Quantcast
Channel: New Drug Approvals
Viewing all 2025 articles
Browse latest View live

LM11A-31-BHS….. (2S,3S)-2-amino-3-methyl-N-(2-morpholinoethyl)-pentanamide

$
0
0

 

LM11A-31-BHS

(2S,3S)-2-amino-3-methyl-N-(2-morpholinoethyl)-pentanamide

2-Amino-3-methyl-N-[2-(4-morpholinyl)ethyl]-pentanamide dihydrochloride

  • CAS Number 1214672-15-7
  • Empirical Formula C12H25N3O2 · 2HCl
  • Molecular Weight 316.27

LM11A-31 is a non-peptide ligand of the p75 neurotrophin receptor (p75NTR). LM11A-31 blocks pro-NGF induced cell death in neuronal cultures, and protects neuronal cells from the the cytotoxic effects of cisplatin or methotrexate. Oral administration of LM11A-31 promotes the survival of oligodendrocytes and myelinated axons in a mouse spinal cord injury model and improves function in both weight-bearing and non-weight bearing tests.Inhibits death of hippocampal neurons at 100–1,000 pM

http://amcrasto.wix.com/anthony-melvin-crasto/apps/blog/lm11a-31-new-drug-can-help-paralyzed

PharmatrophiX

LM11A-31, NEW DRUG CAN HELP PARALYZED PEOPLE WALK AGAIN

Figure 2.

 

LM11A-31, C12 H25 N3 O2, Pentanamide, 2-amino-3-methyl-N-[2-(4-morpholinyl)ethyl]- WO 2010102212 TO LONGO FRANK, PUB 10.09.2010 THE UNIVERSITY OF NORTH CAROLINA AT CHAPEL HILL

PATENT LINK

http://patentscope.wipo.int/search/en/WO2010102212

Scientists have developed a pill which they claim could help paralyzed people walk again.

The new drug allowed mice with no movement in their lower limbs to walk with ‘well-coordinated steps’ and even to replicate swimming motions, researchers said.

The experimental drug, called LM11A-31, was developed by Professor Frank Longo, of Stanford University, California.

The researchers gave three different oral doses of LM11A-31, as well as a placebo, to different groups of mice beginning four hours after injury and then twice daily for a 42 day experimental period, the ‘Daily Mail’ reported.

In tests, the experimental medication did not increase pain in the mice and showed no toxic effects on the animals.

It also efficiently crossed the blood brain barrier, which protects the central nervous system from potentially harmful chemicals carried around in the rest of the bloodstream.

An injury to the spinal cord stops the brain controlling the body and this is the first time an oral drug has been shown to provide an effective therapy.

“This is a first to have a drug that can be taken orally to produce functional improvement with no toxicity in a rodent model,” Professor Sung Ok Yoon, of Ohio State University, Columbus, said.

“So far, in the spinal cord injury field with rodent models, effective treatments have included more than one therapy, often involving invasive means. Here, with a single agent, we were able to obtain functional improvement,” Yoon said.

The small molecule in the study was tested for its ability to prevent the death of cells called oligodendrocytes.

These cells surround and protect axons, long projections of a nerve cell, by wrapping them in a myelin sheath that protect the fibres.

In addition to functioning as axon insulation, myelin allows for the rapid transmission of signals between nerve cells.

The drug preserved oligodendrocytes by inhibiting the activation of a protein called p75. Yoon’s lab previously found p75 is linked to the death of these specialised cells after a spinal cord injury. When they die, axons that are supported by them degenerate.

“Because we know oligodendrocytes continue to die for a long period of time after an injury, we took the approach that if we could put a brake on that cell death, we could prevent continued degeneration of axons,” she said.

FULL TEXT – JOURNAL OF NEUROSCIENCE

Small, Nonpeptide p75NTR Ligands Induce Survival Signaling and Inhibit proNGF-Induced Death  in Journal of neuroscience, 26(20): 5288-5300; doi: 10.1523/​JNEUROSCI.3547-05.2006 by SM Massa – 2006 - Cited by 51 - Related articles
17 May 2006 – At 5 nm, LM11A-24 and -31 inhibit TUNEL staining to a degree  We further prioritized LM11A-31, because preliminary studies

Small, Nonpeptide p75NTR Ligands Induce Survival Signaling and Inhibit proNGF-Induced Death

Figure 1.

2010 SLIDE PRESENTATION RE P75 (E.G. LM11A-31) BY PHARMATROPHIX’S 

investorvillage.com/smbd.asp?mb=160&mn=440341…

3 Nov 2010 – 2010 slide presentation re p75 (e.g. LM11A-31) by PharmatrophiX’s founder. Longo is PharmatrophiX’s founder.

The experimental drug was developed by Prof Frank Longo from Stanford UniversityThe experimental drug was developed by Prof Frank Longo from Stanford University

Prof Frank Longo from Stanford University publications

http://med.stanford.edu/profiles/cancer/frdActionServlet?choiceId=showFacPublications&fid=7249&

Patents

1 US2013005731  (A1) ― 2013-01-03

http://worldwide.espacenet.com/publicationDetails/originalDocument?CC=US&NR=2013005731A1&KC=A1&FT=D&ND=3&date=20130103&DB=worldwide.espacenet.com&locale=en_EP

2 WO2011150347  (A2) ― 2011-12-01

http://worldwide.espacenet.com/publicationDetails/originalDocument?CC=WO&NR=2011150347A2&KC=A2&FT=D&ND=3&date=20111201&DB=worldwide.espacenet.com&locale=en_EP

3 US2011230479  (A1) ― 2011-09-22

http://worldwide.espacenet.com/publicationDetails/originalDocument?CC=US&NR=2011230479A1&KC=A1&FT=D&ND=3&date=20110922&DB=worldwide.espacenet.com&locale=en_EP

<a href=”http://www.bloglovin.com/blog/4674983/?claim=hj3e8pdf2nd”>Follow my blog with Bloglovin</a>

………………..

http://www.google.com.mx/patents/US7723328

TABLE I
Structures of Compounds 1-6
Compound Name
Figure US07723328-20100525-C00018
Compound 1 (also referred to herein as “LM11A-28”)
Figure US07723328-20100525-C00019
Compound 2 (also referred to herein as “LM11A-7”)
Figure US07723328-20100525-C00020
Compound 3 (also referred to herein as “LM11A-24”, “24”, and “C24”)
Figure US07723328-20100525-C00021
Compound 4 (also referred to herein as “LM11A-31” and “31”)
Figure US07723328-20100525-C00022
Compound 5 (also referred to herein as “LM11A-36”, “36”, and “C36”)
Figure US07723328-20100525-C00023
Compound 6 (also referred to herein as “LM11A-38” and “C38”)
Figure US07723328-20100525-C00024
Compound 7

 

…………………….

http://www.google.co.in/patents/WO2010102212A2?cl=en

Table I. Structures of Compounds i-vii

 

Figure imgf000050_0001
Figure imgf000051_0001

Example 32: Preparation of enantiomerically pure 2-amino-3-methyl-N-(2- morpholino-ethyϊ)-pentanamide

[00332] 2-amino-3-methyl-N-(2-morpholinoethyl)-pentanamide can be prepared by a method shown in Scheme 4 below. First, 2-aminoethanol (Compound IE) is transformed to its derivative with a leaving group (Compound 2E). Examples of the leaving group include halides and alkoxy or other activated hydroxyl group. Second, Compound 2E reacts with morpholine at a neutral or basic condition to yield 2-morpholinoethanamine (Compound 3E). The aforementioned two steps may also be performed continuously as one step with Compound 2E being generated in situ. For example, Compound 3 E can be prepared from Compound IE directly through a Mitsunobu reaction wherein the hydroxyl group of Compound IE is activated by diethyl azodicarboxylate (DEAD) before morpholine is added. The final product, 2-amino-3-methyl-N-(2-moipholinoethyl)-pentanamide (Compound 5E), can be obtained by coupling 2-morpholinoethanamine with 2-amino-3- methylpentanoic acid (Compound 4E) via a peptide coupling agent. Examples of the peptide coupling agent include l,r-carbonyldiimidazole (CDI), hydroxybenzotriazole (HOBT), 1,3-dicyclohexylcarbodiimide (DCC), 1- hydroxybenzo-7-azatriazole (HOAt), and the like. Scheme 4:

H2N^0H — H2N^ / LG , p , .

1 Ot= LG: a leaving group

1E zt

 

Figure imgf000099_0001

[00333] A chiral 2-amino-3-methyl-N-(2-moφholinoethyl)-pentanamide (Compound 5E) can be obtained by using the corresponding chiral 2-amino-3- methylpentanoic acid (Compound 4E) in the above coupling step. For example, (2S,3S)-2-amino-3-methyl-N-(2-moφholinoethyl)-pentanamide; (2R,3R)-2-amino- 3 -methyl-N-(2-morpholinoethyl)-pentanamide; (2R,3 S)-2-amino-3 -methyl-N-(2- moφholinoethyl)-pentanamide; and (2S,3R)-2-ammo-3-methyl-N-(2- morpholinoethyl)-pentanamide can be obtained by using (2S,3S)-2-amino-3- methylpentanoic acid, i.e., L-isoleucine; (2R,3R)-2-amino-3-methylpentanoic acid, i.e., D-isoleucine; (2R,3S)-2-amino-3-methylpentanoic acid, i.e., D-alloisoleucine; and (2S,3R)-2-amino-3-methylpentanoic acid, i.e., L-alloisoleucine, respectively. [00334] The chiral purity, also known as, enantiomeric excess or EE, of a chiral Compound 5E can be determined by any method known to one skilled in the art. For example, a chiral Compound 5E can be hydrolyzed to Compound 3E and the corresponding chiral Compound 4E. Then, the chiral Compound 4E obtained through hydrolysis can be compared with a standard chiral sample of Compound 4E to determine the chiral purity of the chiral Compound 5E. The determination can be conducted by using a chiral HPLC.

……………….

http://www.google.co.in/patents/EP2498782A1?cl=en

Scheme A shows the chemical structures of the present compounds.

 

Figure imgf000013_0001

(2S,3S)-2-amino-3-methyl-/V-(2-mor holinoethyl)pentanamide

 

Figure imgf000013_0002

(2R,3R)-2-amin -3-methyl-A/-(2-morpholinoethyl)pentanamide

 

Figure imgf000013_0003

(2S,3R)-2-amino-3-meth l-A/-(2-morpholinoethyl)pentanamide

 

Figure imgf000013_0004

] Q (2R,3S)-2-amino-3-methyl-/ /-(2-morpholinoethyl)pentanamide

The free base compound of 2-amino-3-niethyl- -(2-morpholinoethyl)-pentanamide can be prepared from isoleucine by synthetic methods known to one skilled in the art.

Standard procedures and chemical transformation and related methods are well known to one skilled in the art, and such methods and procedures have been described, for example, in standard references such as Fiesers’ Reagents for Organic Synthesis, John Wiley and Sons, New York, NY, 2002: Organic Reactions, vols, 1-83, John Wiley and Sons, New York, NY, 2006; March J, and Smith M,, Advanced Organic Chemistry, 6th ed., John Wiley and Sons, New York, NY; and Larock R.C., Comprehensive Organic Transformations, Wiley-VCH Publishers, New York, 1999. All texts and references cited herein are incorporated by reference in their entirety. Other related synthetic methods can be found in U.S. Patent Application Publication Nos. 2006/024072 and 2007/0060526, the contents of which are herein incorporated by reference in their entirety for all purposes. The amorphous dihydrochloride (di-HCl) salt of 2-amino-3-methyl-N-(2-morpholinoethyl)-pentanamide can be prepared by mixing two molar ecjuivalents of HC1 with one molar equivalent of 2-amino- 3-methyl-N-(2-morpholinoethyl)~pentanamide in appropriate solvent(s) and then separating the di-HCl salt from the solvent(s) mixture.

The amorphous di-HCl salt of 2-aniino-3-methyl-N-(2-moi holinoethyl)-pentariamide was analyzed via the methods as described above. The XRD analysis indicated it was amorphous/low ordered as shown in Figure 1 , The DSC thermogram exhibited a broad endotherm with onset temperature 37 °C and peak temperature 74 °C and an enthalpy value of ΔΗ = 80 J/g. The TGA thermogram indicated the di-HCl salt is anhydrous and starts to decompose after about 200°C. An overlay of DSC and TGA thermograms are shown in Figure 2. The moisture sorption-desorpiion isotherm of the di-HC! salt (Figures 3 A and 3 B ) was collected using dynamic vapor sorption (DVS) analysis. The material did not adsorb much moisture from 0% to 20% RH, then it showed steady sorption up to 140 wt% moisture at 95% RH (likely deliquescence). This sample showed rapid desorption from 95% to 70% RH and then continues desorbing at a relatively slower pace to a mass about 5 wt% greater than the original value at 0% RH. This sample shows a small hysteresis between the sorption and desorption phase. O verall this material is quite hygroscopic. The crude solubility of the di-HCl salt in water was >30 mg/niL. The proton N MR spectrum of the amorphous di-HCl salt is shown in Figure 4. Example 2. Preparation of 2-amino-3-methyl- -(2-morpholinoethy[)-pentanamide (free base):

Five grams of 2-amino-3-methyl-N-(2-morpholinoethyl)-pentanamide di-HCl salt was dissolved in 150 mL of ethanol. Sodium bicarbonate (5.3 g), dissolved in 100 mL of HPLC water, was added to this solution. The mixed solution was sonicated for ~10 minutes. This solution was concentrated using a rotovap, and the residue was dissolved in 300 mL of methylene chloride. This solution was passed through a short plug of carbonate bonded silica gel. This solution was concentrated using rotovap and the residue was lyophilized to dry, resulting in 3.6 g of the free base as a white solid. Proton NMR, C-13 NMR and LC/MS confirmed the structure of this material as the free base of 2-amino-3-methyl-N-(2- morpholmoethyl)-pentanamide.

In the process of converting the di-HCl salt to free base, the sample was lyophilized to avoid formation of oil. XRD analysis of the lyophilized free base surprisingly re vealed it was crystalline, as shown in Figure 5. The DSC thermogram exhibited an endotherm with extrapolated onset temperature 51 °C and peak temperature 53 °C and an enthalpy value of Δ¾= 104 J/g. The TGA thermogram shows less than 0.6 wt% loss at 105 °C, suggesting it was solvent free. An overlay of the DSC and TGA thermograms can be seen in Figure 6. The crude solubility of free base in water was >30 mg/mL. The proton NMR was consistent with the free base. The NMR and Raman spectra are shown in Figures 7 and 8A and 8B, respectively. The moisture sorption-desorption isotherm (Figures 9 A and 9B) was collected using dynamic vapor sorption (DVS) analysis. The sample did not adsorb much moisture content from 0% to 45% RH under the experimental conditions. Above 45 %RH the sample appears to adsorb moisture of – 10 wt% from 45% to 50% RH followed by rapid sorption up to 96 wt% moisture at 95% RH. In the desorption phase, the free base shows a rapid desorption from 95% to 80°/» RH, then the sample desorbs at a relatively slow pace to the original weight at 0% RH. The sample may form a hydrate near 45 %>RH, The putative hydrate appears to deliquesce resulting in an amorphous glass by the end of the scan.

……………

new patent

WO-2014052659

Crystalline forms of neurotrophin mimetic compounds and their salts

Type II TNF receptor agonist; NGF receptor modulator

Crystalline forms of (2S,3S)-2-amino-3-methyl-N-(2-morpholinoethyl)-pentanamide (LM11A-31-BHS), useful for the treatment of neurodegenerative disorders such as Alzheimer’s disease (AD), Parkinson’s disease and multiple sclerosis. See WO2011066544 claiming deuterated compounds of LM11A-31-BHS, useful for treating neurodegenerative diseases. PharmatrophiX is investigating the p75 neutrophin receptor ligand, LM11A-31-BHS, for the oral treatment of AD. By March 2013, a phase I trial was planned. The drug was formerly being investigated in collaboration with Elan Corp and the deal was terminated by the fourth quarter of 2010.


Filed under: Phase2 drugs, Uncategorized Tagged: LM11A-31-BHS

4-methoxyphenyl methanol, (4-メトキシフェニル)メタノール NMR, IR , MASS

$
0
0

 

(4-methoxyphenyl)methanol

Structure: structure

IUPAC Name: 4-methoxyphenyl methanol

C6H10O3; MW = 138.17

The molecule contains two oxygens, and from the analysis, contains four double bonds, carbonyls or rings. The large number of degrees of unsaturation strongly suggests an aromatic compound (DU = 4).

The mass spectrum displays a molecular ion, which is the base peak, an m-1 and an m-17, all of which are consistent with a simple alcohol.

The 13C spectrum contains six peaks, indicating that the molecule has some elements of symmetry. The quartet at  56 and the triplet at  71 represent a CH3 and a CH2 group which are deshielded by electronegative atoms (most likely oxygen); the peaks at  161 – 128 are in the aromatic region; the fact that two doublets and two singlets are observed strongly suggests 1,4-disubstitution.

The proton NMR also shows evidence for aromatic 1,4-disubstitution and suggests that the methyl and methylene are isolated and adjacent to electronegative groups. A peak consistent with an alcoholic OH can also be seen.

The IR is consistent with an aromatic alcohol containing no carbonyl group, suggesting that the second oxygen is involved in an ether linkage.

The simplest structure which is consistent with all of these data would be an aromatic compound containing an alcohol group and a methyl ether, situated 1,4 relative to each other.

 

IH NMR

The proton NMR has two doublets at  6.9, consistent with aromatic 1,4-disubstitution, and three singlets, areas 3, 2 and 1. The singlets at  3.6 and 4.7 are highly shifted and suggest isolated CH3 and CH2 groups adjacent to one or more electronegative atoms or groups. The singlet, area 1, would be consistent with an alcohol.

