tagged: Dovitinib

Using molecular techniques, researchers improved diagnosis and treatment of cancer

$
0
0

Originally posted on lyranara.me:

Using molecular techniques, researchers improved diagnosis and treatment of cancer

The ABC Medical Center, located in Mexico City, implemented various molecular diagnostic methods that can detect the genetic alterations in several types of cancer, so they can select a personalized therapy for each patient and direct it against the mutated genes that cause disease.

For this the Laboratory of Molecular Pathology was created, thanks to a cancer patient who decided to make a donation to the hospital to acquire the necessary equipment, sensitive to the existing need in Mexico for a place where timely diagnosis of the disease is made.

“While the techniques of molecular biology have been applied to clinical diagnosis for over 20 years, they had just been established in the country. It is estimated that only about five years ago in some of the National Institutes of Health and universities, where it was first used in the field of research, “says Dr. César Lara Torres, head of…

View original 419 more words


Filed under: Uncategorized

Taiho’s Colon Cancer Drug Ups OS in Phase 3

$
0
0

TAS-102 (nonproprietary names: trifluridine and tipiracil hydrochloride)

Taiho’s Colon Cancer Drug Ups OS in Phase 3

 

Taiho Pharmaceutical Co. Ltd. announced results from its global Phase 3 RECOURSE trial on its oral combination anticancer drug TAS-102 in refractory metastatic colorectal cancer (mCRC). Read more…

FULL STORY

http://www.dddmag.com/news/2014/05/taihos-colon-cancer-drug-ups-os-phase-3?et_cid=3937577&et_rid=523035093&type=cta

TAS-102 is an anti-cancer drug under development for colorectal cancer.[1]

Clinical trials

A phase II trial reported in 2011[2] and a phase III trial is due to end in 2014.[1][3]

Mechanism

TAS-102 consists of the cytotoxin trifluridine and the thymidine phosphorylase inhibitor (TPI) tipiracil.[4] Trifluridine is incorporated into DNA during DNA synthesis and inhibits tumor cell growth. Tipiracil protects trifluridine from being broken down when taken orally.[1]

References

 

 

 

Trifluridine.svg

Trifluridine

 

Trifluridine (also called trifluorothymidine or TFT) is an anti-herpesvirus antiviral drug, used primarily on the eye. It was sold under the trade name, Viroptic, by Glaxo Wellcome, now merged into GlaxoSmithKline. The brand is now owned by Monarch Pharmaceuticals, which is wholly owned by King Pharmaceuticals.

It is a nucleoside analogue, a modified form of deoxyuridine, similar enough to be incorporated into viral DNA replication, but the -CF3 group added to the uracil component blocks base pairing.

It is a component of the experimental anti-cancer drug TAS-102.

A Cochrane Systematic Review showed that trifluridine was a more effective treatment than idoxuridine or vidarabine, significantly increasing the relative number of successfully healed eyes in 14 days.[1]

References

  1. Wilhelmus KR (2010). “Antiviral treatment and other therapeutic interventions for herpes simplex virus epithelial keratitis”. Cochrane Database Syst Rev 12: CD002898. doi:10.1002/14651858.CD002898.pub4. PMID 21154352.

External links

  • Costin D, Dogaru M, Popa A, Cijevschi I (2004). “Trifluridine therapy in herpetic in keratitis”. Rev Med Chir Soc Med Nat Iasi 108 (2): 409–12. PMID 15688823.
  • Kuster P, Taravella M, Gelinas M, Stepp P (1998). “Delivery of trifluridine to human cornea and aqueous using collagen shields.”. CLAO J 24 (2): 122–4. PMID 9571274.
  • O’Brien W, Taylor J (1991). “Therapeutic response of herpes simplex virus-induced corneal edema to trifluridine in combination with immunosuppressive agents.”. Invest Ophthalmol Vis Sci 32 (9): 2455–61. PMID 1907950.
  • “Trifluridine Ophthalmic Solution, 1%” (PDF). Retrieved 2007-03-24.

 

Tipiralacil, also known as TPI,  is a thymidine phosphorylase inhibitor (TPI). Tipiracil is one of the active components in TAS-102, which is an anticancer drug candidate currently in clinical trials. TAS-102 consists of the cytotoxin Trifluridine and the thymidine phosphorylase inhibitor (TPI) tipiracil.  Trifluridine is incorporated into DNA during DNA synthesis and inhibits tumor cell growth. Tipiracil protects trifluridine from being broken down when taken orally.

183204-72-0 (Tipiracil -HCl); 183204-74-2(Tipiracil ).

References

1: Peters GJ, Bijnsdorp IV. TAS-102: more than an antimetabolite. Lancet Oncol. 2012 Dec;13(12):e518-9. doi: 10.1016/S1470-2045(12)70426-6. PubMed PMID: 23182191.

2: Yoshino T, Mizunuma N, Yamazaki K, Nishina T, Komatsu Y, Baba H, Tsuji A, Yamaguchi K, Muro K, Sugimoto N, Tsuji Y, Moriwaki T, Esaki T, Hamada C, Tanase T, Ohtsu A. TAS-102 monotherapy for pretreated metastatic colorectal cancer: a double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol. 2012 Oct;13(10):993-1001. doi: 10.1016/S1470-2045(12)70345-5. Epub 2012 Aug 28. PubMed PMID: 22951287.

3: Sobrero A. TAS-102 in refractory colorectal cancer: caution is needed. Lancet Oncol. 2012 Oct;13(10):959-61. doi: 10.1016/S1470-2045(12)70376-5. Epub 2012 Aug 28. PubMed PMID: 22951286.