 

NMR Spectrum

Predict NMR spectrum

 

13C NMR

The 13C spectrum contains six peaks, indicating that the molecule has some elements of symmetry. The quartet at  56 and the triplet at  71 represent a CH3 and a CH2 group which are deshielded by electronegative atoms (most likely oxygen); the peaks at  161 – 128 are in the aromatic region; the fact that two doublets and two singlets are observed strongly suggests 1,4-disubstitution.

13C NMR Data: q-56.0; t-71.0; d-114.3; d-128.3; s-160.9; s-133.2

13C NMR Assignments: C-13 assignments

 

MASS

The mass spectrum consists of a molecular ion at 138, which is also the base peak, an m-1 peak at 137, indicating the presence of a labile hydrogen (OH or CHO), and an m-17 peak (loss of HO-). The spectrum is consistent with an alcohol which cannot readily break down to form other stable radical cations.

Mass Spectrum

 

Mass Spectrum Fragments: C-13 assignments

 

 

IR

 

3400-3200 cm-1: strong OH peak 3100 cm-1: small peak, suggesting possible unsaturated CH 2900 cm-1: strong peak suggesting saturated CH 2200 cm-1: no unsymmetrical triple bonds 1710 cm-1: no carbonyl 1610 and 1500 cm-1: sharp peaks, consistent with aromatic carbon-carbon double bonds

Synthetic Communications, 18, p. 613, 1988 DOI: 10.1080/00397918808064019
Synthesis, p. 1081, 1984
Tetrahedron Letters, 32, p. 3243, 1991

 


Filed under: Anthony crasto, Uncategorized Tagged: (4-methoxyphenyl)methanol, IR, MASS, NMR

Diethyl benzylmalonate, IR, NMR, Mass

$
0
0

structure

The mass spectrum displays a molecular ion, an m-45 and a peak at m/e = 91, all of which are consistent with a molecule containing benzyl and ethoxy groups.

The 13C spectrum contains six peaks, indicating that the molecule has some elements of symmetry. The quartet at  56 and the triplet at  71 represent a CH3 and a CH2 group which are deshielded by electronegative atoms (most likely oxygen); the peaks at  161 – 128 are in the aromatic region; the fact that three doublets and one singlet are observed strongly suggests monosubstitution.

The proton NMR also shows evidence for two ethyl groups, a CH-CH2- group, and a monosubstituted aromatic group; the chemical shift suggests that the carbons of the CH-CH2- adjacent to one or more electronegative groups.

The IR is consistent with an aromatic compound containing a carbonyl group.

The simplest structure which is consistent with all of these data would be an aromatic compound linked via a -CH2CH group to a diethyl ester.

 

 

Structure: structure

IUPAC Name: ethyl ethyl benzylpropanedioate (diethyl benzylmalonate)

607-81-8

C14H18O4; MW = 250.29

 

1H NMR

The proton NMR has a coupled quartet and a triplet, consistent with an ethyl group in which the CH2 (at  4.1) is adjacent to an electronegative atom (most likely oxygen). The presence of a coupled triplet and doublet suggests the presence of a CH-CH2- group in which both carbons are adjacent to one or more electronegative atoms. The singlet at  7.1 is consistent with a monosubstituted aromatic compound.

 

NMR Spectrum

13C NMR

The 13C spectrum contains nine peaks, indicating that the molecule has some elements of symmetry. The quartet at  14 and the triplet at  60 represent a simple CH3 and a CH2 which is deshielded by an electronegative atom (most likely oxygen); the doublet at  58 and the triplet at  36 are CH and CH2 groups which are adjacent to one or more electronegative groups. The peaks at  141 – 125 are in the aromatic region; the fact that three doublets and one singlet are observed strongly suggests monosubstitution.

 

13C NMR Assignments: C-13 assignments

 

 

MASS

The mass spectrum consists of a molecular ion at 250, a base peak at 91 (a benzyl group), an m-45 peak at 205, indicating the presence of an ethoxy group; other significant peaks at 131 and 176 must be consistent with the proposed structure. The spectrum is consistent with a molecule containing ethoxy and benzyl groups.

Mass Spectrum

Mass Spectrum Fragments: C-13 assignments

 

 

IR

3400-3200 cm-1: no OH peak 3100 cm-1: small peak, suggesting unsaturated CH 2900 cm-1: strong peak suggesting saturated CH 2200 cm-1: no unsymmetrical triple bonds 1730 cm-1: strong carbonyl 1610 and 1500 cm-1: weak peaks, vaguely consistent with aromatic carbon-carbon double bonds

 

 

 

 

 

ADDITIONAL INFO FOR READER ON SPECTROSCOPY

SEE THE FUN WHEN A CARBONYL IS INTRODUCED

BENZOYLMALONIC ACID DIETHYL ESTER Structure

1H NMR

 

 

13 C NMR

IR

 

MASS

 

RAMAN

 


Filed under: Uncategorized Tagged: diethyl benzylmalonate, IR, MASS, NMR

1H NMR OF SIMPLE MOLECULES.. Brush up

$
0
0

 

ما هو هذا، لماذا أنت قلق، فرشاة مع الأشياء البسيطة، وسوف يخرج عبقري، وتعلم معي، قطرات صغيرة من الماء جعلالمحيطات، وسوف يكون خبيرا في هذا

what is this, why are you worried, brush up with simple things, you will come out genius, learn with me, small drops of water make an ocean, you will be an expert in this

あなたが心配している理由は、これは簡単なことでブラッシュアップ、、何か、あなたは、天才が出てくる私と一緒に学ぶことが、水の小さな滴が海を作るには、この専門家になる

 

 structure

4-methylbenzaldehyde

NMR Spectrum

The proton NMR has three peaks; a singlet at  2.2 (3H), and a singlet at  10 (1H) and two doublets centered around  7.6. The doublets centered at  7.6 are in the aromatic region; the fact that two doublets are observed (2H each) suggests a 1,4-disubstituted aromatic compound.

The peak at  2.2 is in the region for a methyl group adjacent a mildly electronegative group. The singlet at  10 is in the region observed for aldehydic protons. The presence of two doublets in the aromatic region is highly characteristic of 1,4-disubstitution.

τι είναι αυτό, γιατί είσαι ανήσυχος, βούρτσα με απλά πράγματα, θα βγει ιδιοφυΐα, να μάθουν μαζί μου, μικρές σταγόνες του νερού κάνει έναν ωκεανό, θα είστε ένας εμπειρογνώμονας σε αυτό

 structure

methyl acetate (methyl ethanoate)

NMR Spectrum

The proton NMR has two singlets of equal area. The fact that the molecule has 6 hydrogens means that there must be two non-identical CH3 groups in the molecule and that they are non-adjacent. The peak at  1.95 is in the area generally observed for methyl groups adjacent to carbonyls; the peak at  3.85 is in the region for methyl groups adjacent to electronegative atoms, i.e., oxygen.

আপনি চিন্তিত কেন এই সহজ জিনিস নিয়ে ব্রাশ,, কি, আপনি, প্রতিভা বাইরে আসতে আমার সাথে শিখতে হবে, জলের ছোট ঝরিয়া একটি মহাসাগর না, আপনি এই একজন বিশেষজ্ঞ হতে হবে

 structure
diethyl ketone (3-pentanone)

NMR Spectrum

The proton NMR has a quartet and a triplet, indicating a CH2 adjacent to a CH3. The peak at  2.5 is in the area generally observed for methyl groups adjacent to mildly electronegative groups; the peak at  1.2 is in the region for simple methyl groups adjacent to carbons (CH3CH2). The molecule contains oxygen, but the peak at  2.5 is not shifted enough for this to represent an -O- linkage, so it must represent a carbonyl.

 

מה זה, למה אתה מודאג, לרענן עם דברים פשוטים, אתה תצא גאון, ללמוד איתי, טיפות קטנות של מיםיגרמו לים, אתה תהיה מומחה בזה

 

 structure

acetaldehyde (ethanal)

NMR Spectrum

The proton NMR has two peaks; at high resolution, fine coupling is evident between them (J 1 Hz) with the peak at  9.7 being a quartet and the peak at  2.1 being a doublet, indicating a CH coupled to a CH3. The peak at  9.7 is in the area generally observed for aldehydic protons; the peak at  2.1 is in the region for a methyl group adjacent a carbonyl. The molecule contains oxygen, but the peak at  2.1 is not shifted enough for this to represent an -O- linkage, so it must represent a carbonyl. The very small coupling constant suggests that the proton and the CH3 group are not immediately adjacent, but that this represents an example of cross-space coupling.

o que é isso, por que você está preocupado, retocar com coisas simples, você vai sair gênio, aprender comigo, pequenas gotas de água fazem um oceano, você vai ser um especialista neste

 structure

Name: ethyl acetate (ethyl ethanoate)

NMR Spectrum

The proton NMR has three peaks; a quartet at  4.1 (2H), a triplet at  1.2 (3H) and a singlet at  1.97 (3H). The quartet and triplet suggest a CH2 coupled to a CH3 in an ethyl group. The peak at  4.1 is in the area generally observed for CH groups adjacent to electronegative groups, i.e., oxygen. The peak at  2 is in the region for a methyl group adjacent a carbonyl.

 

நீங்கள் கவலை ஏன் இந்த எளிய பொருட்களை கொண்டு துலக்க, என்ன, நீங்கள், மேதை வெளியே வர எனக்கு கற்று, நீர் சிறு துளிகள் ஒரு கடல் செய்ய, நீங்கள் இந்த ஒரு நிபுணர் இருக்கும்

के तपाईं चिंतित हो किन यो सरल कुरा संग ब्रश,, के हो, तपाईं, प्रतिभा बाहिर आउन मलाई संग सिक्न हुनेछ, पानी सानो थोपा एक महासागर बनाउन, तपाईं यस मा एक विशेषज्ञ हुनेछ\

તમને ચિંતા થતી હોય કે શા માટે આ સરળ બાબતો સાથે બ્રશ, શું છે, તમે પ્રતિભા બહાર આવે મારી સાથે શીખશે, પાણી નાના ટીપાં સમુદ્ર કરો, તો તમે આ એક નિષ્ણાત હશે

kas tas ir, kāpēc jūs uztraucaties, suka ar vienkāršām lietām, jūs iznākt ģēnijs, mācīties kopā ar mani, nelieli ūdens pilieni veikt okeānu, jums būs eksperts šajā

что это такое, почему ты беспокоишься, освежить с простых вещей, вы будете выходить гений, узнать со мной, маленькие капли воды сделать океан, вы будете экспертом в этом

 hvað er þetta, af hverju ert þú áhyggjur, bursta upp með einföldum hlutum, verður þú að koma út snillingur, læra með mér, litla dropa af vatni gera haf, verður þú að vera sérfræðingur í þessu
 structuremethyl vinyl ether (methyl ethenyl ether)
NMR Spectrum
The proton NMR has four groups of peaks; the singlet at  3.6 (3H) is consistent with an isolated CH3 group adjacent to an electronegative center, such as oxygen. The sets of highly split doublets centered at  6.5, 4.2 and 4.0 (there is overlap of these at  4.1) span the region where alkene hydrogens are observed. The complex splitting pattern observed resembles an ABC pattern for a terminal alkene.

这是什么,你为什么担心,刷了简单的事情,你会出来的天才,学我,小水珠做出的海洋,你将在这方面的专家

 당신이 걱정하는 이유는 간단한 것들로 브러시, 무엇인가, 당신은 천재 나올 나와 함께 배울 것, 물 작은 방울은 바다를 만들어,이 분야의 전문가가 될 것입니다
 structureName: 1,2-dimethoxyethane
NMR Spectrum
The proton NMR has two peaks; both singlets are consistent with isolated groups adjacent to an electronegative center, such as oxygen. The fact that the molecule contains ten hydrogens, but only shows singlets for two and three hydrogens requires a great deal of symmetry in the molecule, i.e., both CH3 groups must be identical, as must both CH2 groups.
structure4-isopropyl-1-methoxybenzene (4-(1-methylethyl)-1-methoxybenzene)
NMR Spectrum
The proton NMR has four sets of peaks; a singlet at  3.6 (3H), two sets of doublets centered around  6.9 (4H), a septet at  2.7 (1H) and a doublet at  1.6. The singlet at  3.6 is consistent with an isolated CH3adjacent to an electronegative center, such as an oxygen. The septet and doublet strongly suggest an isopropyl group CH(CH32 in which the carbon is bonded to something mildly electronegative and the two doublets centered at  6.9 strongly suggest a 1,4-disubstituted aromatic compound.
structure 3-bromomethyltoluene (3-bromomethyl-1-methylbenzene)
NMR Spectrum
The proton NMR has three sets of peaks; a singlet at  4.3 (2H), a messy singlet around  7.1 (4H), and a singlet at  2.3 (3H). The singlet at  2.3 is consistent with an isolated CH3 adjacent to a mildly electronegative center. The singlet at 4.3 is consistent with a CH2 group adjacent to two mildly electronegative centers (X-CH2-Y), and the singlet at  7.1 is consistent with a disubstituted aromatic compound. The substitution pattern is not what is normally seen in 1,4-disubstitution (two doublets), suggesting 1,2 or 1,3-disubstitution.
آپ پریشان کیوں ہیں اس سادہ چیزوں کے ساتھ برش،، کیا ہے، آپ، ہوشیار باہر آ میرے ساتھ سیکھ جائے گی، پانی کے چھوٹے چھوٹے قطرے ایک سمندر بنانے کے لئے، آپ کو اس میں ایک ماہر ہو جائے گا

ANTHONY MELVIN CRASTO

THANKS AND REGARD’S
DR ANTHONY MELVIN CRASTO Ph.D

amcrasto@gmail.com

MOBILE-+91 9323115463
GLENMARK SCIENTIST ,  INDIA
web link
http://anthonycrasto.jimdo.com/
Congratulations! Your presentation titled “Anthony Crasto Glenmark scientist, helping millions with websites” has just crossed MILLION views.
アンソニー     安东尼   Энтони    안토니     أنتوني
join my process development group on google
you can post articles and will be administered by me on the google group which is very popular across the world
LinkedIn group
 
blogs are
 
shark

 

 

 


Filed under: Uncategorized Tagged: NMR

13C NMR OF SIMPLE MOLECULES…………..Brush up….???

$
0
0
ما هو هذا، لماذا أنت قلق، فرشاة مع الأشياء البسيطة، وسوف يخرج عبقري، وتعلم معي، قطرات صغيرة من الماء جعلالمحيطات، وسوف يكون خبيرا في هذا

what is this, why are you worried, brush up with simple things, you will come out genius, learn with me, small drops of water make an ocean, you will be an expert in this

あなたが心配している理由は、これは簡単なことでブラッシュアップ、、何か、あなたは、天才が出てくる私と一緒に学ぶことが、水の小さな滴が海を作るには、この専門家になる

 

 

structure

Name: 1,2-dimethoxymethane

C4H10O2

From the molecular formula, the compound has “0 degrees of unsaturation” (no double bonds or rings).

C13 Spectrum

The 13C NMR has two peaks, a quartet at  54 (a CH3) and a triplet at  80 (a CH2). Since the molecule has four carbons and only two 13C NMR peaks, there must be symmetry. Both peaks are in the regions where carbons next to electronegative atoms occur (oxygen).

 

τι είναι αυτό, γιατί είσαι ανήσυχος, βούρτσα με απλά πράγματα, θα βγει ιδιοφυΐα, να μάθουν μαζί μου, μικρές σταγόνες του νερού κάνει έναν ωκεανό, θα είστε ένας εμπειρογνώμονας σε αυτό

আপনি চিন্তিত কেন এই সহজ জিনিস নিয়ে ব্রাশ,, কি, আপনি, প্রতিভা বাইরে আসতে আমার সাথে শিখতে হবে, জলের ছোট ঝরিয়া একটি মহাসাগর না, আপনি এই একজন বিশেষজ্ঞ হতে হবে

 

 

C5H7O2N

From the molecular formula, the compound has “3 degrees of unsaturation” (3 double bonds or rings).

 

structure

: ethyl cyanoacetate

C13 Spectrum

The 13C NMR has 5 peaks, a quartet at  14 (a CH3), a triplet at  59 (a CH2), another triplet at  22 (another CH2), and two singlets, one at  118 and one at  172. Since the molecule has five carbons and five 13C NMR peaks, there must be no symmetry. The singlet at  172 is in the carbonyl region, most likely an acid or an ester. The CH2 at  59 is in the region where carbons next to electronegative atoms occur (i.e., oxygen) and the CH3 at  14 is a simple terminal methyl, suggesting an -O-CH2CH3 residue. The singlet at  118 would be consistent with a nitrile carbon and the shielded CH2 at  22 suggests that it may be adjacent to the sp-carbon of the nitrile

 

 

 

מה זה, למה אתה מודאגלרענן עם דברים פשוטיםאתה תצא גאון, ללמוד איתי, טיפות קטנות של מיםיגרמו לים, אתה תהיה מומחה בזה
C6H10OFrom the molecular formula, the compound has “2 degrees of unsaturation” (2 double bonds or rings).

C13 Spectrum

structure

2-butanon-4-ene

The 13C NMR has 6 peaks, a quartet at  25 (a CH3), a triplet at  49 (a CH2), another quartet at  17 (another CH3), two doublets (a CH) , one at  124 and one at  131, and one singlet at  207. Since the molecule has six carbons and six 13C NMR peaks, there must be no symmetry. The singlet at  207 is in the carbonyl region, most likely an aldehyde or ketone. The CH3 groups at  17 and 25 are consistent with simple terminal methyl groups, with one slightly shifted by an mildly electronegative group (a carbonyl?). The doublets at  124 and 131 are in the alkene region, suggesting a -CHCH- group. The remaining CH2 group at  49 is probably deshielded by two electronegative groups.