4: Doi T, Ohtsu A, Yoshino T, Boku N, Onozawa Y, Fukutomi A, Hironaka S, Koizumi W, Sasaki T. Phase I study of TAS-102 treatment in Japanese patients with advanced solid tumours. Br J Cancer. 2012 Jul 24;107(3):429-34. doi: 10.1038/bjc.2012.274. Epub 2012 Jun 26. PubMed PMID: 22735906; PubMed Central PMCID: PMC3405214.

5: Suzuki N, Nakagawa F, Nukatsuka M, Fukushima M. Trifluorothymidine exhibits potent antitumor activity via the induction of DNA double-strand breaks. Exp Ther Med. 2011 May;2(3):393-397. Epub 2011 Mar 21. PubMed PMID: 22977515; PubMed Central PMCID: PMC3440718.

6: Shintani M, Urano M, Takakuwa Y, Kuroda M, Kamoshida S. Immunohistochemical characterization of pyrimidine synthetic enzymes, thymidine kinase-1 and thymidylate synthase, in various types of cancer. Oncol Rep. 2010 May;23(5):1345-50. PubMed PMID: 20372850.

7: Temmink OH, Bijnsdorp IV, Prins HJ, Losekoot N, Adema AD, Smid K, Honeywell RJ, Ylstra B, Eijk PP, Fukushima M, Peters GJ. Trifluorothymidine resistance is associated with decreased thymidine kinase and equilibrative nucleoside transporter expression or increased secretory phospholipase A2. Mol Cancer Ther. 2010 Apr;9(4):1047-57. doi: 10.1158/1535-7163.MCT-09-0932. Epub 2010 Apr 6. PubMed PMID: 20371715.

8: Bijnsdorp IV, Kruyt FA, Fukushima M, Smid K, Gokoel S, Peters GJ. Molecular mechanism underlying the synergistic interaction between trifluorothymidine and the epidermal growth factor receptor inhibitor erlotinib in human colorectal cancer cell lines. Cancer Sci. 2010 Feb;101(2):440-7. doi: 10.1111/j.1349-7006.2009.01375.x. Epub 2009 Sep 29. PubMed PMID: 19886911.

9: Bijnsdorp IV, Peters GJ, Temmink OH, Fukushima M, Kruyt FA. Differential activation of cell death and autophagy results in an increased cytotoxic potential for trifluorothymidine compared to 5-fluorouracil in colon cancer cells. Int J Cancer. 2010 May 15;126(10):2457-68. doi: 10.1002/ijc.24943. PubMed PMID: 19816940.

10: Bijnsdorp IV, Kruyt FA, Gokoel S, Fukushima M, Peters GJ. Synergistic interaction between trifluorothymidine and docetaxel is sequence dependent. Cancer Sci. 2008 Nov;99(11):2302-8. doi: 10.1111/j.1349-7006.2008.00963.x. Epub 2008 Oct 18. PubMed PMID: 18957056.

11: Overman MJ, Kopetz S, Varadhachary G, Fukushima M, Kuwata K, Mita A, Wolff RA, Hoff P, Xiong H, Abbruzzese JL. Phase I clinical study of three times a day oral administration of TAS-102 in patients with solid tumors. Cancer Invest. 2008 Oct;26(8):794-9. doi: 10.1080/07357900802087242. PubMed PMID: 18798063.

12: Overman MJ, Varadhachary G, Kopetz S, Thomas MB, Fukushima M, Kuwata K, Mita A, Wolff RA, Hoff PM, Xiong H, Abbruzzese JL. Phase 1 study of TAS-102 administered once daily on a 5-day-per-week schedule in patients with solid tumors. Invest New Drugs. 2008 Oct;26(5):445-54. doi: 10.1007/s10637-008-9142-3. Epub 2008 Jun 5. PubMed PMID: 18528634.

13: Temmink OH, Emura T, de Bruin M, Fukushima M, Peters GJ. Therapeutic potential of the dual-targeted TAS-102 formulation in the treatment of gastrointestinal malignancies. Cancer Sci. 2007 Jun;98(6):779-89. Epub 2007 Apr 18. Review. PubMed PMID: 17441963.

14: Temmink OH, Hoebe EK, van der Born K, Ackland SP, Fukushima M, Peters GJ. Mechanism of trifluorothymidine potentiation of oxaliplatin-induced cytotoxicity to colorectal cancer cells. Br J Cancer. 2007 Jan 29;96(2):231-40. PubMed PMID: 17242697; PubMed Central PMCID: PMC2360012.

 

 


Filed under: cancer, Phase3 drugs Tagged: CANCER, PHASE 3, TAIHO, Taiho's Colon Cancer Drug

Timely post on litigation and polymorphism: Case study with Celgene’s Revlimid

$
0
0

Originally posted on Developing the Process:

Hi there, folks.  It has been a while since I blogged.  I decided that I would continue once again and share some of the references that I came across while perusing the journals, while using the free blog-hosting services that are available.  This particular post was something I came across while looking at some stocks online.  I was searching taking a look at Celgene (CELG) stock and came across this discussion regarding the tricky world of patent litigation and more specifically, its application to matters concerning crystal form and polymorphism.  Having worked both sides of generic and brand-name pharma, I am interested in how this case will work out.