 

o que é isso, por que você está preocupado, retocar com coisas simples, você vai sair gênio, aprender comigo, pequenas gotas de água fazem um oceano, você vai ser um especialista neste

C8H8O

From the molecular formula, the compound has “5 degrees of unsaturation” (5 double bonds or rings).

structure

acetophenone

 

C13 Spectrum

The 13C NMR has 6 peaks, a quartet at  27 (a CH3), three doublets (CH groups), at  129, 128 and 133, and two singlets, one at  137 and one at  197. Since the molecule has eight carbons and six 13C NMR peaks, there must some degree of symmetry. The singlet at  197 is in the carbonyl region, most likely an aldehyde or ketone. The CH3 groups at  27 is consistent with a simple terminal methyl group, slightly shifted by an mildly electronegative group (a carbonyl?). The doublets at  129, 128 and 133 and the singlet at 137 are in the aromatic region, suggesting a monosubstituted aromatic group, with symmetry in four of the six carbons.

 

நீங்கள் கவலை ஏன் இந்த எளிய பொருட்களை கொண்டு துலக்க, என்ன, நீங்கள், மேதை வெளியே வர எனக்கு கற்று, நீர் சிறு துளிகள் ஒரு கடல் செய்ய, நீங்கள் இந்த ஒரு நிபுணர் இருக்கும்

के तपाईं चिंतित हो किन यो सरल कुरा संग ब्रश,, के हो, तपाईं, प्रतिभा बाहिर आउन मलाई संग सिक्न हुनेछ, पानी सानो थोपा एक महासागर बनाउन, तपाईं यस मा एक विशेषज्ञ हुनेछ\

તમને ચિંતા થતી હોય કે શા માટે આ સરળ બાબતો સાથે બ્રશ, શું છે, તમે પ્રતિભા બહાર આવે મારી સાથે શીખશે, પાણી નાના ટીપાં સમુદ્ર કરો, તો તમે આ એક નિષ્ણાત હશે

kas tas ir, kāpēc jūs uztraucaties, suka ar vienkāršām lietām, jūs iznākt ģēnijs, mācīties kopā ar mani, nelieli ūdens pilieni veikt okeānu, jums būs eksperts šajā

что это такое, почему ты беспокоишься, освежить с простых вещей, вы будете выходить гений, узнать со мной, маленькие капли воды сделать океан, вы будете экспертом в этом

 hvað er þetta, af hverju ert þú áhyggjur, bursta upp með einföldum hlutum, verður þú að koma út snillingur, læra með mér, litla dropa af vatni gera haf, verður þú að vera sérfræðingur í þessu
C6H8OFrom the molecular formula, the compound has “3 degrees of unsaturation” (3 double bonds or rings). 

structure

cyclohexanon-2-ene

C13 Spectrum
The 13C NMR has 6 peaks, three triplets (CH2 groups) at  46, 30 and 41, two doublets (CH groups), at  129 and 145, and one singlet at  198. Since the molecule has six carbons and six 13C NMR peaks, there must be no symmetry. The singlet at  198 is in the carbonyl region, most likely an aldehyde or ketone. Two of the three CH2 groups are shifted by electronegative groups, suggesting a X-CH2-CH2-CH2-Y unit. The doublets at  129 and 145 are in the alkene region, suggesting a -CHCH- group. The three degrees of unsaturation suggests that the molecule also has a ring.

这是什么,你为什么担心,刷了简单的事情,你会出来的天才,学我,小水珠做出的海洋,你将在这方面的专家

 당신이 걱정하는 이유는 간단한 것들로 브러시, 무엇인가, 당신은 천재 나올 나와 함께 배울 것, 물 작은 방울은 바다를 만들어,이 분야의 전문가가 될 것입니다
 
  • Electronegative groups are “deshielding” and tend to move NMR signals from attached carbons further “downfield” (to higher ppm values). 
  • The -system of alkenes, aromatic compounds and carbonyls strongly deshield C nuclei and move them “downfield” to higher ppm values. 
  • Carbonyl carbons are strongly deshielded and occur at very high ppm values. Within this group, carboxylic acids and esters tend to have the smaller  values, while ketones and aldehydes have values  200.
The 13C chemical shift is dependent both on the presence of electronegative groups and on the steric environment. This is best demonstrated by examining a variety of hexane isomers:

 

Simple interior (primary and secondary) carbons tend to be in the range  25 – 45. Methyl groups which terminate unbranched alkyl chains, however, are significantly shielded (moved to lower  values), as shown by the examples above ( 14, 14.3 and 8.7). The origin of this effect is thought to be steric compression in the gamma () position due to gauche interactions. This is shown schematically below and the gamma position is marked above in the example for hexane.

 

The presence of an electronegative atom such as oxygen tends to move the chemical shift of the Œ-carbon down into the region  65 – 90, as shown in the examples below:

 

Halogens, however, have effects which are difficult to predict and carbons adjacent to halogens tend to have chemical shifts in the  30 – 50 region, as shown below. The effects are not simply additive, however, and multiple substitution can often be shielding (move the signal to lower  values). The nitrile carbon is significantly shielding and adjacent carbons tend to occur in the  20 – 25 region.

 

Alkene carbons tend to have chemical shifts in the range  110 – 140, as shown in the examples below. Conjugation between alkene centers has little effect, as demonstrated by the two middle structures shown below. Conjugation with an oxygen, however, has a dramatic shielding effect, which is attributed to contributions from the resonance forms shown below.


Alkyne carbons occur in the region  65 -85, and are significantly shielding to the carbons which are immediately adjacent ( 1.5 for the terminal methyl of 2-pentyne).

 

Carbonyls are the most highly deshielded carbons which are typically encountered. Their intensity is usually weak, since there are no attached hydrogens to contribute to the Nuclear Overhauser Effect enhancement (with the exception of aldehydes). Typical chemical shifts occur in the region  170 – 210 with esters, carboxylic acids and amides at the low end, and simple ketones and aldehydes at the high end of the range.

 

Aromatic carbons have chemical shifts in the range  120 – 140 and are shifted within this range by the nature of the attached substituent. The multiplicity of aromatic peaks in the non-decoupled spectrum is useful for identifying aromatic substitution patterns.

آپ پریشان کیوں ہیں اس سادہ چیزوں کے ساتھ برش،، کیا ہے، آپ، ہوشیار باہر آ میرے ساتھ سیکھ جائے گی، پانی کے چھوٹے چھوٹے قطرے ایک سمندر بنانے کے لئے، آپ کو اس میں ایک ماہر ہو جائے گا

ANTHONY MELVIN CRASTO

THANKS AND REGARD’S
DR ANTHONY MELVIN CRASTO Ph.D

amcrasto@gmail.com

MOBILE-+91 9323115463
GLENMARK SCIENTIST ,  INDIA
web link
http://anthonycrasto.jimdo.com/
Congratulations! Your presentation titled “Anthony Crasto Glenmark scientist, helping millions with websites” has just crossed MILLION views.
アンソニー     安东尼   Энтони    안토니     أنتوني
join my process development group on google
you can post articles and will be administered by me on the google group which is very popular across the world
LinkedIn group
 
blogs are
 
shark

 

 

 


Filed under: Uncategorized Tagged: NMR

Dacomitinib in phase 3 for lung (non-small cell) (NSCLC) Cancer

$
0
0

File:Dacomitinib.svg

 

Dacomitinib

(2E)-N-{4-[(3-Chloro-4-fluorophenyl)amino]-7-methoxy-6-quinazolinyl}-4-(1-piperidinyl)-2-butenamide

4-Piperidin-1-yl-but-2-enoic acid [4-(3-chloro-4-fluoro-phenylamino)-7-methoxy-quinazolin-6-yl]-amide 

4-Piperidin-1-yl-but-2-enoic acid [4-(3-chloro-4-fluoro-phenylamino)-7-methoxy-quinazolin-6-yl]-amide

1042385-75-0

pf299804…… pfizer

EGFR (HER1; erbB1) Inhibitors
HER4 (erbB4) Inhibitors
HER2 (erbB2) Inhibitors 

  • Molecular formula:C24H25ClFN5O2
  • Molecular mass:469.95

Dacomitinib (PF-00299804) is an experimental drug candidate under development by Pfizer for the treatment of non-small-cell lung carcinoma. It is a selective and irreversible inhibitor of EGFR.[1]

Dacomitinib has advanced to several Phase III clinical trials. The results of the first trials were disappointing, with a failure to meet the study goals,[2][3][4] Additional Phase III trials are ongoing.[2]

Dacomitinib is a HER (erbB) inhibitor in clinical trial development at Pfizer for the treatment of advanced non-small cell lung cancer (NSCLC) and for the treatment of relapsed/recurrent glioblastoma.

No recent development has been reported for research into the treatment of recurrent and/or metastatic head and neck squamous cell cancer. In 2012, Pfizer and SFJ Pharmaceuticals signed a codevelopment agreement for dacomitinib for the treatment of patients with locally advanced or metastatic NSCLC with activating mutations of epidermal growth factor receptor.

 

 

Substituted 4-phenylamino-quinazolin-6-yl-amides useful in the treatment of cancer have been described in the art, including those of U.S. Pat. No. 5,457,105 (Barker), U.S. Pat. No. 5,760,041 (Wissner et al.), U.S. Pat. No. 5,770,599 (Gibson), U.S. Pat. No. 5,929,080 (Frost), U.S. Pat. No. 5,955,464 (Barker), U.S. Pat. No. 6,251,912 (Wissner et al.), U.S. Pat. No. 6,344,455 (Bridges et al.), U.S. Pat. No. 6,344,459 (Bridges et al.), U.S. Pat. No. 6,414,148 (Thomas et al.), U.S. Pat. No. 5,770,599 (Gibson et al.), U.S. patent application 2002/0173509 (Himmelsbach et al.), and U.S. Pat. No. 6,323,209 (Frost).

Dacomitinib is a pan-human epidermal growth factor receptor (pan-HER) inhibitor developed by Pfizer, as ー small molecules targeting ffiR-1, HER-2 and HER-4 tyrosine kinase inhibitor by irreversibly binding to HER-l, HER-2, HER-4 and anti-tumor effect. Ni-line treatment of non-small cell lung cancer (NSCLC) display, Dacomitinib in non-small cell lung cancer Dinner erlotinib compared to some extend on progression-free survival and quality of life have mentioned the smell.

_4] Structural formula for Dacomitinib

Figure CN103304492AD00051

[0005] U.S. patent US7772243 Dacomitinib first proposed a synthesis method, first, a fluorine-2_ _4_ amino acid and formamidine ring closure reaction to give 7 – fluoro-4 – quinazolinone, nitration and then successively chlorination reaction, to give 4 – chloro-7 – fluoro-6 – nitro-quinazoline; another aspect ー 3 – chloro-4 – amino-substituted on a fluoroaniline to give 3 – chloro – # – (3,4 – ni section yl methoxy)-4_ fluoro-aniline, obtained after the coupling of both an amino-protected N-(3 – chloro-4 – fluorophenyl)-7 – fluoro-6 – nitro-quinazoline -4 – amine, protected amino N-(3 – chloro-4 – fluorophenyl

Yl)-7_ fluoro-6 – nitro-quinazolin-4 – amine is of formula

Figure CN103304492AD00052

Followed by a methoxy group, an amidation reaction and hydrogenation, the final deprotection ko under the action of trifluoroacetic acid to give the final product Dacomitinib. Throughout the reaction as follows:

 

synthesis

http://www.google.com/patents/CN103304492A?cl=en

Synthesis ー kind EGFR inhibitors Dacomitinib, synthetic route for

Figure CN103304492AD00071

 

A synthetic method EGFR inhibitors Dacomitinib, concrete steps are as follows:

Step I, 7 – fluoro-4 – Synthesis of quinazolinone:

Figure CN103304492AD00101

30 g (0.1934mol) 2 – fluoro-amino acid was dissolved in 250 ml _4_ formamide among the reaction was heated to 150 ° C for 6 inch, TLC plates to determine the point of completion of the reaction. The reaction was poured hot into 2000 ml of ice water, filtered, the filter cake was washed with water, vacuum dried at 50 ° C for 14 hours to give a pale brown solid powder 7 – fluoro-4 – quinazolinone, 28 g, yield 88%.

[0021] 2 walk 7 – fluoro-6 – nitro-4_ (hydrogen) _ Synthetic quinazolinones of:

Figure CN103304492AD00102

Concentrated sulfuric acid (50 ml) and fuming nitric acid (50 ml) mixture was cooled with an ice bath to (TC hereinafter under stirring slowly added 25 g (0.1523mol) 7 – fluoro-4 – quinazolinone , the addition was complete, the reaction mixture was stirred at room temperature for I hour and then the reaction was heated to 110 ° C for 2 inch, TLC plates to determine the point of completion of the reaction the reaction was cooled to room temperature, 300 ml of ice water, the precipitated solid was stirred for 30 minutes , filtered, the filter cake was washed with water, vacuum dried at 50 ° C in 14 hours to give a yellow solid powder 7 – fluoro-6 – nitro-4 – (hydrogen) – quinazolinone, 26 g, yield 82%.

[0022] Step 3 6 – amino-7 – fluoro-4 – (hydrogen) – quinazolinone Synthesis:

Figure CN103304492AD00103

24 g (0.1148mol) 7 – fluoro-6 – nitro _4_ (hydrogen) – quinazolinone was dissolved in 400 ml of methanol was added 2 g of palladium / carbon catalyst was added 8 ml of concentrated hydrochloric acid, and hydrogen was 2 small inch atmospheric reaction, TLC plates to determine the point of the reaction is complete. The catalyst was removed by suction filtration through celite, washed several fitness methanol, and the filtrate was concentrated by rotary evaporation to dryness to give 6 – amino-7_ fluoro-4 – (hydrogen) – quinazolinone, yellow powder, 20 g, yield 97%.

[0023] 4 walk, ⑶ -4 – (piperidin – Suites yl) -2 – butene acid methyl ester synthesis:

Figure CN103304492AD00104

18 g (0.1006mol) 4 – bromo-methyl crotonate dissolved in 180 ml of methylene chloride ni added 27.9 g (0.2019mol) potassium carbonate, cooled to ice-bath (TC, was slowly added dropwise 10 ml (0.1012mol ) piperidine, (I reaction was stirred under a small inch TC, TLC plates to determine the point of completion of the reaction was concentrated by rotary evaporation to dryness, to give (E) -4 – (piperidin-1 – yl) – 2 – butenoic acid methyl Cool as a yellow solid, 17.1 g, yield 93%.

[0024] 5th walk, Buddhist) -4 – (piperidin-1 – yl) -2 – butene acid hydrochloride synthesis:

Figure CN103304492AD00105

16 g (0.0873mol) of W) -4 – (piperidin-_1_ yl) -2 – butenyl acetate and 80 ml of concentrated hydrochloric acid was added to 250 ml of 1,4 – ni oxygen dioxane, heated under reflux 20 hours inch, TLC plate point the reaction was determined complete, the reaction solution was concentrated by rotary evaporation to dryness surplus was recrystallized from isopropanol to give a pale yellow solid, Buddhist) _4-(piperidin-1 – yl) -2 – butene acid hydrochloride, 14.5 g, yield 81%.

[0025] Step 6, (E) -4 – (piperazine Jie fixed -1 – yl) – 2 – butenyl chloride synthesis:

Figure CN103304492AD00111

13 g (0.0632mol) of (K) ~ 4 ~ (piperidin-1 – yl) -2 – butene acid hydrochloride was dissolved in 750 ml of methylene chloride ni, 5 ml of DMF, was slowly added dropwise 8 ml ( 0.0933mol) of oxalyl chloride, the reaction was stirred at room temperature for I h, TLC plates to determine the point of completion of the reaction, the reaction solution was concentrated to dryness by rotary evaporation to give a pale yellow oil, Buddhist) _4-(piperidin-1 – yl) -2 – butyl allyl chloride, 11.8 g, yield 99%.

[0026] Step 7 (cargo) – # – (7 – fluoro-4 – oxo-3 ,4 – ni hydrogen quinazolin-6 – yl) -4 – (piperidin-1 – yl) -2 – butene amide Synthesis:

Figure CN103304492AD00112

11 g (0.0586mol) of the) -4 – (piperidin-1 – yl) – 2 – butenyl chloride ni chloride (50 ml) was slowly added dropwise to 6 – amino-1 – fluoro-4 – ( hydrogen) – quinazolinone (7 g, 0.0391mmol), three ko amine (14 ml) and the mixture was ni chloride (200 ml), the reaction mixture was stirred at room temperature for 2 hours the reaction inch, TLC determined the completion of reaction points board , was added 800 liters of halo ni halo chloroformate and 500 liters of burning the separated organic phase was washed with 500 liters of halo, halo and then with 500 liters of brine, dried over magnesium sulfate, and concentrated by rotary evaporation to dryness was subjected to silica gel surplus Column chromatography (30% acid ko ko acetate / hexane) to give (M)-N-(7 – fluoro-4 – oxo-3 ,4 – ni hydrogen quinazolin-6 – yl) -4 – (piperidin-1 – yl) -2 – butenamide, as a pale yellow solid, 12.3 g, yield 95%.

Step 8 [0027] (2 ^) – # – (7 – methoxy – 4 – oxo _3, 4_ ni hydrogen quinazolinyl _6_ yl)-4_ (piperidin-1 – yl) – Synthesis 2_ butenamide:

Figure CN103304492AD00113

I ^ xN MeONa N. Under nitrogen atmosphere, to 100 ml of anhydrous methanol was slowly added 1.52 g of sodium metal (0.0661mol), stirred for 10 minutes to dissolve all of the sodium metal to the completion of the reaction, to obtain a freshly prepared solution of sodium methoxide, and the The sodium methoxide solution was added 11 g (0.0333mol) of (receive) (7 – fluoro-4 – oxo-3 ,4 – ni hydrogen quinazolin-6 – yl) -4 – (piperidin-1 – yl) 2_ butene-amide, the reaction was heated to reflux for 3 inch, TLC plates to determine completion of the reaction point, cooled to room temperature, acidified with 2N hydrochloric acid solution to pH = 3 ~ 4, and concentrated by rotary evaporation to dryness, the residue was washed with water beating, filtration, The filter cake was washed with water, vacuum dried at 50 ° C in 14 hours to give (article) – # – (7 – methoxy – 4 – oxo – ni hydrogen quinazolin-6 – yl) – 4_ (piperidin-1 – yl)-2_ butenamide yellow solid, 10.6 g, yield 93%.

[0028] Step 9, {W,-N-(4 – chloro-7 – methoxy-quinazoline _6_ yl)-4_ (piperidin _1_ yl)-amide <EMI butene 2_:

Figure CN103304492AD00121

9 g (0.0263mol) of (receive) – # – (7 – methoxy _4_ oxo – ni hydrogen quinazolin-6 – yl)-4_ (piperidin-1 – yl) – 2_ butenamide were added to 40 ml of phosphorus oxychloride was heated under reflux for 2 inch, TLC plates to determine the point of completion of the reaction, the reaction solution was concentrated to dryness by rotary evaporation, ice water was added surplus, beating, filtered, the cake washed with washed with water, vacuum dried at 50 ° C in 14 hours to give {W,-N-(4 – chloro-7 – methoxy-quinazolin-6 – yl) -4 – (piperidin-1 – yl) – 2 – butene amide as a yellow solid, 7 g, yield 74%

Figure CN103304492AD00122

(2E)-N-(4 – chloro-7 – methoxy-quinazolin-6 – yl) -4 – (piperidin-1 – yl) -2 – butene amide (6 g,

0.0166mol), 3 – chloro-4-fluoro-aniline (2.6 g, 0.0179mol) and three ko amine (2.6 ml, 0.0186mol) was added to 140 ml of isopropanol and the reaction was heated to reflux for 3 inch, TLC plates to determine the point completion of the reaction, cooled to room temperature, filtered, the filter cake washed with methanol, vacuum dried at 50 ° C in 14 hours to give the final product Dacomitinib, a yellow solid, 6.6 g, yield 84%.

 

/////////////////////////

synthesis

US7772243

http://www.google.com/patents/US7772243

 Scheme 1, wherein the 4-position aniline group is represented a 4-fluoro-3-chloro aniline group.

Figure US07772243-20100810-C00005

4-Chloro-7-fluoro-6-nitroquinazoline (7) can be prepared by methods similar to those described in J.Med. Chem. 1996, 39, 918-928. Generally, 2-amino-4-fluoro-benzoic acid (1) can be reacted with formamidine (2) and acetic acid (3) in the presence of 2-methoxyethanol to provide 7-Fluoro-3H-quinazolin-4-one (4). The 7-fluoro-3H-quinazolin-4-one (4) can be nitrated to 7-fluoro-6-nitro-3H-quinazolin-4-one (5), which can be treated with thionyl chloride to yield 4-chloro-6-nitro-7-fluoro-3H-quinazoline (6). The 4-chloro-quinazoline compound (6) can be combined with a desirably substituted aniline, represented above by 4-fluoro-3-chloro-aniline, in the presence of a tertiary amine and isopropanol to provide the 4-anilino-6-nitro-7-fluoro-quinazoline (7).

The 4-anilino-6-nitro-7-fluoro-quinazoline (7) may be reacted with an alcohol of the formula R3OH, wherein Ris as defined above, to yield the 7-alkoxylated compound (8). Reduction of the 6-nitro compound (8) provides the 6-amino analog (9).

The 6-position amino compound (9) may be reacted with a haloalkenoyl chloride (12), such as a 4-bromo-but-2-enoyl chloride, 5-bromo-pent-2-enoyl chloride, 4-chloro-but-2-enoyl chloride, or 5-chloro-pent-2-enoyl chloride, to provide an alkenoic acid[4-anilino]-7-alkoxylated-quinazolin-6-yl-amide (13). Haloalkenoyl chloride agents useful in this scheme may be prepared by methods known in the art, such as the treatment of a relevant haloalkenoic acid, represented by bromoalkenoic acid ester (10), with a primary alcohol, yielding the corresponding haloalkenoic acid (11), which may in turn be treated with oxalyl chloride to provide the desired haloalkenoyl chloride (12).

Finally, the quinazoline-6-alkanoic acid compound (13) may be treated with a cyclic amine, such as piperidine, piperazine, etc., to provide the desired final compound (14).

EXAMPLE 2

4-Piperidin-1-yl-but-2-enoic acid [4-(3-chloro-4-fluoro-phenylamino)-7-methoxy-quinazolin-6-yl]-amide (Synthetic Route No. 1)

 

Figure US07772243-20100810-C00008

 

The title compound and other 7-methoxy analogs of this invention can be prepared as described in Example 1 by replacing the 2-fluoroethanol used in Example 1 with stoichiometric amount of methanol.

EXAMPLE 3 4-Piperidin-1 -yl-but-2-enoic acid [4-(3-chloro-4-fluoro-phenylamino)-7-methoxv -quinazolin-6-yl]-amide (Synthetic Route No. 2)

An alternative synthetic route for compounds of this invention involves preparing the 6-position substituent chain as a Het-alkenoyl chloride as depicted in Scheme 2, below.

Figure US07772243-20100810-C00009
Figure US07772243-20100810-C00010

It will be understood that other compounds within this invention may be prepared using Het-butenoyl halide, Het-pentenoyl halide and Het-hexenoyl halide groups of the formula:

Figure US07772243-20100810-C00011


wherein Ris as described herein and halo represents F, Cl, Br or I, preferably Cl or Br. One specific group of these Het-alkenoyl halides includes those compounds in which halo is Cl or Br, Ris —(CH2)m-Het, m is an integer from 1 to 3, and Het is piperidine or the substituted piperidine moieties disclosed above.

EXAMPLE 4 4-Piperidin-1-yl-but-2-enoic acid [4-(3-chloro-4-fluoro-phenylamino)-7-methoxy-quinazolin-6-yl]-amide (Synthetic Route No. 3)

Figure US07772243-20100810-C00012
Figure US07772243-20100810-C00013

3-Chloro-4-fluoro-phenylamine 15 (50.31, 345.6 mmole) and 3,4-Dimethoxy-benzaldehyde 16 (57.43 g, 345.6 mmole) were mixed in 500 ml of IPA and cooled in an ice-water. The glacial acetic acid was added (20.76 g, 345.6 mole) and then sodium cyanoborohydride in one portion. The reaction was stirred at room temperature (RT) for 24 hrs. 250 mL of 10% NaOH was added dropwise at RT after the reaction was completed. The mixture was stirred for ½ hr. The slurry was then filtered and washed with IPA and dried in vacuo. The mass weight 88.75 g (17, 87%).

Compounds 6 (3 g, 13.18 mmole) and 17 (3.9 g, 13.18 mmole) were combined in CH3CN (25 mL) and heated for one hr. Mass spectroscopy indicated no starting material. Saturated K2COwas added and the reaction was extracted 3× with EtOAc. The organic layers were combined, washed with brine and concentrated in vacuo to give 6.48 g of 7 (78.4%).

Compound 7 (72.76 g, 149.4 mmole) was added to a cool solution of NaOMe in 1.5 L of dry MeOH under N2. The cooling bath was removed and the mixture was heated to reflux and stirred for 1 hr. The reaction was cooled to room temperature and quenched with water until the product precipitated out. The solid was filtered and washed with water and hexanes. The product was slurred in refluxing EtOAc and filtered hot to provide 68.75 g of yellow soled 8 (73%).

Compound 8 (63.62 g, 127.5 mole) was hydrogenated using Raney/Ni as catalyst to obtain 43.82 g of 9 (100%). Oxalyl chloride (6.5 g, 51.18 mmole) was added slowly to a suspension of 13 (10.5 g, 51.2 mmole) in 200 ml of dichloromethane containing 8 drops of DMF, after the reaction become homogeneous, the solvent was removed and the residual light yellow solid was slurred in 200 ml of DMAC and 9 (20 g, 42.65 mmole) was added gradually as a solid. The reaction was stirred for 15 min. and poured slowly into 1N NaOH. The mixture was extrated 3× EtOAc. The combined organic layers were washed with brine, filtered and concentrated in vacuo to obtain 28.4 g (100%) 10.

Compound 10(13.07 g, 21.08 mmole) was dissolved in trifluoroacetic acid (TFA) (74 g, 649 mmole) and heated to 30° C. for 24 hrs. The reaction was cooled to RT and poured gradually into a cooled 1 N NaO H-brine solution. Precipitate formed and was filtered and washed with 3X water then dried. The precipitate was recrystallized from toluene to obtain pure 4-Piperidin-1-vl-but-2-enoic acid [4-(3-chloro-4-fluoro-phenylamino) -7-methoxv-puinazolin-6-yl]-amide (9.90 g, 89%).

Example 1 is similar but not same…caution

EXAMPLE 1 4-Piperidin-1-yl-but-2-enoic acid [4-(3-chloro-4-fluoro-phenylamino)-7-(2-fluoro-ethoxy)-quinazolin-6-yl]-amide

 

Figure US07772243-20100810-C00007

 

7-fluoro-6-nitro-4-chloroquinazoline (14.73,g, 65 mmol) was combined with 3-choro-4-fluoroaniline (9.49 g, 65 mmol) and triethylamine (10 mL, 72 mmol) in 150 mL of isopropanol. The reaction was stirred at room temperature for 1.5 hours, resulting in a yellow slurry. The solid was collected by filtration, rinsing with isopropanol and then water. The solid was dried in a 40° C. vacuum oven overnight to give 19.83 g (91%) of the product as an orange solid.

MS (APCI, m/z, M+1): 337.0

NaH (60% in mineral oil, 3.55 g, 88 mmol) was added, in portions, to a solution of 2-fluoroethanol (5.19 g, 80 mmol) in 200 mL THF. The reaction was stirred for 60 minutes at room temperature. To the reaction was added 7-fluoro-6-nitro-4-(3-chloro-4-fluoroaniline)quinazoline (18.11 g, 54 mmol) as a solid, rinsing with THF. The reaction was heated to 65° C. for 26 hours. The reaction was cooled to room temperature and quenched with water. THF was removed in vacuo. The resulting residue was sonicated briefly in water then the solid collected by filtration. The solid was triturated with MeOH, filtered and dried in a 40° C. vacuum oven overnight to 12.63 g of the product. Additional product was obtained by concentrating the MeOH filtrate to dryness and chromatography eluting with 50% EtOAc/hex. The isolated material was triturated with MeOH (2×), filtered and dried. 3.90 g

Total yield: 16.53 g, 81%

MS (APCI, m/z, M+1): 381.0

7-(2-fluoroethoxy)-6-nitro-4-(3-chloro-4-fluoroaniline)quinazoline (0.845 g, 2.2 mmol) in 50 mL THF was hydrogenated with Raney nickel (0.5 g) as the catalyst over 15 hours. The catalyst was filtered off and the filtrate was evaporated to give 0.77 g of product. (99%)

MS (APCI, m/z, M+1): 351.2

Methyl 4-bromocrotonate (85%, 20 mL, 144 mmol) was hydrolyzed with Ba(OH)in EtOH/H2O as described in J.Med.Chem. 2001, 44(17), 2729-2734.

MS (APCI, m/z, M−1): 163.0

To a solution of 4-bromocrotonic acid (4.17 g, 25 mmol) in CH2Cl(20 mL) was added oxalyl chloride (33 mL, 38 mmoL) and several drops of DMF. The reaction was stirred at room temperature for 1.5 hours. The solvent and excess reagent was removed in vacuo. The resulting residue was dissolved in 10 mL THF and added to a 0° C. mixture of 6-amino-7-(2-fluoroethoxy)-4-(3-chloro-4-fluoroaniline)quinazoline (5.28 g, 15 mmol) and triethylamine (5.2 mL, 37 mmol). The reaction was stirred at 0° C. for 1 hour. Water was added to the reaction and the THF removed in vacuo. The product was extracted into CH2Cl(400 mL). The organic layer was dried over MgSO4, filtered and concentrated. The crude material was chromatographed on silica gel eluting with 0-4% MeOH/CH2Cl2. An isolated gold foam was isolated. Yield: 4.58 g, 61%

MS (APCI, m/z, M−1): 497.1

Piperidine (0.75 mL, 6.7 mmol) was added to a solution of the above compound (3.35 g, 6.7 mmol) and TEA (2.80 mL, 20 mmol) in 10 mL DMA at 0° C. The reaction was stirred at 0° C. for 17 hours. Water was added to the reaction until a precipitate was evident. The reaction was sonicated for 40 minutes and the liquid decanted. The residue was dissolved in CH2Cl2, dried over MgSO4, filtered and concentrated. The material was chromatographed on silica gel eluting with 4-10% MeOH/CH2Cl2. The isolated residue was triturated with acetonitrile (2×) and collected by filtration. Impurity found: Michael addition of piperidine (2.2% in first trituration of acetonitrile). Additional material can be obtained from the acetonitrile filtrates.

Yield: 0.95 g, 27%

MS (APCI, m/z, M+1): 502.3

 

……………

US 20050250761 A1, 

References

  1.  “Dacomitinib”. NCI Drug Dictionary.
  2.  Zosia Chustecka (January 27, 2014). “Dacomitinib Fails in Pretreated Non-small Cell Lung Cancer”. Medscape.
  3.  “Blow to Pfizer as dacomitinib fails in lung cancer trials”. pmlive.com. 28th January 2014.
  4.  “Pfizer Announces Top-Line Results From Two Phase 3 Trials Of Dacomitinib In Patients With Refractory Advanced Non-Small Cell Lung Cancer”. Pfizer Press Release. January 27, 2014.
  5. Tyrosine kinase inhibitors.17. Irreversible inhibitors of the epidermal growth factor receptor: 4-(Phenylamino)quinazoline- and 4-(phenylamino)pyrido[3,2-d]pyrimidine-6-acrylamides baring additional solubilizing functions
    J Med Chem 2000, 43(7): 1380

 

WO1996033980A1 * 23 Apr 1996 31 Oct 1996 Keith Hopkinson Gibson Quinazoline derivatives
WO1997038983A1 * 8 Apr 1997 23 Oct 1997 Alexander James Bridges Irreversible inhibitors of tyrosine kinases
WO2002050043A1 * 12 Dec 2001 27 Jun 2002 Boehringer Ingelheim Pharma Quinazoline derivatives, medicaments containing said compounds, their utilization and method for the production thereof
WO2004069791A2 * 3 Feb 2004 19 Aug 2004 Hubert Gangolf Klemens Barth Preparation of substituted quinazolines
US5760041 * 21 Jan 1997 2 Jun 1998 American Cyanamid Company 4-aminoquinazoline EGFR Inhibitors

Filed under: Phase3 drugs Tagged: Dacomitinib, PHASE 3

‘Shocking’ study gives hope to paralyzed people; shows electricity helps them stand, move legs

$
0
0

Originally posted on Atasteofcreole's Blog:

http://www.nydailynews.com/life-style/health/shocking-study-hope-paralyzed-people-article-1.1749234

‘It could still be a life-changer for them,’ experts say, but they also add that while this breakthrough is promising, ‘there is no miracle cure on the way’ for paralysis.

Electricity may provide hope to men and women who suffer paralysis.

Three years ago, doctors reported that zapping a paralyzed man’s spinal cord with electricity allowed him to stand and move his legs. Now they’ve done the same with three other patients, suggesting their original success was no fluke.

“This is wonderful news. Spinal cord injury need no longer be a lifelong sentence of paralysis,” said Dr. Roderic Pettigrew, director of the National Institute of Biomedical Imaging and Bioengineering, one of the National Institutes of Health, according to NBC News. “It is just downright marvelous.”

“The big message here is that people with spinal cord injury of the type these men had no longer need to think they have…

View original 773 more words


Filed under: Uncategorized

EU and Australia link up on orphan drugs


DRUG PROCESS CHEMISTRY

$
0
0

 

Process chemistry is the arm of pharmaceutical chemistry concerned with the development and optimization of a synthetic scheme and pilot plant procedure to manufacture compounds for the drug development phase. Process chemistry is distinguished from medicinal chemistry, which is the arm of pharmaceutical chemistry tasked with designing and synthesizing molecules on small scale in the early drug discovery phase.

Medicinal chemists are largely concerned with synthesizing a large number of compounds as quickly as possible from easily tunable chemical building blocks (usually for SAR studies). In general, the repertoire of reactions utilized in discovery chemistry is somewhat narrow (for example, the Buchwald-Hartwig amination, Suzuki coupling and reductive amination are commonplace reactions).[1] In contrast, process chemists are tasked with identifying a chemical process that is safe, cost and labor efficient, “green,” and reproducible, among other considerations.

Oftentimes, in searching for the shortest, most efficient synthetic route, process chemists must devise creative synthetic solutions that eliminate costly functional group manipulations and oxidation/reduction steps.

This article will focus exclusively on the chemical and manufacturing processes associated with the production of small molecule drugs. Biological medical products (more commonly called “biologics”) represent a growing proportion of approved therapies, but the manufacturing processes of these products are beyond the scope of this article.