I know that this article’s subject matter is on the periphery of what is involved in developing a process.  I have worked on crystallizations and been concerned with polymorph control, so this is a textbook case, that is current, about patent…

View original 256 more words


Filed under: Uncategorized

Saracatinib AZD0530 in phase 3 for Ovary Cancer,

$
0
0

Saracatinib
NCGC00241099, cas 379231-04-6

893428-71-2 (trihydrate)

N-(5-Chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methyl-1-piperazinyl)ethoxy]-5-[(tetrahydro-2H-pyran-4-yl)oxy]-4-quinazolinamine

N-(5-Chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)ethoxy]-5-(tetrahydropyran-4-yloxy)quinazolin-4-amine

4-(6-chloro-2,3-methylenedioxyanilino)-7-[2-(4-methyIpiperazin-l-yI)ethoxy]- 5-tetrahydropyran-4-yloxyquinazoline

4-(6-chloro-2,3-methylenedioxyanilino)- 7-[2-(4-methylpiperazin-l -yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline

AZD0530

C27H32ClN5O5

542.03

AstraZeneca Pharmaceuticals LP

Astrazeneca Ab, Astrazeneca Uk Ltd,

Saracatinib (AZD0530) is a highly selective, orally available, dual-specific Src/Abl kinase inhibitor with IC50 of 2.7 and 30 nM for c-Src and Abl kinase, respectively.Saracatinib (AZD0530) demonstrated potent antimigratory and antiinvasive effects in vitro, and inhibited metastasis in a murine model of bladder cancer. Antiproliferative activity of AZD0530 in vitro varied between cell lines (IC50=0.2 ~10 mM).

c-Src, Bcr–Abl, Yes1, Lck.target

AZD0530 is orally available 5-, 7-substituted anilinoquinazoline with anti-invasive and anti-tumor activities. AZD0530 is a dual-specific inhibitor of Src and Abl, protein tyrosine kinases that are overexpressed in chronic myeloid leukemia cells. This agent binds to and inhibits these tyrosine kinases and their effects on cell motility, cell migration, adhesion, invasion, proliferation, differentiation, and survival. Specifically, AZD0530 inhibits Src kinase-mediated osteoclast bone resorption.

AZD-0530 is a highly selective, dual-specific small molecule Src/Abl kinase inhibitor currently in phase II/III clinical trials at AstraZeneca for the treatment of ovarian cancer. Phase II clinical trials are also under way at the company for the treatment of solid tumors and hematological neoplasms. The Mayo Clinic is developing AZD-0530 in phase II clinical studies for the treatment of metastatic pancreas cancer.

Additional phase II trials are under way at the National Cancer Institute (NCI) for the treatment of colorectal cancer, prostate cancer, breast cancer, lung cancer, stomach cancer, soft tissue sarcoma, stage II or IV melanoma and thymic malignancies. A phase II trial for pancreatic cancer has been suspended. Src and Abl kinase are highly expressed in various human tumor types. No recent development has been reported for research into the treatment of head and neck cancer.

 

Phase II study of Saracatinib (AZD0530) for for the treatment of patients with hormone receptor-negative metastatic breast cancer : Nine patients were treated on study. After a median of 2 cycles (range 1-3), no patient had achieved CR, PR, or SD >6 months. The median time to treatment failure was 82 days (12-109 days).The majority (89%) of patients discontinued saracatinib because of disease progression. One patient acquired potentially treatment-related grade 4 hypoxia with interstitial infiltrates and was removed from the study. Common adverse events included fatigue, elevated liver enzymes, nausea, hyponatremia, dyspnea, cough, and adrenal insufficiency. CONCLUSIONS:  These efficacy results were not sufficiently promising to justify continued accrual to this study. Based on this series, saracatinib does not appear to have significant single-agent activity for the treatment of patients with ER(-)/PR(-) MBC. (source: Clin Breast Cancer. 2011 Oct;11(5):306-11.)

Phase II study of  Saracatinib (AZD0530) in patients with metastatic or locally advanced gastric or gastro esophageal junction (GEJ) adenocarcinoma:  Saracatinib has insufficient activity as a single agent in patients with advanced gastric adenocarcinoma to warrant further investigation. Further development in gastric cancer would require rational drug combinations or identification of a tumor phenotype sensitive to Src inhibition. (source: Invest New Drugs. 2011 Mar 12. [Epub ahead of print]).

  

Phase II study of saracatinib (AZD0530) for patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). Nine patients were enrolled. All patients had received prior radiotherapy and six patients had received prior chemotherapy for recurrent or metastatic disease. The most common adverse event was fatigue. Eight patients had progression of disease by response evaluation criteria in solid tumors (RECIST) within the first eight-week cycle and one patient was removed from the study after 11 days due to clinical decline with stable disease according to the RECIST criteria. Median overall survival was six months. The study was closed early due to lack of efficacy according to the early stopping rule. CONCLUSION: Single-agent saracatinib does not merit further study in recurrent or metastatic HNSCC. (source: Anticancer Res. 2011 Jan;31(1):249-53.)

893428-72-3 Saracatinib difumarate

893428-73-4 also

Saracatinib (AZD0530) is a Src inhibitor for c-Src with IC50 of 2.7 nM.

 

…………………………….

 

 

WO 2001094341

http://www.google.com/patents/WO2001094341A9?cl=en

………………….

WO 2006064217

http://www.google.fm/patents/EP1871769A2?cl=en

4-(6-chloro-2,3-methylenedioxyanilino)- 7-[2-(4-methylpiperazin-l -yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline which compound is disclosed as Compound No. 73 within the Table in Example 14 of International Patent Application WO 01/94341. That compound is described herein by way of the Formula I

 

and as AZD0530, the code number by which the compound is known.

AZD0530 is an inhibitor of the Src family of non-receptor tyrosine kinase enzymes and, thereby, is a selective inhibitor of the motility of tumour cells and a selective inhibitor of the dissemination and invasiveness of mammalian cancer cells leading to inhibition of metastatic tumour growth. In particular, the compound AZD0530 is an inhibitor of c-Src non-receptor tyrosine kinase and should be of value as an anti-invasive agent for use in the containment and/or treatment of solid tumour disease in the human or animal body. The route for preparing the compound of the Formula I that is disclosed in International Patent Application WO 01/94341 involves the reaction of the compound 4-(6-chloro-2,3-methylenedioxyanilino)-7-hydroxy-5-tetrahydropyran-4-yloxyquinazoline with an alkylating agent to form the 2-(4-methylpiperazin-l-yl)ethoxy side-chain at the 7-position. The product of the reaction is disclosed in WO 01/94341 in the form of a dihydrochloride salt and in the form of a free base.

 

Example 14 4-(6-chloro-2,3-methylenedioxyaniIino)-7-[2-(4-methyIpiperazin-l-yI)ethoxy]- 5-tetrahydropyran-4-yloxyquinazoline (route 4)

Under an atmosphere of nitrogen gas, l-(2-hydroxyethyl)-4-methylpiperazine (13.93 g) was added to a stirred mixture of 4-(6-chloro-2,3-methylenedioxyanilino)-7-fluoro- 5-tetrahydropyran-4-yloxyquinazoline (12.9 g), sodium te/t-pentoxide (9.87 g) and 1 ,2-diethoxyethane (37.5 ml). Water (1.34 g) and 1,2-diethoxyethane (25 ml) were added and the resultant reaction mixture was stirred and heated to 86°C for 18 hours. The reaction mixture was cooled to 5O0C and, under vacuum distillation at approximately 60 millibar pressure, approximately 50 ml of reaction solvent was distilled off. The reaction mixture was neutralised to pH 7.0 to 7.6 by the addition of a mixture of concentrated aqueous hydrochloric acid (36%, 10 ml) and water (84 ml) at a rate that kept the temperature of the reaction mixture at a maximum of 6O0C. With the temperature of the reaction mixture being kept at 6O0C, the reaction mixture was extracted with ethyl acetate (225 ml). The organic solution was washed with water (50 ml). Water (25 ml) was added and, with the temperature being kept at 6O0C, the mixture was stirred for 10 minutes, then allowed to stand for 30 minutes and the aqueous layer was separated. The organic layer was concentrated to a volume of about 100 ml by distillation of solvent at about 9O0C under atmospheric pressure. The residual mixture was cooled during 1 hour to 450C and held at that temperature for 2 hours to allow crystallisation of product. The mixture was warmed briefly to 550C and then cooled during 4 hours to 180C and held at that temperature for 1 hour. The crystalline precipitate was isolated by filtration and washed in turn with water (17 ml) and with tø’t-butyl methyl ether (17 ml). There was thus obtained 4-(6-chloro-2,3-πiethylenedioxyanilino)-7-[2-(4-methylpiperazin-l-yl)ethoxy]- 5-tetrahydropyran-4-yloxyquinazoline as a trihydrate (11 g; 88% purity by HPLC using Method B, retention time 7.3 minutes); NMR Spectrum: (CDCl3) 1.65 (br s, 3H), 1.9-2.05 (m, 2H), 2.2-2.3 (m, 2H), 2.31 (s, 3H), 2.4-2.8 (m, 8H), 2.9 (m, 2H), 3.6-3.7 (m, 2H), 3.95-4.05 (m, 2H), 4.2-4.25 (m, 2H), 4.8 (m,lH), 6.05 (s, 2H), 6.55 (s, IH), 6.75 (d, IH), 6.85 (s, IH), 7.0 (d, IH), 8.55 (s, IH), 9.25 (s, IH).

A portion (10 g) of the material so obtained was placed on a filter and dried at ambient temperature in a stream of dry nitrogen gas. The resultant material was dissolved at 6O0C in dry isopropanol (140 ml) whilst maintaining a dry nitrogen atmosphere. The solution was allowed to cool to ambient temperature and to stand under a dry nitrogen atmosphere for 2 days. The resultant crystalline solid was isolated by filtration under a dry nitrogen atmosphere. The material (8 g) so obtained was a crystalline anhydrous form of 4-(6-chloro-2,3-methylenedioxyanilino)-7-[2-(4-methylpiperazin-l -yl)ethoxy]- 5-tetrahydropyran-4-yloxyquinazoline, m.p. 142 to 1440C.

Example 15

4-(6-chloro-2,3-methylenedioxyanilino)-7-[2-(4-methylpiperazin-l-yl)ethoxy]- 5-tetrahydropyran-4-yloxyquinazoline difumarate salt

A mixture of 4-(6-chloro-2,3-methylenedioxyanilino)-7-[2-(4-methylpiperazin- l-yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline trihydrate (27.1 g), isopropanol (200 ml) and water (10 ml) was heated to 75°C. A mixture of fumaric acid (12.8 g), isopropanol (200 ml) and water (40 ml) was heated to 8O0C. A portion (80 ml) of the warmed solution of the quinazoline compound was added to the fumaric acid solution whilst the temperature was maintained at 750C. The resultant mixture was stirred at 750C for 75 minutes. The remainder of the quinazoline compound solution was added during 1 hour whilst the temperature was maintained at 750C. Isopropanol (50 ml) was added and the resultant mixture was stirred at 750C for 7 hours. The mixture was cooled slowly over at least 25 minutes to 5O0C and was stirred at that temperature for 6 hours. The mixture was cooled slowly over at least 20 minutes to 2O0C and was stirred at that temperature for 18.5 hours. The crystalline solid was isolated by filtration, washed twice with a 10:1 mixture of isopropanol and water (50 ml and 100 ml respectively) and dried in vacuo at 450C to constant weight. There was thus obtained 4-(6-chloro- 2,3-methylenedioxyanilino)-7-[2-(4-methylρiperazin-l-yl)ethoxy]-5-tetrahydropyran- 4-yloxyquinazoline difumarate salt (37.0 g); m.p. 233-2370C; NMR Spectrum: (DMSOd6) 1.76-1.88 (m, 2H), 2.1-2.17 (m, 2H)5 2.33 (s, 3H), 2.6 (br s, 8H), 2.78 (t, 2H), 3.51-3.6 (m, 2H)3 3.83-3.9 (m, 2H), 4.24 (t, 2H)5 4.98-5.07 (m, IH), 6.07 (s, 2H)3 6.6 (s, 4H)5 6.83 (d5 IH)3 6.84 (d, IH)5 6.91 (d3 IH)5 7.05 (d, IH)3 8.33 (s, IH)3 9.18 (s, IH).