Additionally, the many complex factors associated with chemical plant engineering (for example, heat transfer and reactor design) and drug formulation will be treated cursorily.

Process Chemistry Considerations

Cost efficiency is of paramount importance in process chemistry and, consequently, is a focus in the consideration of pilot plant synthetic routes. The drug substance that is manufactured, prior to formulation, is commonly referred to as the active pharmaceutical ingradient (API) and will be referred to as such herein.

API production cost can be broken into two components: the “material cost” and the “conversion cost.”[2] The ecological and environmental impact of a synthetic process should also be evaluated by an appropriate metric (e.g. the EcoScale).

An ideal process chemical route will score well in each of these metrics, but inevitably tradeoffs are to be expected. Most large pharmaceutical process chemistry and manufacturing divisions have devised weighted quantitative schemes to measure the overall attractiveness of a given synthetic route over another. As cost is a major driver, material cost and volume-time output are typically weighted heavily.

The chemical and processing industries (CPI) provide the building blocks for many products. By using large amounts of heat and energy to physically or chemically transform materials, these industries help meet the world’s most fundamental needs for food, shelter and health, as well as products that are vital to such advanced technologies as computing, telecommunications and biotechnology. 

These industries face major challenges to meet the needs of the present without compromising the needs of the future generations in the face of increasing industrial competitiveness. This translates into the need to make processes much more energy efficient, safer and more flexible, and to reduce emissions to meet the many competitive challenges within a global economy.

The chemical and processing industries refer to processes where materials undergo chemical conversion during their production into finished products, as well as – or instead of – the physical conversions common to industry in general. 

In the chemical process industry the products differ chemically from the raw materials as a result of undergoing one or more chemical reactions during the manufacturing process.

The chemical process industries broadly include the traditional chemical industries, both organic and inorganic; the petroleum industry; the petrochemical industry, which produces the majority of plastics, synthetic fibers, and synthetic rubber from petroleum and natural-gas raw materials; and a series of allied industries in which chemical processing plays a substantial part. 

While the chemical process industries are primarily the realm of the chemical engineer and the chemist, they also involve a wide range of other scientific, engineering, and economic specialists.

Material Cost

The material cost of a chemical process is the sum of the costs of all raw materials, intermediates, reagents, solvents and catalysts procured from external vendors. Material costs may influence the selection of one synthetic route over another or the decision to outsource production of an intermediate.

Conversion Cost

The conversion cost of a chemical process is a factor of that procedure’s overall efficiency, both in materials and time, and its reproducibility. The efficiency of a chemical process can be quantified by its atom economy, yield, volume-time output, and environmental factor (E-factor), and its reproducibility can be evaluated by the Quality Service Level (QSL) and Process Excellence Index (PEI) metrics.

 

An illustrative example of atom economy using the Claisen rearrangement and Wittig reaction.

Atom Economy

The atom economy of a reaction is defined as the number of atoms from the starting materials that are incorporated into the final product. Atom economy can be viewed as an indicator of the “efficiency” of a given synthetic route.[3]

 
AE = \frac{\text{MW(product)}}{\sum \text{MW(raw materials)}}*100%

 

For example, the Claisen rearrangement and the Diels-Alder cycloaddition are examples of reaction that are 100 percent atom economical. On the other hand, a prototypical Wittig reaction has especially poor atom economy (merely 20 percent in the example shown).

Process synthetic routes should be designed such that atom economy is maximized for the entire synthetic scheme. Consequently, “costly” reagents such as protecting groups and high molecular weight leaving groups should be avoided where possible. An atom economy value in the range of 70 to 90 percent for an API synthesis is ideal, but it may be impractical or impossible to access certain complex targets within this range. Nevertheless, atom economy is a good metric to compare two routes to the same molecule.

Yield

Yield is defined as the amount of product obtained in a chemical reaction. According to Vogel’s Textbook of Practical Organic Chemistry, yields around 100% are called quantitative, yields above 90% are excellent, yields above 80% are very good, yields above 70% are good, yields above 50% are fair, and yields below 40% are poor. The yield that has practical significance in a process chemistry setting is the isolated yield, referring to the yield of the isolated product after all extraction and purification steps. In a final API synthesis, isolated yields of 80 percent or above for each synthetic step are expected.

 

 

An illustrative example of convergent synthesis.

There are several strategies that are employed in the design of a process route to ensure adequate overall yield of the pharmaceutical product. The first is the concept of convergent synthesis. Assuming a very good to excellent yield in each synthetic step, the overall yield of a multistep reaction can be maximized by combining several key intermediates at a late stage that are prepared independently from each other.

Another strategy to maximize isolated yield (as well as time efficiency) is the concept of telescoping synthesis (also called one-pot synthesis). This approach describes the process of eliminating workup and purification steps from a reaction sequence, typically by simply adding reagents sequentially to a reactor. In this way, unnecessary losses from these steps can be avoided.

Finally, to minimize overall cost, synthetic steps involving expensive reagents, solvents or catalysts should be designed into the process route as late stage as possible, to minimize the amount of reagent used.

In a pilot plant or manufacturing plant setting, yield can have a profound effect on the material cost of an API synthesis, so the careful planning of a robust route and the fine-tuning of reaction conditions are crucially important. After a synthetic route has been selected, process chemists will subject each step to exhaustive optimization in order to maximize overall yield. Low yields are typically indicative of unwanted side product formation, which can raise red flags in the regulatory process as well as pose challenges for reactor cleaning operations.

Volume-Time Output

The volume-time output (VTO) of a chemical process represents the cost of occupancy of a chemical reactor for a particular process or API synthesis. For example, a high VTO indicates that a particular synthetic step is costly in terms of “reactor hours” used for a given output. Mathematically, the VTO for a particular process is calculated by the total volume of all reactors (m3) that are occupied times the hours per batch divided by the output for that batch of API or intermediate (measured in kg).

VTO=\frac{\text{nominal volume of all reactors} [m^3]*\text{time per batch} [h]}{\text{output per step} [kg]}

The process chemistry group at Boehringer-Ingelheim, for example, targets a VTO of less than 1 for any given synthetic step or chemical process.

Additionally, the raw conversion cost of an API synthesis (in dollars per batch) can be calculated from the VTO, given the operating cost and usable capacity of a particular reactor. Oftentimes, for large-volume APIs, it is economical to build a dedicated production plant rather than to use space in general pilot plants or manufacturing plants.

Environmental Factor (E-factor) and Process Mass Intensity (PMI)

Both of these measures, which capture the environmental impact of a synthetic reaction, intend to capture the significant and rising cost of waste disposal in the manufacturing process. The E-factor for an entire API process is computed by the ratio of the total mass of waste generated in the synthetic scheme to the mass of product isolated.
E=\frac{\sum \text{mass of waste}}{\text{mass of isolated product}}=\frac{\sum \text{mass of materials}-\text{mass of isolated product}}{\text{mass of isolated product}}
A similar measure, the process mass intensity (PMI) calculates the ratio of the total mass of materials to the mass of the isolated product.
PMI=\frac{\sum \text{mass of materials}}{\text{mass of isolated product}}
For both metrics, all materials used in all synthetic steps, including reaction and workup solvents, reagents and catalysts, are counted, even if solvents or catalysts are recycled in practice. Inconsistencies in E-factor or PMI computations may arise when choosing to consider the waste associated with the synthesis of outsourced intermediates or common reagents. Additionally, the environmental impact of the generated waste is ignored in this calculation; therefore, the environmental quotient (EQ) metric was devised, which multiplies the E-factor by an “unfriendliness quotient” associated with various waste streams. A reasonable target for the E-factor or PMI of a single synthetic step is any value between 10 and 40.

Quality Service Level (QSL)

The final two “conversion cost” considerations involve the reproducibility of a given reaction or API synthesis route. The quality service level (QSL) is a measure of the reproducibility of the quality of the isolated intermediate or final API. While the details of computing this value are slightly nuanced and unimportant for the purposes of this article, in essence, the calculation involves the ratio of satisfactory quality batches to the total number of batches. A reasonable QSL target is 98 to 100 percent.

Process Excellence Index (PEI)

Like the QSL, the process excellence index (PEI) is a measure of process reproducibility. Here, however, the robustness of the procedure is evaluated in terms of yield and cycle time of various operations. The PEI yield is defined as follows:

\text{PEI yield}=\frac{\text{average yield}*100%}{\text{aspiration level yield}}=\frac{\text{average yield}*100%}{\frac{\text{median yield}+\text{best yield}}{2}}
In practice, if a process is high-yielding and has a narrow distribution of yield outcomes, then the PEI should be very high. Processes that are not easily reproducible may have a higher aspiration level yield and a lower average yield, lowering the PEI yield.

Similarly, a PEI cycle time may be defined as follows:

\text{PEI cycle time}=\frac{\text{aspiration level cycle time}*100%}{\text{average cycle time}}=\frac{\frac{\text{median cycle time}+\text{best cycle time}}{2}}{\text{average cycle time}}
For this expression, the terms are inverted to reflect the desirability of shorter cycle times (as opposed to higher yields). The reproducibility of cycle times for critical processes such as reaction, centrifugation or drying may be critical if these operations are rate-limiting in the manufacturing plant setting. For example, if an isolation step is particularly difficult or slow, it could become the bottleneck for an API synthesis, in which case the reproducibility and optimization of that operation become critical.

For an API manufacturing process, all PEI metrics (yield and cycle times) should be targeted at 98 to 100 percent.

EcoScale

In 2006, Van Aken, et al.[4] developed a quantitative framework to evaluate the safety and ecological impact of a chemical process, as well as minor weighting of practical and economical considerations. Others have modified this EcoScale by adding, subtracting and adjusting the weighting of various metrics. Among other factors, the EcoScale takes into account the toxicity, flammability and explosive stability of reagents used, any nonstandard or potentially hazardous reaction conditions (for example, elevated pressure or inert atmosphere), and reaction temperature. Some EcoScale criteria are redundant with previously considered criteria (e.g. E-factor).

Synthetic Case Studies

Boehringer Ingelheim HCV Protease Inhibitor (BI 201302)

Macrocyclization is a recurrent challenge for process chemists, and large pharmaceutical companies have necessarily developed creative strategies to overcome these inherent limitations. An interesting case study in this area involves the development of novel NS3 protease inhibitors to treat Hepatitis C patients by scientists at Boehringer-Ingelheim.[5] The process chemistry team at BI was tasked with developing a cheaper and more efficient route to the active NS3 inhibitor BI 201302, a close analog of BILN 2061. Two significant shortcomings were immediately identified with the initial scale-up route to BILN 2061, depicted in the scheme below.[6] The macrocyclization step posed four challenges inherent to the cross-metathesis reaction.

  1. High dilution is typically necessary to prevent unwanted dimerization and oligomerization of the diene starting material. In a pilot plant setting, however, a high dilution factor translates into lower throughput, higher solvent costs and higher waste costs.
  2. High catalyst loading was found to be necessary to drive the RCM reaction to completion. Because of high licencing costs of the ruthenium catalyst that was used (1st generation Hoveyda catalyst), a high catalyst loading was financially prohibitive. Recycling of the catalyst was explored, but proved impractical.
  3. Long reaction times were necessary for reaction completion, due to slow kinetics of the reaction using the selected catalyst. It was hypothesized that this limitation could be overcome using a more active catalyst. However, while the second-generation Hoveyda and Grubbs catalysts were kinetically more active than the first-generation catalyst, reactions using these catalysts formed large amounts of dimeric and oligomeric products.
  4. An epimerization risk under the cross-methathesis reaction conditions. The process chemistry group at Boehringer-Ingelheim performed extensive mecahnistic studies showing that epimerization most likely occurs through a ruthenacyclopentene intermediate.[7] Furthermore, the Hoveyda catalyst employed in this scheme minimizes epimerization risk compared with the alalogous Grubbs catalyst.

Additionally, the final double SN2 sequence to install the quinoline heterocycle was identified as a secondary inefficiency in the synthetic route.

BILN 2061 Synthetic Scheme.pdf

Analysis of the cross-methathesis reaction revealed that the conformation of the acyclic precursor had a profound impact on the formation of dimers and oligomers in the reaction mixture. By installing a Boc protecting group at the C-4 amide nitrogen, the Boehringer-Ingelheim chemists were able to shift the site of initiation from the vinylcyclopropane moiety to the nonenoic acid moiety, improving the rate of the intramolecular reaction and decreasing the risk of epimerization. Additionally, the catalyst employed was switched from the expensive 1st generation Hoveyda catalyst to the more reactiveless expensive Grela catalyst.[8] These modifications allowed the process chemists to run the reaction at a standard reaction dilution of 0.1-0.2 M, given that the rates of competing dimerization and oligomerization reactions was so dramatically reduced.

Additionally, the process chemistry team envisioned a SNAr strategy to install the quinoline heterocycle, instead of the SN2 strategy that they had employed for the synthesis of BILN 2061. This modification prevented the need for inefficient double inversion by proceeding through retention of stereochemistry at the C-4 position of the hydroxyproline moiety.[9]

BI 201302 Synthetic Scheme.pdf

It is interesting to examine this case study from a VTO perspective. For the unoptimized cross-metathesis reaction using the Grela catalyst at 0.01 M diene, the reaction yield was determined to be 82 percent after a reaction and workup time of 48 hours. A 6-cubic meter reactor filled to 80% capacity afforded 35 kg of desired product. For the unoptimized reaction:

VTO Initial Route BI 201302.png

This VTO value was considered prohibitively high and a steep investment in a dedicated plant would have been necessary even before launching Phase III trials with this API, given its large projected annual demand. But after reaction development and optimization, the process team was able to improve the reaction yield to 93 percent after just 1 hour (plus 12 hours for workup and reactor cleaning time) at a diene concentration of 0.2 M. With these modifications, a 6-cubic meter reactor filled to 80% capacity afforded 799 kg of desired product. For this optimized reaction:

VTO Optimized Route BI 201302.png

Thus, after optimization, this synthetic step became less costly in terms of equipment and time and more practical to perform in a standard manufacturing facility, eliminating the need for a costly investment in a new dedicated plant.

 

Simvastatin, originally developed by Merck, is the most frequently prescribed statin today, with more nearly 100 million prescriptions filled in 2010, according to IMS Health. The traditional synthesis of the drug entailed a multi-step chemical process starting from Lovastatin. The chemical process was using large amounts of hazardous reagents as well as large quantities of solvents.

Professor Yi Tang, at UCLA conceived an initial synthesis that used an engineered enzyme. Codexis Inc. licensed the intellectual property from UCLA, optimized the initial enzyme and developed the new process for commercial use as shown in Figure 1. Following the quantitative hydrolysis of lovastatin to monacolin J acid, Codexis developed a novel, non-natural acyl donor enzyme to regioselectively acylate the C8 position and effect cyclization to simvastatin. This mild bioenzymatic process reduces the 4 steps chemical synthesis to only two steps. The Codexis process is significantly more efficient, cost effective and environmentally friendly.

 

This is the reaction scheme for producing the drug Simvastatin. The process was an award-winner at last month’s Green Chemistry Challenge Awards held by the Environmental Protection Agency

“We started working on Simvastatin in 2008 and completed the planning process in 2010,” Huisman said. “Then, we started the commercialization process, which takes time because you need regulatory approval of the new process we were working on. We licensed some technology from Yi Tang and UCLA and were then able to continue.”

Codexis took the three-step process used to make Simvastatin and cut out two of the steps, Huisman said.

“From the starting material, it (Simvastatin) has three reactive groups, or hydroxy groups, and what we need to do is convert two of the three groups,” Huisman explained. “We took out a protective step and a de-protective step. We took out two of the steps, and it was intense chemical processing. We then were able to accomplish everything in one step. We also circumvented the use of several nasty chemicals, as well.”

By cutting out two steps, “the overall yield goes up tremendously, about 35 percent,” Huisman added. “And we’re generating 25 times less waste than we did in the old process.”

Huisman said the new process doesn’t change the drug’s effects at all, and that scientists have been trying to do this type of work on commercial drugs for decades.

“In order for this to be a commercial process, the enzyme needs to be improved,” he said. “We needed to speed up the enzyme 1,000-fold to make this process workable; it took a team of scientists about nine months to optimize the enzymes and speed it up.”

 Codexis 2 step enzymatic process versus the 4 step chemical synthesis

AZIDES

A popular procedure for making 5-substituted tetrazoles is the reaction of sodium azide with a nitrile, often in the presence of an ammonium salt. The example shown below is from Organic Syntheses (Novartis Process R&D and Ley’s group at Cambridge), providing the useful enantiocatalyst shown on an 80 mmol scale. The excess sodium azide was destroyed with sodium nitrite and sulfuric acid, which converts hydrazoic acid into nitrogen and nitrous oxide gases.

Novartis/Cambridge example of the reaction of a  nitrile with sodium azide to produce a tetrazole

While the above procedure may be popular, any time you use sodium azide you should be thinking, “hydrazoic acid can be generated, it’s explosive and toxic, and I need to take the appropriate safety precautions.” That’s precisely what happened during some recent process R&D work at Merck Frosst on the steroyl-CoA desaturase inhibitor MK-8245. The discovery chemistry route used NaN3/pyridinium chloride as shown below, but the process group felt that the potential for significant amounts of hydrazoic acid generation was too high.