Example 16

4-(6-chloro-2,3-methyIenedioxyaniIino)-7-[2-(4-methyIpiperazin-l-yl)ethoxy]- 5-tetrahydropyran-4-yIoxyquinazolme difumarate salt

A mixture of 4-(6-chloro-2,3-methylenedioxyanilino)-7-[2-(4-methylpiperazin- l-yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline trihydrate (27.1 g), isopropanol (210 ml) and water (30 ml) was heated to 4O0C and the mixture was filtered. The filter was washed with isopropanol (20 ml) and the washings were added to the warm filtrate. The resultant solution was warmed to 75°C.

A mixture of fumaric acid (12.8 g), isopropanol (200 ml) and water (20 ml) was heated to 700C and the resultant mixture was filtered. A portion (110 ml) of the fumaric acid solution was added to the warmed solution of 4-(6-chloro-2,3-methylenedioxyanilino)- 7-[2-(4-methylpiperazin- 1 -yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline whilst the temperature was maintained at 75°C. Seed crystals of 4-(6-chloro-

253-methylenedioxyanilino)-7-[2-(4-methylpiperazin-l-yl)ethoxy]-5-tetrahydropyran- 4-yloxyquinazoline difumarate salt (0.02 g) were added and the resultant mixture was stirred at 750C for 1 hour. The remainder of the fumaric acid solution was added during 1 hour whilst the temperature was maintained at 750C and the resultant mixture was stirred at 750C for 14 hours.

The mixture was cooled slowly over at least 2 hours to 200C and was stirred at that temperature for 1 hour. The crystalline solid was isolated by filtration, washed twice with a 10:1 mixture of isopropanol and water (50 ml and 100 ml respectively) and dried in vacuo at 450C to constant weight. There was thus obtained 4-(6-chloro-253-methylenedioxyanilino)- 7-[2-(4-methylpiperazin-l-yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline difumarate salt (35.8 g); m.p. 234-237°C; NMR Spectrum: (DMSOd6) 1.76-1.88 (m, 2H)5 2.1-2.17 (m5 2H)5 2.33 (s5 3H)5 2.6 (br s, 8H), 2.78 (t, 2H), 3.51-3.6 (m, 2H), 3.83-3.9 (m, 2H), 4.24 (t, 2H)5 4.98-5.07 (m, IH), 6.07 (s, 2H)5 6.6 (s, 4H), 6.83 (d, IH)5 6.84 (d, IH)5 6.91 (d, IH)5 7.05 (d, IH)5 8.33 (s5 IH)5 9.18 (s5 IH).

Example 17 4-(6-chloro-2,3-methylenedioxyanilino)-7-[2-(4-methyIpiperazin-l-yI)ethoxy]- 5-tetrahydropyran-4-yloxyquinazoline sesquifumarate salt

A mixture of 4-(6-chloro-253-methylenedioxyanilino)-7-[2-(4-methylpiperazin- l-yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline difurnarate (0.15 g) and water (20 ml) was warmed using a heat gun to obtain a solution. The sample was allowed to evaporate slowly at ambient temperature to a volume of about 3 ml under a flow of air for 24 hours whereupon a precipitate had started to form. The mixture was placed in a refridgerator at 4°C for 2 days. The resultant precipitate was isolated by filtration and washed with water. There was thus obtained 4-(6-chloro-253-methylenedioxyanilino)-7-[2-(4-methylpiperazin-l-yl)ethoxy]- 5-tetrahydropyran-4-yloxyquinazoline as a sesquifumarate tetrahydrate salt (0.084 g) which was characterised using XRPD5 DSC5 TGA5 FTIR and solution NMR techniques.

………………..

A simplified process for the manufacture of AZD0530, a potent SRC kinase inhibitor
Org Process Res Dev 2011, 15(3): 688

http://pubs.acs.org/doi/abs/10.1021/op100161y

Abstract Image

Process research and development of a synthetic route towards a novel SRC kinase inhibitor is described. The Medicinal Chemistry route was very long and suffered from extensive use of chlorinated solvents and chromatography. A number of steps in the Medicinal Chemistry route were also unattractive for large-scale use for a variety of reasons. The route was modified to produce a shorter synthetic scheme that started from more readily available materials. By using the modified route, the title compound was manufactured on kilogram scale without recourse to chromatography and in significantly fewer steps. The scaled synthesis required two Mitsunobu couplings, which were developed and scaled successfully. An interesting hydrazine impurity was identified in the second Mitsunobu coupling; a mechanism for its formation is proposed, and a method for its control is described. The formation and control of some other interesting impurities are also described.

N-(5-Chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)ethoxy]-5-(tetrahydro-2H-pyran-4-yloxy)quinazolin-4-amine Difumarate (AZD0530 Difumarate)
To a slurry of 30 (2.139 kg at 92% w/w, 4.73 mol) and DTAD (2.771 kg, 12.03 mol) in THF (31 L) at ambient temperature in a 100 L vessel was added a solution of triphenylphosphine (3.057 kg, 11.66 mol) in THF (8 L) over 15 min. A THF (2 L) line wash was applied, and the mixture was stirred for 10 min. The reaction mixture was cooled to 15 °C, and a filtered (to remove undissolved particulates) solution of 31 (1.050 L, 1.049 kg, 7.27 mol) in THF …DELETED…………………………….The mixture was filtered and the cake washed with IPA (7 L as a slurry wash and 7 L as a displacement wash) before drying to constant weight under reduced pressure at 50 °C to give AZD0530 difumarate (3.546 kg at 89% w/w, 4.08 mol, 86% yield).
Final Purification of N-(5-Chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)ethoxy]-5-(tetrahydro-2H-pyran-4-yloxy)quinazolin-4-amine Difumarate (AZD0530 Difumarate)
AZD0530 difumarate (4.234 kg at 89% w/w, 4.87 mol) was refluxed in a mixture of IPA (10.L) and water (10.L, Fresenius). A solution was not obtained, so further IPA (450 mL) and water (450 mL, Fresenius) were added, and the mixture was refluxed. The resulting solution was cooled to 68 °C and screened over 3.5 min through a 20 μm in-line filter into a vessel preheated to 65 °C. IPA(20.4 L) at 65 °C was added via the first vessel and in-line filter, and the resulting solution was stirred at 65 °C for 2 h. Crystallisation was evident after 20 min. The mixture was allowed to self-cool to ambient temperature overnight before filtering and washing the cake with a mixture (prescreened through a 20 μm membrane) of water (640 mL) and IPA (5.76 L). The cake was washed with IPA (6.4 L, prescreened) and MTBE (6.4 L, prescreened) and dried to constant weight under reduced pressure at 50 °C to give AZD0530 difumarate (2.865 kg, at 95.2% w/w, 3.52 mol, 72% yield). Spectroscopic analysis was in agreement with the reported data…………Ford, J. G.; McCabe, J. F.; O’Kearney-McMullan, A.; O’Keefe, P.; Pointon, S. M.; Powell, L.; Purdie, M.; Withnall, J. WO/2006/064217, 2006.

 

……………………….

SEE…………N-(5-Chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)ethoxy]-5-(tetrahydro-2H-pyran-4-yloxy)quinazolin-4-amine, a novel, highly selective, orally available, dual-specific c-Src/Abl kinase inhibitor
J Med Chem 2006, 49(22): 6465

 

…………..

1: Hannon RA, Finkelman RD, Clack G, Iacona RB, Rimmer M, Gossiel F, Baselga J, Eastell R. Effects of Src kinase inhibition by saracatinib (AZD0530) on bone turnover in advanced malignancy in a Phase I study. Bone. 2012 Jan 8. [Epub ahead of print] PubMed PMID: 22245630.

2: Gucalp A, Sparano JA, Caravelli J, Santamauro J, Patil S, Abbruzzi A, Pellegrino C, Bromberg J, Dang C, Theodoulou M, Massague J, Norton L, Hudis C, Traina TA. Phase II trial of saracatinib (AZD0530), an oral SRC-inhibitor for the treatment of patients with hormone receptor-negative metastatic breast cancer. Clin Breast Cancer. 2011 Oct;11(5):306-11. Epub 2011 May 3. PubMed PMID: 21729667; PubMed Central PMCID: PMC3222913.

3: Mackay HJ, Au HJ, McWhirter E, Alcindor T, Jarvi A, Macalpine K, Wang L, Wright JJ, Oza AM. A phase II trial of the Src kinase inhibitor saracatinib (AZD0530) in patients with metastatic or locally advanced gastric or gastro esophageal junction (GEJ) adenocarcinoma: a trial of the PMH phase II consortium. Invest New Drugs. 2011 Mar 12. [Epub ahead of print] PubMed PMID: 21400081.

4: Fury MG, Baxi S, Shen R, Kelly KW, Lipson BL, Carlson D, Stambuk H, Haque S, Pfister DG. Phase II study of saracatinib (AZD0530) for patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). Anticancer Res. 2011 Jan;31(1):249-53. PubMed PMID: 21273606.

5: Renouf DJ, Moore MJ, Hedley D, Gill S, Jonker D, Chen E, Walde D, Goel R, Southwood B, Gauthier I, Walsh W, McIntosh L, Seymour L. A phase I/II study of the Src inhibitor saracatinib (AZD0530) in combination with gemcitabine in advanced pancreatic cancer. Invest New Drugs. 2010 Dec 18. [Epub ahead of print] PubMed PMID: 21170669.

6: Dalton RN, Chetty R, Stuart M, Iacona RB, Swaisland A. Effects of the Src inhibitor saracatinib (AZD0530) on renal function in healthy subjects. Anticancer Res. 2010 Jul;30(7):2935-42. PubMed PMID: 20683035.

7: Arcaroli JJ, Touban BM, Tan AC, Varella-Garcia M, Powell RW, Eckhardt SG, Elvin P, Gao D, Messersmith WA. Gene array and fluorescence in situ hybridization biomarkers of activity of saracatinib (AZD0530), a Src inhibitor, in a preclinical model of colorectal cancer. Clin Cancer Res. 2010 Aug 15;16(16):4165-77. Epub 2010 Aug 3. PubMed PMID: 20682712.

8: Morrow CJ, Ghattas M, Smith C, Bönisch H, Bryce RA, Hickinson DM, Green TP, Dive C. Src family kinase inhibitor Saracatinib (AZD0530) impairs oxaliplatin uptake in colorectal cancer cells and blocks organic cation transporters. Cancer Res. 2010 Jul 15;70(14):5931-41. Epub 2010 Jun 15. PubMed PMID: 20551056; PubMed Central PMCID: PMC2906706.