Armed with the ability to detect hydrazoic acid in the headspace above the reaction mixture using online IR, the Merck Frosst researchers surveyed alternatives. Sharpless’s zinc bromide procedure, proposed to minimize hydrazoic acid formation by control of the pH, led to a reading of 2000 ppm of HN3 in the headspace, which is below the detonation threshold of 15,000 ppm but was still felt to be undesirable.  In their own survey of conditions, the Merck Frosst scientists found something quite new and significant: Reaction with sodium azide in the presence of a catalytic amount of zinc oxide in aqueous THF (pH 8) proceeded efficiently, and most notably, with only 2 ppm of HN3 in the headspace! They were able to make 7 kg of the tetrazole in one run in nearly quantitative yield. Nice!

Sharpless and Merck Frosst Modifications

 

I’d be remiss if I didn’t mention Bu3SnN3 and Me3SiN3/Cu(I) as sodium azide surrogates, sometimes used on large scale. Shown below is an application to valsartan (see here and here) with recycling of the tin by-products. The intermediate stannyl tetrazole and leftover Bu3SnN3 were converted with HCl to Bu3SnCl, which was then converted to the fluoride, which was removed by filtration and recycled to Bu3SnCl.

Valsartan synthesis

Additional Topics

Transition-Metal Catalysis and Organocatalysis

Biocatalysis and Enzymatic Engineering

Recently, large pharmaceutical process chemists have relied heavily on the development of enzymatic reactions to produce important chiral building blocks for API synthesis. Many varied classes of naturally occurring enzymes have been co-opted and engineered for process pharmaceutical chemistry applications. The widest range of applications come from ketoreductases and transaminases, but there are isolated examples from hydrolases, aldolases, oxidative enzymes, esterases and dehalogenases, among others.[10]

One of the most prominent uses of biocatalysis in process chemistry today is in the synthesis of Januvia®, a DPP-4 inhibitor developed by Merck for the management of type II diabetes. The traditional process synthetic route involved a late-stage enamine formation followed by rhodium-catalyzed asymmetric hydrogenation to afford the API sitagliptin. This process suffered from a number of limitations, including the need to run the reaction under a high-pressure hydrogen environment, the high cost of a transition-metal catalyst, the difficult process of carbon treatment to remove trace amounts of catalyst and insufficient stereoselectivity, requiring a subsequent recrystallization step before final salt formation.[11][12]

Comparison of the chemosynthetic and biosynthetic routes toward sitagliptin.

Merck’s process chemistry department contracted Codexis, a medium-sized biocatalysis firm, to develop a large-scale biocatalytic reductive amination for the final step of its sitagliptin synthesis. Codexis engineered a transaminase enzyme from the bacteria Arthrobacter through 11 rounds of directed evolution. The engineered transaminase contained 27 individual point mutations and displayed activity four orders of magnitude greater than the parent enzyme. Additionally, the enzyme was engineered to handle high substrate concentrations (100 g/L) and to tolerate the organic solvents, reagents and byproducts of the transamination reaction. This biocatalytic route successfully avoided the limitations of the chemocatalyzed hydrogenation route: the requirements to run the reaction under high pressure, to remove excess catalyst by carbon treatment and to recrystallize the product due to insufficient enantioselectivity were obviated by the use of a biocatalyst. Merck and Codexis were awarded the Presidential Green Chemistry Challenge Award in 2010 for the development of this biocatalytic route toward Januvia®.[13]

 

 

ATORVASTATIN

Biocatalytic process development firm Codexis was recognized with the award in the greener reaction conditions category for developing a “green-by-design” enzymatic process to replace a chemical process for making ethyl (R)-4-cyano-3-hydroxybutyrate. This chemical, also known as hydroxynitrile, is the key chiral building block used to make atorvastatin, the active ingredient in Pfizer‘s cholesterol-lowering drug Lipitor.

The new process is helping to lower atorvastatin’s long-term production costs, according to John H. Grate, senior vice president of R&D and chief technology officer at Codexis. The savings could be financially significant for Pfizer and future generics manufacturers given that Lipitor is the world’s top pharmaceutical, with annual sales of about $13 billion.

1

Hydroxynitrile is used in the early stages of atorvastatin synthesis to build the chiral dihydroxy acid side chain that’s essential to the drug’s activity, Grate told C&EN. Demand for the intermediate is about 200 metric tons per year, and it’s currently being made by several fine chemicals producers. The competition to supply the intermediate to Pfizer has spurred several firms to chase after a better way to prepare hydroxynitrile (Angew. Chem. Int. Ed. 2005, 44, 362).

 

 

Chemical engineering professor Galen J. Suppes of the University of Missouri, Columbia, was honored with the academic award for his group’s work to create a low-cost catalytic process to convert the glycerol by-product from biodiesel production into propylene glycol–turning 1,2,3-propanetriol into 1,2-propanediol. At first glance, this achievement may not sound that exciting. But the repercussions of Suppes’s accomplishment are expected to have a major impact on the future use of biodiesel fuel, the world glycerol market, and the environmental health and safety of antifreeze and deicing chemicals.

4Photo by Rob Hill/MU Publications
GREEN SOLUTION Suppes and his group uncovered ideal reaction conditions for the catalytic conversion of by-product glycerol to useful propylene glycol.

 

Biodiesel is a mixture of fatty acid methyl esters made by esterifying soybean oil or other vegetable oil or animal fat. The triglycerides in the oil consist of three long fatty acid chains connected to a propyl headgroup. Sodium hydroxide is used to cleave the chains, which in turn are reacted with methanol to form methyl esters, leaving the residual glycerol headgroup as a by-product. About 1 kg of crude glycerol is formed for every 9 kg of biodiesel produced.

Millions of gallons of glycerol are flooding the world market as biodiesel production is ramping up in the U.S. and Europe, Suppes explained. The fallout from this glycerol glut is that chemical companies have shuttered some glycerol production plants and are considering glycerol as a starting material to make a host of feedstock chemicals (C&EN, Feb. 6, page 7).

Suppes entered the picture about four years ago when he realized that an inexpensive method to convert glycerol to propylene glycol could be valuable, he said. Utilizing the glycerol not only would help offset the cost of biodiesel production, but the inexpensive propylene glycol could be used as a low-toxicity replacement for ethylene glycol in automotive antifreeze.

Suppes’s system involves low-pressure hydrogenolysis of glycerol using a copper chromite catalyst, CuO•Cr2O3 (Appl. Catal. A 2005, 281, 225). In the two-step process, glycerol is first dehydrated to form acetol (1-hydroxy-2-propanone), which is then hydrogenated to form propylene glycol.

6
GREEN LEFTOVERS Glycerol by-product from biodiesel production can be used as a feedstock in Suppes’ process to produce acetol or propylene glycol from renewable resources.

Copper chromite hydrogenolysis catalysts aren’t new, but the success of the Missouri process is in achieving high selectivity for propylene glycol by controlling the temperature and hydrogen pressure of the reaction, Suppes noted. In the past, researchers tended to use reaction temperatures that were too high, leading to a higher percentage of by-products. Thus, they “missed the window of opportunity to achieve high selectivity,” Suppes said. Tinkering with temperature, pressure, and several different catalysts, Suppes and his colleagues optimized the system to operate at about 220 °C and less than 10 bar versus about 260 °C and more than 150 bar for other systems.

Another key part of the synthesis is the ability to isolate the acetol intermediate, Suppes added. Acetol is a synthetic starting material used to make polyols. But when made from petroleum, it costs about $5.00 per lb, discouraging its widespread use. Suppes envisions that producing acetol from biomass-based glycerol using his process could lower the cost to 50 cents per lb, “opening up even more potential applications and markets for products made from glycerol.”

Suppes’s propylene glycol process has been patented and is being licensed through the Missouri Soybean Merchandising Council, which provided partial funding for the research. The first commercial facility, with an annual capacity of 11.5 million gal, is being built in an undisclosed location in the U.S. by Senergy Chemical. It’s expected to be in operation by the end of this year.

 

 

E7398, INN eribulin mesylate

The most awe-inspiring example of a positive tangible outcome from the combination of basic research into the synthesis of a system, and a correctly weighted assessment of ‘scalability’, is Halaven® (2, E7398, INN eribulin mesylate). Most chemists in industry and academia alike would have considered using total synthesis to support clinical development and commercialization of this compound a ‘fool’s errand,’ but the Kishi group and Eisai Inc. did not. The fact is that this compound solves a major clinical problem, so taking on the issues (length of synthesis, stability limitations, stereochemical problems, etc.) had a big payoff (reducing the relative weighting or importance of these factors in assessing the viability of a commercial chemical synthesis). As depicted below, a highly convergent approach, combined with powerful methodology for stitching together key fragments 5 and 6 (Nozaki–Hiyama–Kishi (NHK) coupling) and a strategy of targeting crystalline intermediates were all key elements that culminated in this landmark accomplishment

 

The commercial synthesis of Halaven® (2), a landmark achievement in process chemistry

 

Artemisinin (Cook, 2012). 

(+)-Artemisinin (41) is currently the most effective drug against Plasmodium falciparum malaria as part of an artemisinin-based combination therapy (ACT). Although it can be isolated on an industrial scale from Artemisia annua, the market price of artemisinin (41) has fluctuated widely and traditional extraction does not provide enough material to meet the worldwide demand. Interestingly, recent efforts towards a cheaper and more efficient production of artemisinin (41) have mainly taken place in the areas of synthetic biology, semisynthesis and plant engineering, while there has been a lack of practical approaches using a straightforward total synthesis. Despite the fact that all the total syntheses of artemisinin, until 2010, were impressive from a feasibility point of view, none of them provided a solution for the low-cost synthesis of 41. This changed when Cook’s group recently published a scalable synthesis of artemisinin (41), which provides a blueprint for the cost-effective production of 41 and its derivatives below Key to their successful strategy was the use of reaction cascades that rapidly built complexity, starting from the cheap feedstock chemical, cyclohexenone (42). The latter was first subjected to a one-pot conjugate addition/alkylation sequence, to give ketone 43. A three-step sequence consisting of formylation, cycloaddition and a Wacker-type oxidation, yielded 9.4 g of methyl ketone 44. The challenging formation of the unusual peroxide bridge was initially met with failure, but was eventually realized by a reaction with singlet oxygen to give 41 amongst other oxidized intermediates. The entire synthetic sequence was conducted on a gram scale, required only three chromatographic purifications and was carried out in only five flasks. Considering the low cost of the commodity chemicals used and the conciseness of Cook’s synthesis, it is certainly worth being further investigated. 

image file: c3np70090a-s10.tif
 Cook’s scalable route to (+)-artemisinin (41).

Continuous/Flow Manufacturing

In recent years, much progress has been made in the development and optimization of flow reactors for small-scale chemical synthesis (the Jamison Group at MIT and Ley Group at Cambridge University, among others, have pioneered efforts in this field). The pharmaceutical industry, however, has been slow to adopt this technology for large-scale synthetic operations. For certain reactions, however, continuous processing may possess distinct advantages over batch processing in terms of safety, quality and throughput.

A case study of particular interest involves the development of a fully continuous process by the process chemistry group at Eli Lilly and Company for an asymmetric hydrogenation to access a key intermediate in the synthesis of LY500307,[14] a potent ERβ agonist that is entering clinical trials for the treatment of patients with schizophrenia, in addition to a regimen of standard antipsychotic medications. In this key synthetic step, a chiral rhodium-catalyst is used for the enantioselective reduction of a tetrasubstituted olefin. After extensive optimization, it was found that in order to reduce the catalyst loading to a commercially practical level, the reaction required hydrogen pressure up to 70 atm. The pressure limit of a standard chemical reactor is about 10 atm, although high-pressure batch reactors may be acquired at significant capital cost for reactions up to 100 atm. Especially for an API in the early stages of chemical development, such an investment clearly bears a large risk.

An additional concern was that the hydrogenation product has an unfavorable eutectic point, so it was impossible to isolate the crude intermediate in more than 94 percent ee by batch process. Because of this limitation, the process chemistry route toward LY500307 necessarily involved a kinetically controlled crystallization step after the hydrogenation to upgrade the enantiopurity of this penultimate intermediate to >99 percent ee.

Comparison of the batch and continuous flow processes toward LY500307.

The process chemistry team at Eli Lilly successfully developed a fully continuous process to this penultimate intermediate, including reaction, workup and kinetically controlled crystallization modules (the engineering considerations implicit in these efforts are beyond the scope of this article). An advantage of flow reactors is that high-pressure tubing can be utilized for hydrogenation and other hyperbaric reactions. Because the head space of a batch reactor is eliminated, however, many of the safety concerns associated with running high-pressure reactions are obviated by the use of a continuous process reactor. Additionally, a two-stage mixed suspension-mixed product removal (MSMPR) module was designed for the scalable, continuous, kinetically controlled crystallization of the product, so it was possible to isolate in >99 percent ee, eliminating the need for an additional batch crystallization step.

This continuous process afforded 144 kg of the key intermediate in 86 percent yield, comparable with a 90 percent isolated yield using the batch process. This 73-liter pilot-scale flow reactor (occupying less than 0.5 m3 space) achieved the same weekly throughput as theoretical batch processing in a 400-liter reactor. Therefore, the continuous flow process demonstrates advantages in safety, efficiency (eliminates the need for batch crystallization) and throughput, compared with a theoretical batch process.

 

US scientists have found a way to stop solid byproducts clogging channels in continuous flow reactors, a problem that has hampered their progress for use in manufacturing pharmaceuticals.

Klavs Jensen, Stephen Buchwald and their team at the Massachusetts Institute of Technology believe that flow methods will become increasingly important in the future of pharmaceuticals and chemical manufacturing. ‘One of the biggest hurdles is handling solids,’ says group member Timothy Noël. ‘Precipitates can form during the reactions, which usually lead to irreversible clogging of microchannels in the reactors.’ Previous methods suggested to overcome this problem include introducing another solvent to dissolve the solids, but this can reduce the overall efficiency of the reactions. Now, the team have used an ultrasound bath to break up the byproducts to prevent clogging.

Traditionally, pharmaceutical manufacture is done in a batch-based system, but the process suffers from interruptions and the need to transport material between batch reactors. Performing these reactions in a continuous flow system would speed up the process and reduce chemical waste.

Unclogging the problems of flow chemistry

Reagents were introduced into a tube, which was then placed in an ultrasonic bath heated to 60 degrees Celsius. When the reagents exited the reactor, the reaction was mixed with a quench of water and ethyl acetate in a larger tube, allowing plenty of time for salt byproducts to dissolve

 

The team tested the method on palladium-catalysed C-N cross-coupling reactions, making amines that are common in biologically active molecules. The reactions couple aryl halides to nitrogen nucleophiles and form byproducts – inorganic salts – that are insoluble in the solvents used.

As a result, says Noël, they were able to obtain diarylamine products with reaction times ranging from 20 seconds to 10 minutes. At very short residence times (time in the reactor under reaction conditions) they observed a significantly higher rate for the reaction in flow compared to the equivalent batch experiments. With high conversions in short reaction times, they were able to reduce the catalyst loading in flow to just 0.1 mol per cent. ‘Extremely low catalyst loadings such as these are of particular interest to the pharmaceutical industry,’ says Noël.

Noël believes that in the future microfluidics will be used to construct increasingly complex molecules. Different devices will automate and integrate many synthetic steps that are currently performed using the more traditional and time-consuming batch-based practices.

Oliver Kappe, from the Christian Doppler Laboratory for Microwave Chemistry, Institute of Chemistry, Karl-Franzens-University Graz says: ‘Jensen and Buchwald clearly demonstrate that immersing a flow device into an ultrasound bath can prevent clogging problems that unfortunately are all too familiar to the flow/microreactor community.’

 

Direct Fluorination and Microreactor Technology

Elemental fluorine has long been considered to be too reactive and uncontrollable for use as a reagent in organic synthesis and this perception still predominates. Prof. Poliakoff’s comments on the popular Periodic Table video series (www.PeriodicVideos.com), ‘It was much more exciting than I thought …you see the flames,’ and general comments in standard advanced organic chemistry textbooks (J. March, Advanced Organic Chemistry, ‘Direct fluorination of aromatic rings with F2 is not feasible at room temperature because of the extreme reactivity of F2….not yet of preparative significance) are typical.

Despite this background, research into the use of elemental fluorine for organic synthesis at Durham has overcome the many problems of using fluorine gas for the safe synthesis of fine chemicals, in particular, by use of dilute fluorine gas in nitrogen, appropriate solvent choice (high dielectric constant media such as formic acid, sulfuric acid or acetonitrile), reactor vessel design, gas flow regulator systems and stainless steel/monel fluorine gas handling lines have developed over the years to allow selective direct fluorination of a range of aliphatic, dicarbonyl, aromatic, heteroaromatic, heterocyclic, steroid and carbohydrate derivatives to be established and the mechanism (regiochemistry, stereochemistry, selectivity, etc.) of these processes to be assessed. Indeed, direct fluorination of aromatic rings is feasible at room temperature !  Research expanding the use of fluorine gas continues to develop new selective fluorination methodology for the synthesis of a range of aromatic, heterocylic and aliphatic systems.2,3

In particular, a process for the synthesis of a fluoroketoester first carried out in Durham was developed by our industrial collaborators, F2 Chemicals Ltd (UK), for the Pfizer company and forms a key starting material in the multi step synthesis of the widely used anti-fungal agent V-Fend (Voriconazole) throughout the clinical trial, launch and commercialization periods. In the period from January 2008 to March 2011 approximately 17 tonnes of the fluoroketoester were manufactured for Pfizer by F2 Chemicals Ltd. Global sales of V-Fend in the 2008-2010 period total $2.4 billion (Pfizer annual financial reports) and in 2010 was 17th position in Pfizer’s best selling products and it is one of the global top 100 best selling pharmaceutical products.