9: Hannon RA, Clack G, Rimmer M, Swaisland A, Lockton JA, Finkelman RD, Eastell R. Effects of the Src kinase inhibitor saracatinib (AZD0530) on bone turnover in healthy men: a randomized, double-blind, placebo-controlled, multiple-ascending-dose phase I trial. J Bone Miner Res. 2010 Mar;25(3):463-71. PubMed PMID: 19775203.

10: Rajeshkumar NV, Tan AC, De Oliveira E, Womack C, Wombwell H, Morgan S, Warren MV, Walker J, Green TP, Jimeno A, Messersmith WA, Hidalgo M. Antitumor effects and biomarkers of activity of AZD0530, a Src inhibitor, in pancreatic cancer. Clin Cancer Res. 2009 Jun 15;15(12):4138-46. Epub 2009 Jun 9. PubMed PMID: 19509160.

11: Chen Y, Guggisberg N, Jorda M, Gonzalez-Angulo A, Hennessy B, Mills GB, Tan CK, Slingerland JM. Combined Src and aromatase inhibition impairs human breast cancer growth in vivo and bypass pathways are activated in AZD0530-resistant tumors. Clin Cancer Res. 2009 May 15;15(10):3396-405. PubMed PMID: 19451593.

12: Lara PN Jr, Longmate J, Evans CP, Quinn DI, Twardowski P, Chatta G, Posadas E, Stadler W, Gandara DR. A phase II trial of the Src-kinase inhibitor AZD0530 in patients with advanced castration-resistant prostate cancer: a California Cancer Consortium study. Anticancer Drugs. 2009 Mar;20(3):179-84. PubMed PMID: 19396016; PubMed Central PMCID: PMC3225398.

13: Green TP, Fennell M, Whittaker R, Curwen J, Jacobs V, Allen J, Logie A, Hargreaves J, Hickinson DM, Wilkinson RW, Elvin P, Boyer B, Carragher N, Plé PA, Bermingham A, Holdgate GA, Ward WH, Hennequin LF, Davies BR, Costello GF. Preclinical anticancer activity of the potent, oral Src inhibitor AZD0530. Mol Oncol. 2009 Jun;3(3):248-61. Epub 2009 Feb 7. PubMed PMID: 19393585.

14: de Vries TJ, Mullender MG, van Duin MA, Semeins CM, James N, Green TP, Everts V, Klein-Nulend J. The Src inhibitor AZD0530 reversibly inhibits the formation and activity of human osteoclasts. Mol Cancer Res. 2009 Apr;7(4):476-88. PubMed PMID: 19372577.

15: Schweppe RE, Kerege AA, French JD, Sharma V, Grzywa RL, Haugen BR. Inhibition of Src with AZD0530 reveals the Src-Focal Adhesion kinase complex as a novel therapeutic target in papillary and anaplastic thyroid cancer. J Clin Endocrinol Metab. 2009 Jun;94(6):2199-203. Epub 2009 Mar 17. PubMed PMID: 19293266; PubMed Central PMCID: PMC2690419.

16: Purnell PR, Mack PC, Tepper CG, Evans CP, Green TP, Gumerlock PH, Lara PN, Gandara DR, Kung HJ, Gautschi O. The Src inhibitor AZD0530 blocks invasion and may act as a radiosensitizer in lung cancer cells. J Thorac Oncol. 2009 Apr;4(4):448-54. PubMed PMID: 19240653; PubMed Central PMCID: PMC2716757.

17: Gwanmesia PM, Romanski A, Schwarz K, Bacic B, Ruthardt M, Ottmann OG. The effect of the dual Src/Abl kinase inhibitor AZD0530 on Philadelphia positive leukaemia cell lines. BMC Cancer. 2009 Feb 13;9:53. PubMed PMID: 19216789; PubMed Central PMCID: PMC2654659.

18: Chang YM, Bai L, Liu S, Yang JC, Kung HJ, Evans CP. Src family kinase oncogenic potential and pathways in prostate cancer as revealed by AZD0530. Oncogene. 2008 Oct 23;27(49):6365-75. Epub 2008 Aug 4. PubMed PMID: 18679417.

Src inhibition with saracatinib reverses fulvestrant resistance in ER-positive ovarian cancer models in vitro and in vivo.
Simpkins et al. Clin Cancer Res. 2012 Aug 15. PMID: 22896656.

Saracatinib (AZD0530) is a potent modulator of ABCB1-mediated multidrug resistance in vitro and in vivo.
Liu et al. Int J Cancer. 2012 May 24. PMID: 22623106.

Common PIK3CA mutants and a novel 3′ UTR mutation are associated with increased sensitivity to saracatinib.
Arcaroli et al. Clin Cancer Res. 2012 May 1;18(9):2704-14. PMID: 22553375.

Phase I study of saracatinib (AZD0530) in combination with paclitaxel and/or carboplatin in patients with solid tumours.
Kaye et al. Br J Cancer. 2012 May 22;106(11):1728-34. PMID: 22531637.


Filed under: Phase3 drugs, Uncategorized Tagged: AZD0530, PHASE 3, Saracatinib

GSK2606414 is an orally available, potent, and selective PERK inhibitor

$
0
0

GSK2606414 structure

GSK2606414

1-(5-(4-amino-7-methyl-7H-pyrrolo[2,3-d]pyrimidin-5-yl)indolin-1-yl)-2-(3-(trifluoromethyl)phenyl)ethanone

1-[5-(4-Amino-7-methyl-7H-pyrrolo[2,3-d]pyrimidin-5-yl)indolin-1-yl]-2-(3-trifluoromethylphenyl)ethanone

CAS: 1337531-36-8

 Formula: C24H20F3N5O
Exact Mass: 451.16199

Glaxosmithkline Llc  innovator

CS-1428, QC-9698, GSK 2606414, KB-145925, GSK2606414|1337531-36-8|GSK-2606414

  nmr       ………http://www.medkoo.com/Product-Data/GSK2606414/GSK2606414-QC-APC40116Web.pdf

GSK2606414  is an orally available, potent, and selective PERK inhibitor. GSK2606414 inhibits PERK activation in cells and inhibits the growth of a human tumor xenograft in mice. Protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is activated in response to a variety of endoplasmic reticulum stresses implicated in numerous disease states. Evidence that PERK is implicated in tumorigenesis and cancer cell survival stimulated our search for small molecule inhibitors. (12/13/2013).