Further reaction control in selective fluorination reactions was achieved by the design, fabrication and commissioning of single and multi-channel continuous flow reactor systems, establishing the use of convenient, inexpensive flow reactors for gas – liquid processes using flow regimes in the laboratory. Techniques for the supply of individual gas and liquid reagents from single sources to a parallel array of many flow channels at the same flow rate and pressure whilst maintaining laminar flow within the reactor channels and telescoped gas – liquid / liquid – liquid processes involving fluorination and ring formation in one continuous flow process have been developed.

References

  1. Roughley, S. D.; Jordan, A. M. (2011). “The medicinal chemist’s toolbox: an analysis of reactions used in the pursuit of drug candidates”. J. Med. Chem. 54: 3451.
  2. Dach, R.; Song, J. J.; Roschangar, F.; Samstag, W.; Senanayake, C. H. (2012). “The eight criteria defining a good chemical manufacturing process”. Org. Process Res. Dev. 16: 1697.
  3. Trost, B. M. (1991). “The atom economy – a search for synthetic efficiency”. Science 254: 1471.
  4. Van Aken, K.; Strekowski, L.; Patiny, L. (2006). “EcoScale, a semi-quantitative tool to select an organic preparation based on economical and ecological parameters”. Beilstein J. Org. Chem. 2 (No. 3).
  5. Faucher, A-M.; Bailey, M. D.; Beaulieu, P. L.; Brochu, C.; Duceppe, J-S.; Ferland, J-M.; Ghiro, E.; Gorys, V.; Halmos, T.; Kawai, S. H.; Poirier, M.; Simoneau, B.; Tsantrizos, Y. S.; Llinas-Brunet, M. (2004). “Synthesis of BILN 2061, an HCV NS3 protease inhibitor with proven antiviral effect in humans”. Org. Lett. 6: 2901.
  6. Yee, N. K.; Farina, V.; Houpis, I. N.; Haddad, N.; Frutos, R. P.; Gallou, F.; Wang, X-J.; Wei, X.; Simpson, R. D.; Feng, X.; Fuchs, V.; Xu, Y.; Tan, J.; Zhang, L.; Xu, J.; Smith-Keenan, L. L.; Vitous, J.; Ridges, M. D.; Spinelli, E. M.; Johnson, M. (2006). “Efficient large-scale synthesis of BILN 2061, a potent HCV protease inhibitor, by a convergent approach based on ring-closing metathesis”. J. Org. Chem. 71: 7133.
  7. Zeng, X.; Wei, X.; Farina, V.; Napolitano, E.; Xu, Y.; Zhang, L.; Haddad, N.; Yee, N. K.; Grinberg, N.; Shen, S.; Senanayake, C. H. (2006). “Epimerization reaction of a substituted vinylcyclopropane catalyzed by ruthenium carbenes: mechanistic analysis”. J. Org. Chem. 71: 8864.
  8. Grela, K.; Harutyunyan, S.; Michrowska, A. (2002). “A highly efficient ruthenium catalyst for metathesis reactions”. Angew. Chem. Int. Ed. 41: 4038.
  9. Wei, X.; Shu, C.; Haddad, N.; Zeng, X.; Patel, N. D.; Tan, Z.; Liu, J.; Lee, H.; Shen, S.; Campbell, S.; Varsolona, R. J.; Busacca, C. A.; Hossain, A.; Yee, N. K.; Senanayake, C. H. (2013). “A highly convergent and efficient synthesis of a macrocyclic hepatitis C virus protease inhibitor BI 201302″. Org. Lett. 15: 1016.
  10. Bornscheuer, U. T.; Huisman, G. W.; Kazlauskas, R. J.; Lutz, S.; Moore, J. C.; Robins, K. (2012). “Engineering the third wave of biocatalysis”. Nature 485: 185.
  11. Savile, C. K.; Janey, J. M.; Mundorff, E. C.; Moore, J. C.; Tam, S.; Jarvis, W. R.; Colback, J. C.; Krebber, A.; Fleitz, F. J.; Brands, J.; Devine, P. N.; Huisman, G. W.; Hughes, G. J. (2010). “Biocatalytic asymmetric synthesis of chiral amines applied to sitagliptin manufacture”. Science 329: 305.
  12. Desai, A. A. (2011). “Sitagliptin manufacture: a compelling tale of green chemistry, process intensification, and industrial asymmetric catalysis”. Angew. Chem. Int. Ed. 50: 1974.
  13. Busacca, C. A.; Fandrick, D. R.; Song, J. J.; Sananayake, C. H. (2011). “The growing impact of catalysis in the pharmaceutical industry”. Adv. Synth. Catal. 353: 1825.
  14. Johnson, M. D.; May, S. A.; Calvin, J. R.; Remacle, J.; Stout, J. R.; Dieroad, W. D.; Zaborenko, N.; Haeberle, B. D.; Sun, W-M.; Miller, M. T.; Brannan, J. (2012). “Development and scale-up of a continuous, high-pressure, asymmetric hydrogenation reaction, workup, and isolation”. Org. Process Res. Rev. 16: 1017.

Filed under: PROCESS, Uncategorized Tagged: PROCESS

Apigenin: this chemical breaks the immortality of cancer cells

$
0
0
parsley apigenin
Apigenin, which abounds in particular parsley, have protective effects against cancer. Indeed, a U.S. study showed that apigenin alters the process of gene regulation in cancer cells, which has the effect of making them sensitive to the new process of cell death. Credits: H. Zell
Apigenin, a very natural chemical compound present in the Mediterranean diet, breaks immortality of cancer cells. A result obtained by researchers at the Ohio State University (USA).
Apigeninapigenin
Apigenin is found in many fruits and vegetables, but parsleycelery and chamomile tea are the most common sources

Filed under: cancer Tagged: CANCER, epigenin

Dextrose and Morrhuate Sodium Injections for Knee Osteoarthritis

$
0
0

Originally posted on lyranara.me:

A new nonsurgical approach to treating chronic pain and stiffness associated with knee osteoarthritis has demonstrated significant, lasting improvement in knee pain, function, and stiffness. This safe, two-solution treatment delivered in a series of injections into and around the knee joint is called prolotherapy.

David Rabago, MD, and a team of researchers from the University of Wisconsin School of Medicine and Public Health, and Meriter Health Services, Madison, WI, report substantial improvement among participants in the one-year study who received at least three of the two-solution injections. Symptom improvement ranged from 19.5-42.9% compared to baseline status.

As described in the article “Dextrose and Morrhuate Sodium Injections (Prolotherapy) for Knee Osteoarthritis: A Prospective Open-Label Trial“, reported improvement in knee pain, function, and stiffness scores exceeded the minimum for a “clinically important difference” in 50-75% of patients.

Here is the full text of this article: http://online.liebertpub.com/doi/full/10.1089/acm.2013.0225

View original


Filed under: Uncategorized

Milk Thistle Promising for Colorectal Cancer Prevention

$
0
0

Originally posted on lyranara.me:

Colorectal cancer stem cells thrive in conditions of inflammation. A University of Colorado Cancer Center study presented today at the American Association for Cancer Research (AACR) Annual Meeting 2014 shows that the chemical silibinin, purified from milk thistle extract, affects cell signaling associated with inflammation and thus also the formation and survival of colorectal cancer stem cells.

“We have been deeply involved in this line of research that extends from silibinin to its chemopreventive properties in colorectal cancer, and the current study takes another important step: we see both a likely chemopreventive mechanism and the result of this mechanism in animal models,” says Sushil Kumar, PhD, postdoctoral fellow in the lab of Rajesh Agarwal, PhD, co-program leader of Cancer Prevention and Control at the CU Cancer Center and professor at the Skaggs School of Pharmacy and Pharmaceutical Sciences.

The group compared mice chemically treated to develop inflammation-dependent colorectal cancer…

View original 301 more words


Filed under: Uncategorized

سیاه‌دانه Nigella Sativa حبة البركة Kills 89% of Lung Cancer Cells in Vitro

$
0
0
Nigella Sativa Kills 89% of Lung Cancer Cells in Vitro: Researchers have just shown that nigella sativa (also known as black seed or black cumin) seed oil kills up to 89% of human lung cancer cells (A-549) after just 24 hours, while a non-oil extract from the seeds killed up to 77% of the cancer cells. The extracts were prepared from seeds obtained at a local market. Nigella sativa is a powerful medicinal herb which has been used for thousands of years in traditional Chinese, Ayurvedic, Unani and Arabic medicine. It is best known for its potent anti-inflammatory and antioxidant properties, and has been used to suppress coughs, treat kidney stones, diarrhea and stomach pain. But modern science has now also uncovered nigella’s powerful anti-diabetes and anti-cancer effects. This super herb has already shown potent activity against cancer of the breast, prostate, kidney, pancreas, liver, colon and cervix in previous lab studies, and this new study has shown new activity against lung cancer. Good health and cancer prevention should always start with a well-balanced diet focused on organic vegetables, fruit and whole foods (consuming at least half in the raw state). But nigella sativa may offer sizeable benefits for those wanting an extra measure of protection.<br /><br /><br /><br />
#NigellaSativa #BlackCumin #BlackSeed<br /><br /><br /><br />
<a href=http://www.ncbi.nlm.nih.gov/pubmed/24568529&#8243; width=”395″ height=”395″ />

Nigella Sativa Kills 89% of Lung Cancer Cells in Vitro: Researchers have just shown that nigella sativa (also known as black seed or black cumin) seed oil killsup to 89% of human lung cancer cells (A-549) after just 24 hours, while a non-oil extract from the seeds killed up to 77% of the cancer cells.

The extracts were prepared from seeds obtained at a local market. Nigella sativa is a powerful medicinal herb which has been used for thousands of years in traditional Chinese, Ayurvedic, Unani and Arabic medicine. It is best known for its potent anti-inflammatory and antioxidant properties, and has been used to suppress coughs, treat kidney stones, diarrhea and stomach pain. But modern science has now also uncovered nigella’s powerful anti-diabetes and anti-cancer effects.

This super herb has already shown potent activity against cancer of the breast, prostate, kidney, pancreas, liver, colon and cervix in previous lab studies, and this new study has shown new activity against lung cancer. Good health and cancer prevention should always start with a well-balanced diet focused on organic vegetables, fruit and whole foods (consuming at least half in the raw state). But nigella sativa may offer sizeable benefits for those wanting an extra measure of protection.


read at

http://www.ncbi.nlm.nih.gov/pubmed/24568529

 

Nigella sativa is an annual flowering plant, native to south and southwest Asia. It grows to 20–30 cm (7.9–11.8 in) tall, with finely divided, linear (but not thread-like) leaves. The flowers are delicate, and usually coloured pale blue and white, with five to ten petals. The fruit is a large and inflated capsule composed of three to seven united follicles, each containing numerous seeds. The seed is used as a spice.

Etymology

Nigella sativa seed

The scientific name is a derivative of Latin niger (black).[2]

Common names

In EnglishNigella sativa seed is variously called fennel flower,[3] nutmeg flower,[3] black caraway,[3] Roman coriander,[3] and also called black cumin.[3] Other names used, sometimes misleadingly, are onion seed and black sesame, both of which are similar-looking, but unrelated.Blackseed and black caraway may also refer to Bunium persicum.[4]

The seeds are frequently referred to as black cumin (as in Assamesekaljeera or kolajeera or Bengali kalo jeeray), But black cumin (kala Jeera)[clarification needed] is different than Nigella sativa (Kali Jeeri).[citation needed] In south Indian language Kannada it is called [ಕೃಷ್ಣ ಜೀರಿಗೆ] “Krishna Jeerige”, but this is also used for a different spice, Bunium persicum.

In English-speaking countries with large immigrant populations, it is also variously known as kaljeera (Assamese কালজীৰা kalzira or ক’লাজীৰাkolazira), kalo jira (Bengaliকালোজিরা kalojira, black cumin), karum cheerakamhabbat al-barakah (Arabic حبة البركة) Kurdish “reşke” (rashkeh) (Tamil கருஞ்சீரகம்), kalonji (Hindi कलौंजी kalauṃjī or कलोंजी kaloṃjīUrdu كلونجى kaloṃjī) or mangrail (Hindi मंगरैल maṃgarail), “Kala Jira in Marathi” ketzakh (Hebrew קצח), chernushka (Russian), çörek otu (Turkish), garacocco (Cypriot Turkish), ḥebbit al-barakah, seed of blessing (Arabic), siyah daneh (Persian سیاه‌دانه siyâh dâne), jintan hitam (Indonesian), karim jeerakam (കരിംജീരകം) in Malayalamor කළු දුරු in SinhalaKarto Jeera in Beary.

It is used as part of the spice mixture paanch phoran or panch phoron (meaning a mixture of five spices) and by itself in a great many recipes in Bengali cookery and most recognizably in naan bread.[5]

The Turkish name çörek otu literally means “bun’s herb” from its use in flavouring the çörek buns. Such braided-dough buns are widespread in the cuisines of Turkey and its neighbours (see Tsoureki τσουρέκι). In Bosnian, the Turkish name for Nigella sativa is respelled as čurekot. The seed is used in Bosnia, and particularly its capital Sarajevo, to flavour pastries (Bosnian: somun) often baked on Muslim religious holidays.

The Arabic approbation about Bunium bulbocastanum (Kaala Jeera) Hebbit il barakah, meaning the “seed of blessing” is also applied toNigella sativa (Kali Jeeri).

Characteristics

Nigella sativa has a pungent bitter taste and smell. It is used primarily in confectionery and liquors. Peshawari naan is, as a rule, topped with kalonji seeds. Nigella is also used in Armenian string cheese, a braided string cheese called Majdouleh or Majdouli in the Middle East.

History

According to Zohary and Hopf, archaeological evidence about the earliest cultivation of N. sativa “is still scanty”, but they report supposed N. sativa seeds have been found in several sites from ancient Egypt, including Tutankhamun‘s tomb.[6] Although its exact role in Egyptian culture is unknown, it is known that items entombed with a pharaoh were carefully selected to assist him in the afterlife.

The earliest written reference to N. sativa is thought to be in the book of Isaiah in the Old Testament, where the reaping of nigella and wheat is contrasted (Isaiah 28: 25, 27). Easton’s Bible dictionary states the Hebrew word ketsah refers to N. sativa without doubt (although not all translations are in agreement). According to Zohary and Hopf, N. sativawas another traditional condiment of the Old World during classical times; and its black seeds were extensively used to flavour food.[6]

Found in Hittite flask in Turkey from 2nd millennium BCE.[7]

History of medicineIn the Unani Tibb system of medicine, black cumin (Bunium bulbocastanum) is regarded as a valuable remedy for a number of diseases. Sayings of the Islamic prophet Muhammadunderline the significance of black cumin. According to a hadith narrated by Abu Hurairah, he says, “I heard Allah’s Apostle saying, ‘There is healing in black seed (haba sowda) for all diseases except death.’” [8] [9]

The black cumin (Bunium bulbocastanum) seeds have been traditionally used in the Middle East and Southeast Asian countries for a variety of ailments. Nigella seeds are sold as black cumin in small bundles to be rubbed until warm, when they emit an aroma similar to black cumin which opens clogged sinuses in the way that do eucalyptus or Vicks.

Nestlé has purportedly filed a patent application covering use of Nigella sativa as a food allergy treatment.[10] Yet the firm denies the claim of patenting the plant, stating that the patent would only cover “the specific way that thymoquinone - a compound that can be extracted from the seed of the fennel flower – interacts with opioid receptors in the body and helps to reduce allergic reactions to food”.[11]

Medical studies

Thymoquinone, found in the seed oil extract of N. sativa, has been shown to have anti-neoplastic effects in rats and mice and in cultured human cells from several types of cancer, including pancreatic ductal adenocarcinoma.[12] It has protective antioxidant and anti-inflammatory effects, and promotes apoptosis (cell death) of the cancer cells.[12]

Black cumin

Nigella sativa oil

Original black cumin (Bunium bulbocastanum) is rarely available, so N. sativa is widely used instead; in India, Carum carvi is the substitute. Cumins are from the Apiaceae (Umbelliferae) family, but N. sativa is from Ranunculaceae family. Black cumin (not N. sativa) seeds come as paired or separate carpels, and are 3–4 mm long. They have a striped pattern of nine ridges and oil canals, and are fragrant (Ayurveda says, “Kaala jaaji sugandhaa cha” (black cumin seed is fragrant itself)), blackish in colour, boat-shaped, and tapering at each extremity, with tiny stalks attached; it has been used for medicinal purposes for centuries, both as a herb and pressed into oil, in Asia, the Middle East, and Africa.

Chemistry

Nigella sativa oil contains an abundance of conjugated linoleic (18:2) acidthymoquinone, nigellone (dithymoquinone),[13] melanthinnigilline,damascenine, and tannins. Melanthin is toxic in large doses and nigelline is paralytic, so this spice must be used in moderation.

References

  1.  “The Plant List: A Working List of All Plant Species”.
  2.  New International Encyclopedia
  3.  “USDA GRIN Taxonomy”.
  4.  Bunium persicum - (Boiss.) B.Fedtsch. Common Name Black Caraway
  5.  Indian Naan with Nigella Seeds Recipe
  6.  Zohary, Daniel; Hopf, Maria (2000). Domestication of plants in the Old World (3 ed.). Oxford University Press. p. 206. ISBN 0-19-850356-3.
  7.  http://dx.doi.org/10.1016/j.jep.2009.05.039
  8.  Sunan Ibn Majah.
  9.  “71″Sahih Bukhari 7. 592.
  10.  Hammond, Edward (2012). “Food giant Nestlé claims to have invented stomach soothing use of habbat al-barakah (Nigella sativa)”Briefing Paper. Third World Network. Retrieved 23 April 2013.
  11.  “Is Nestlé trying to patent the fennel flower?”, http://www.nestle.com.
  12.  Chehl, N.; Chipitsyna, G.; Gong, Q.; Yeo, C.J.; Arafat, H.A. (2009). “Anti-inflammatory effects of the Nigella sativa seed extract, thymoquinone, in pancreatic cancer cells”. HPB (Oxford) 11 (5): 373–381. doi:10.1111/j.1477-2574.2009.00059.xPMID 19768141.
  13.  Mohammad Hossein Boskabady, Batool Shirmohammadi (2002). “Effect of Nigella Sativa on Isolated Guinea Pig Trachea”Arch Iranian Med 5 (2): 103–107.