GSK2606414 structure.png

In vitro protocol:

J Med Chem. 2012 Aug 23;55(16):7193-207

In vivo protocol:

J Med Chem. 2012 Aug 23;55(16):7193-207

……………..

The chemical structures of GSK2606414  and GSK2656157 are very similar. The following graphic is a side-by-side comparison.

GSK2606414-GSK2656157 structure

 

GSK2606414 is a drug which is the first selective inhibitor discovered for the enzyme protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), which is involved in various processes relating to cancer and neurodegenerative disorders. GSK2606414 was found to be a potent and selective inhibitor of PERK, with good oral bioavailability and blood-brain barrier penetration.[1] PERK mediates the unfolded protein response pathway which is involved in the initiation of protein synthesis, and this pathway has been implicated in the neurotoxicity of various diseases including prion and Alzheimer’s diseases. Treatment with GSK2606414 was found to be neuroprotective in mice against damage caused by prions, and prevented the development of cognitive deficits and other clinical manifestations of prion disease. Extension of lifespan in treated mice was, however, not recorded. However, side effects such as weight loss and elevated blood glucose levels were also observed, likely due to unwanted inhibition of PERK in the pancreas gland, where it is involved in regulating insulin production.[2]

WO 2011119663

http://www.google.com/patents/WO2011119663A1?cl=en

Example 35

5-{1 -[(3-fluorophenyl)acetyl]-2,3-dihydro-1 H-indol-5-yl}-7-methyl-7H-pyrrolo[2,3- d]pyrimidin-4-amine

Figure imgf000116_0001

In a 20 mL vial with cap, to the solution of 5-(2,3-dihydro-1 H-indol-5-yl)-7-methyl-7H- pyrrolo[2,3-d]pyrimidin-4-amine2HCI (70.6 mg, 0.209 mmol), (3-fluorophenyl)acetic acid (32.2 mg, 0.209 mmol), HATU (79 mg, 0.209 mmol) in DMF (2 mL) was added Hunig’s base (0.146 mL, 0.836 mmol). The mixture was stirred at rt for over night. LCMS showed reaction was completed. The reaction was poured into water, white solid formed. The solid was filtered and dried to afford a white solid as the product. 1H NMR (400 MHz, DMSO- cfe) δ ppm 3.23 (t, J=8.46 Hz, 2 H), 3.73 (s, 3 H), 3.92 (s, 2 H), 4.19 – 4.26 (m, 2 H), 7.08 – 7.1 1 (m, 1 H), 7.12 – 7.17 (m, 2 H), 7.23 (d, J=8.34 Hz, 1 H), 7.25 (s, 1 H), 7.31 (s, 1 H), 7.36 (s, 1 H), 7.39 (d, J=6.82 Hz, 1 H), 8.10 – 8.17 (m, 2 H).

References

  1. Axten, J. M.; Medina, J. S. R.; Feng, Y.; Shu, A.; Romeril, S. P.; Grant, S. W.; Li, W. H. H.; Heerding, D. A.; Minthorn, E.; Mencken, T.; Atkins, C.; Liu, Q.; Rabindran, S.; Kumar, R.; Hong, X.; Goetz, A.; Stanley, T.; Taylor, J. D.; Sigethy, S. D.; Tomberlin, G. H.; Hassell, A. M.; Kahler, K. M.; Shewchuk, L. M.; Gampe, R. T. (2012). “Discovery of 7-Methyl-5-(1-{\3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-pyrrolo\2,3-d]pyrimidin-4-amine (GSK2606414), a Potent and Selective First-in-Class Inhibitor of Protein Kinase R (PKR)-like Endoplasmic Reticulum Kinase (PERK)”. Journal of Medicinal Chemistry 55 (16): 7193–7207. doi:10.1021/jm300713s.PMID 22827572. edit
  2.  Moreno, J. A.; Halliday, M.; Molloy, C.; Radford, H.; Verity, N.; Axten, J. M.; Ortori, C. A.; Willis, A. E.; Fischer, P. M.; Barrett, D. A.; Mallucci, G. R. (2013). “Oral Treatment Targeting the Unfolded Protein Response Prevents Neurodegeneration and Clinical Disease in Prion-Infected Mice”. Science Translational Medicine 5 (206): 206ra138. doi:10.1126/scitranslmed.3006767. edit

1: Axten JM, Medina JR, Feng Y, Shu A, Romeril SP, Grant SW, Li WH, Heerding DA,  Minthorn E, Mencken T, Atkins C, Liu Q, Rabindran S, Kumar R, Hong X, Goetz A, Stanley T, Taylor JD, Sigethy SD, Tomberlin GH, Hassell AM, Kahler KM, Shewchuk LM, Gampe RT. Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK). J Med Chem. 2012 Aug 23;55(16):7193-207. doi: 10.1021/jm300713s. Epub 2012 Aug 8. PubMed PMID: 22827572

.


Filed under: Preclinical drugs, Uncategorized Tagged: CS-1428, GSK 2606414, GSK2606414, QC-9698

New breast cancer stem cell findings explain how cancer spreads

Viewing all 2925 articles
Browse latest View live


<script src="https://jsc.adskeeper.com/r/s/rssing.com.1596347.js" async> </script>