Filed under: AYURVEDA, cancer, Herbals, Uncategorized Tagged: AYURVEDA, CANCER, nigella sativa

Doravirine, MK-1439 reverse transcriptase inhibitor In Phase 3 for treatment of HIV-1 infection

$
0
0

Doravirine.svg

 

Doravirine, MK-1439……….. AN ANTIVIRAL

3-Chloro-5-({1-[(4-methyl-5-oxo-4,5-dihydro-1H-1,2,4-triazol-3-yl)methyl]-2-oxo-4-(trifluoromethyl)-1,2-dihydro-3-pyridinyl}oxy)benzonitrile

Benzonitrile, 3-chloro-5-[[1-[(4,5-dihydro-4-methyl-5-oxo-1H-1,2,4-triazol-3-
yl)methyl]-1,2-dihydro-2-oxo-4-(trifluoromethyl)-3-pyridinyl]oxy]-

3-chloro-5-({1-[(4-methyl-5-oxo-4,5-dihydro-1H-1,2,4-triazol-3-yl)methyl]-2-
oxo-4-(trifluoromethyl)-1,2-dihydropyridin-3-yl}oxy)benzonitrile

1338225-97-0 CAS

MOLECULAR FORMULA C17H11ClF3N5O3
MOLECULAR WEIGHT 425.7

Merck Sharp & Dohme Corp

 reverse transcriptase inhibitor

Doravirine (MK-1439) is a non-nucleoside reverse transcriptase inhibitor under development by Merck & Co. for use in the treatment of HIV infection. Doravirine demonstrated robust antiviral activity and good tolerability in a small clinical study of 7-day monotherapy reported at the 20th Conference on Retroviruses and Opportunistic Infections in March 2013. Doravirine appeared safe and generally well tolerated with most adverse events being mild-to-moderate.[1][2]

investigational next-generation, non-nucleoside reverse transcriptase inhibitor (NNRTI), at the 21st Conference on Retroviruses and Opportunistic Infections (CROI). Interim data demonstrating potent antiretroviral (ARV) activity for four doses (25, 50, 100 and 200 mg) of once-daily, oral doravirine in combination with tenofovir/emtricitabine in treatment-naïve, HIV-1 infected adults after 24 weeks of treatment were presented during a late-breaker oral session. Based on these findings as well as other data from the doravirine clinical program, Merck plans to initiate a Phase 3 clinical trial program for doravirine in combination with ARV therapy in the second half of 2014.

“Building on our long-standing commitment to the HIV community, Merck continues to evaluate new candidates we believe have the potential to make a meaningful difference in the lives of HIV patients,” said Daria Hazuda, Ph.D., vice president, Infectious Diseases, Merck Research Laboratories. “We look forward to advancing doravirine into Phase 3 clinical trials in the second half of 2014.”

Doravirine Clinical Data

This randomized, double-blind clinical trial examined the safety, tolerability and efficacy of once-daily doravirine (25, 50, 100 and 200 mg) in combination with once-daily tenofovir/emtricitabine versus efavirenz (600 mg), in treatment-naïve, HIV-1 infected patients. The primary efficacy analysis was percentage of patients achieving virologic response (< 40 copies/mL).

At 24 weeks, doravirine doses of 25, 50, 100, and 200 mg showed virologic response rates consistent with those observed for efavirenz at a dose of 600 mg. All treatment groups showed increased CD4 cell counts.

Proportion of Patients with Virologic
Response at 24 weeks (95% CI)

Mean CD4 Change
from Baseline (95% CI)

Treatment* Dose (mg) n/N

% <40
copies/mL

cells/μL

Doravirine 25 32/40 80.0 (64.6, 90.9) 158 (119, 197)
50 32/42 76.2 (60.5, 87.9) 116 (77, 155)
100 30/42 71.4 (55.4, 84.3) 134 (100, 167)
200 32/41 78.0 (62.4, 89.4) 141 (96, 186)
Efavirenz 600 27/42 64.3 (48.0, 78.4) 121 (73, 169)
Missing data approach: Non-completer = Failure Observed Failure

*In combination with tenofovir/emtricitabine

The incidence of drug-related adverse events was comparable among the doravirine-treated groups. The overall incidence of drug-related adverse events was lower in the doravirine-treated groups (n=166) than the efavirenz-treated group (n=42), 35 percent and 57 percent, respectively. The most common central nervous system (CNS) adverse events at week 8, the primary time point for evaluation of CNS adverse experiences, were dizziness [3.0% doravirine (overall) and 23.8% efavirenz], nightmare [1.2% doravirine (overall) and 9.5% efavirenz], abnormal dreams [9.0% doravirine (overall) and 7.1% efavirenz], and insomnia [5.4% doravirine (overall) and 7.1% efavirenz].

Based on the 24-week data from this dose-finding study, a single dose of 100 mg doravirine was chosen to be studied for the remainder of this study, up to 96 weeks.

About Doravirine

DORAVIRINE

Doravirine, also known as MK-1439, is an investigational next-generation, NNRTI being evaluated by Merck for the treatment of HIV-1 infection. In preclinical studies, doravirine demonstrated potent antiviral activity against HIV-1 with a characteristic profile of resistance mutations selected in vitro compared with currently available NNRTIs. In early clinical studies, doravirine demonstrated a pharmacokinetic profile supportive of once-daily dosing and did not show a significant food effect.

Merck’s Commitment to HIV

For more than 25 years, Merck has been at the forefront of the response to the HIV epidemic, and has helped to make a difference through our proud legacy of commitment to innovation, collaborating with the community, and expanding global access to medicines. Merck is dedicated to applying our scientific expertise, resources and global reach to deliver healthcare solutions that support people living with HIV worldwide.

About Merck

Today’s Merck is a global healthcare leader working to help the world be well. Merck is known as MSD outside the United States and Canada. Through our prescription medicines, vaccines, biologic therapies, and consumer care and animal health products, we work with customers and operate in more than 140 countries to deliver innovative health solutions. We also demonstrate our commitment to increasing access to healthcare through far-reaching policies, programs and partnerships. For more information, visit www.merck.com and connect with us on TwitterFacebook and YouTube.

Discovery of MK-1439, an orally bioavailable non-nucleoside reverse transcriptase inhibitor potent against a wide range of resistant mutant HIV viruses
Bioorg Med Chem Lett 2014, 24(3): 917

http://www.sciencedirect.com/science/article/pii/S0960894X13014546

The optimization of a novel series of non-nucleoside reverse transcriptase inhibitors (NNRTI) led to the identification of pyridone 36. In cell cultures, this new NNRTI shows a superior potency profile against a range of wild type and clinically relevant, resistant mutant HIV viruses. The overall favorable preclinical pharmacokinetic profile of 36 led to the prediction of a once daily low dose regimen in human. NNRTI 36, now known as MK-1439, is currently in clinical development for the treatment of HIV infection.

Full-size image (16 K)

Full-size image (10 K)

Scheme 1. 

Reagents and conditions: (a) K2CO3, NMP, 120 °C; (b) KOH, tert-BuOH, 75 °C; (c) Zn(CN)2, Pd(PPh3)4, DMF, 100 °C.

Full-size image (12 K)

Scheme 3.

Reagents and conditions: (a) K2CO3, DMF, −10 °C; (b) MeI or EtI, K2CO3, DMF.

 

36 IS DORAVIRINE

 

 

WO 2011120133

http://www.google.com/patents/WO2011120133A1?cl=en

Scheme I depicts a method for preparing compounds of Formula I in which hydroxypyridine 1-1 is alkylated with chlorotriazolinone 1-2 to provide 1-3 which can be selectively alkylated with an alkyl halide (e.g., methyl iodide, ethyl iodide, etc.) to afford the desired 1-4. Scheme I

Figure imgf000039_0001

Scheme II depicts an alternative route to compounds of the present invention, wherein fluorohydroxypyridine II-l can be alkylated with chlorotriazolinone II-2 to provide the alkylated product II-3 which can be converted to the desired II-5 via nucleophilic aromatic substitution (S] fAr) using a suitable hydroxyarene II-4.

Scheme II

Figure imgf000039_0002

Hydroxypyridines of formula I-l (Scheme 1) can be prepared in accordance with Scheme III, wherein a SNAr reaction between pyridine III-l (such as commercially available 2- chloro-3-fluoro-4-(trifluoromethyl)pyridine) and hydroxyarene H-4 can provide chloropyridine III-2, which can be hydrolyzed under basic conditions to the hydroxypyridine I-l. Scheme III

Figure imgf000040_0001

Another method for preparing hydroxypyridines of formula I-l is exemplified in Scheme IV, wherein S Ar coupling of commercially available 2-chloro-3-fluoro-4- nitropyridone-N-oxide IV-1 with a suitable hydroxyarene II-4 provides N-oxide IV-2, which can first be converted to dihalides IV-3 and then hydro lyzed to hydroxypyridine IV-4. Further derivatization of hydroxypyridine IV-4 is possible through transition metal-catalyzed coupling processes, such as Stille or boronic acid couplings using a PdLn catalyst (wherein L is a ligand such as triphenylphosphine, tri-tert-butylphosphine or xantphos) to form hydroxypyridines IV-5, or amination chemistry to form hydroxypyridines IV-6 in which R2 is N(RA)RB.

Scheme IV

Figure imgf000040_0002

IV-1

Figure imgf000040_0003

- – Scheme V depicts the introduction of substitution at the five-position of the hydroxypyridines via bromination, and subsequent transition metal-catalyzed chemistries, such as Stille or boronic acid couplings using PdLn in which L is as defined in Scheme IV to form hydroxypyridines V-3, or amination chemistry to form hydroxypyridines V-4 in which R3 is N(RA)RB.

Scheme V

Figure imgf000041_0001

As shown in Scheme IV, fiuorohydroxypyridines II-l (Scheme II) are available from the commercially available 3-fluoroypridines VI- 1 through N-oxide formation and rearrangement as described in Konno et al., Heterocycles 1986, vol. 24, p. 2169.

Scheme VI

Figure imgf000041_0002

The following examples serve only to illustrate the invention and its practice. The examples are not to be construed as limitations on the scope or spirit of the invention.

The term “room temperature” in the examples refers to the ambient temperature which was typically in the range of about 20°C to about 26°C.

EXAMPLE 1

3-Chloro-5-({ l-[(4-methyl-5-oxo-4,5-dihydro-lH-l ,2,4-triazol-3-yl)methyl]-2-oxo-4- (trifluoromethyl)-l ,2-dihydropyridin-3-yl}oxy)benzonitrile (1-1)

 

Figure imgf000042_0001

Step 1(a):

 

Figure imgf000042_0002

A mixture of the 3-bromo-5-chlorophenol (3.74 g; 18.0 mmol), 2-chloro-3-fluoro- 4-(trifluoromethyl)pyridine (3.00 g; 15.0 mmol) and 2CO3 (2.49 g; 18.0 mmol) in NMP (15 mL) was heated to 120°C for one hour, then cooled to room temperature. The mixture was then diluted with 250 mL EtOAc and washed with 3 x 250 mL 1 :1 H20:brine. The organic extracts were dried (Na2S04) and concentrated in vacuo. Purification by ISCO CombiFlash (120 g column; load with toluene; 100:0 to 0:100 hexanes:CH2Cl2 over 40 minutes) provided title compound (1-2) as a white solid. Repurification of the mixed fractions provided additional title compound. lH NMR (400 MHz, CDCI3): δ 8.55 (d, J = 5.0 Hz, 1 H); 7.64 (d, J = 5.0 Hz, 1 H);

7.30 (s, 1 H); 6.88 (s, 1 H); 6.77 (s, 1 H).

3-(3-bromo-5-chlorophenoxy)-4-(trifluoromethyl)pyridin-2-ol (1-3)

 

Figure imgf000042_0003

To a suspension of 3-(3-bromo-5-chlorophenoxy)-2-chloro-4- (trifluoromethyl)pyridine (1-2; 3.48 g; 8.99 mmol) in lBuOH (36 mL) was added KOH (1.51 g; 27.0 mmol) and the mixture was heated to 75°C overnight, at which point a yellow oily solid had precipitated from solution, and LCMS analysis indicated complete conversion. The mixture was cooled to room temperature, and neutralized by the addition of -50 mL saturated aqueous NH4CI. The mixture was diluted with 50 mL H2O, then extracted with 2 x 100 mL EtOAc. The combined organic extracts were dried (Na2S04) and concentrated in vacuo. Purification by ISCO CombiFlash (120 g column; dry load; 100:0 to 90: 10 CH2Cl2:MeOH over 40 minutes) provided the title compound (1-3) as a fluffy white solid. lH NMR (400 MHz, DMSO): δ 12.69 (s, 1 H); 7.59 (d, J = 6.9 Hz, 1 H); 7.43 (t, J = 1.7 Hz, 1 H); 7.20 (t, J = 1.9 Hz, 1 H); 7.13 (t, J = 2.0 Hz, 1 H); 6.48 (d, J = 6.9 Hz, 1 H).

3-chloro-5-{[2-hydroxy-4-(trifluoromethyl)pyridin-3-yl]oxy}benzonitrile (1-4)

 

Figure imgf000043_0001

To a suspension of 3-(3-bromo-5-chlorophenoxy)-4-(trifluoromethyl)pyridin-2-ol (1-3; 3.25 g; 8.82 mmol) in NMP (29 mL) was added CuCN (7.90 g; 88 mmol) and the mixture was heated to 175°C for 5 hours, then cooled to room temperature slowly. With increased fumehood ventilation, 100 mL glacial AcOH was added, then 100 mL EtOAc and the mixture was filtered through Celite (EtOAc rinse). The filtrate was washed with 3 x 200 mL 1 : 1 H20:brine, then the organic extracts were dried (Na2S04) and concentrated in vacuo.

Purification by ISCO CombiFlash (120 g column; dry load; 100:0 to 90:10 CH2Cl2:MeOH over 40 minutes), then trituration of the derived solid with Et20 (to remove residual NMP which had co-eluted with the product) provided the title compound (1-4). lH NMR (400 MHz, DMSO): δ 12.71 (s, 1 H); 7.75 (s, 1 H); 7.63-7.57 (m, 2 H); 7.54 (s, 1 H); 6.49 (d, J = 6.9 Hz, 1 H).

Step 1(d): 5-(chloromethyl)-2,4-dihydro-3H-l,2,4-triazol-3-one (1-5)

Figure imgf000043_0002

The title compound was prepared as described in the literature: Cowden, C. J.; Wilson, R. D.; Bishop, B. C; Cottrell, I. F.; Davies, A. J.; Dolling, U.-H. Tetrahedron Lett. 2000, 47, 8661.

3 -chloro-5 -( { 2-oxo- 1 – [(5 -oxo-4,5 -dihydro- 1 H- 1 ,2,4-triazol-3 -yl)methyl] – 4- (trifiuoromethyl)- 1 ,2-dihydropyridin-3 -yl } oxy)benzonitrile (1-6)

Figure imgf000044_0001

A suspension of the 3-chloro-5-{[2-hydroxy-4-(trifluoromethyl)pyridin-3- yl]oxy}benzonitrile (1-4; 2.00 g; 6.36 mmol), 5-(chloromethyl)-2,4-dihydro-3H-l,2,4-triazol-3- one (1-5; 0.849 g; 6.36 mmol) and K2CO3 (0.878 g; 6.36 mmol) in DMF (32 mL) was stirred for 2 hours at room temperature, at which point LCMS analysis indicated complete conversion. The mixture was diluted with 200 mL Me-THF and washed with 150 mL 1 : 1 : 1 H20:brine:saturated aqueous NH4CI, then further washed with 2 x 150 mL 1 : 1 H20:brine. The aqueous fractions were further extracted with 150 mL Me-THF, then the combined organic extracts were dried (Na2S04) and concentrated in vacuo. Purification by ISCO CombiFlash (80 g column; dry load; 100:0 to 90:10 EtOAc:EtOH over 25 minutes) provided the title compound (1-6) as a white solid. lH NMR (400 MHz, DMSO): δ 1 1.46 (s, 1 H); 1 1.39 (s, 1 H); 7.93 (d, J = 7.3 Hz, 1 H); 7.76 (s, 1 H); 7.58 (s, 1 H); 7.51 (s, 1 H); 6.67 (d, J = 7.3 Hz, 1 H); 5.02 (s, 2 H).

Step 1(f): 3 -chloro-5 -( { 1 – [(4-methyl-5-oxo-4,5 -dihydro- 1 H- 1 ,2,4-triazol-3 -yl)methyl] -2- oxo-4-(trifluoromethyl)- 1 ,2-dihydropyridin-3 -yl } oxy)benzonitrile (1 -1 )

A solution of 3-chloro-5-({2-oxo-l -[(5-oxo-4,5-dihydro-lH-l,2,4-triazol-3- yl)methyl]- 4-(trifluoromethyl)-l ,2-dihydropyridin-3-yl}oxy)benzonitrile (1-6; 2.37 g; 5.76 mmol) and K2CO3 (0.796 g; 5.76 mmol) in DMF (58 mL) was cooled to 0°C, then methyl iodide (0.360 mL; 5.76 mmol) was added. The mixture was allowed to warm to room

temperature, and stirred for 90 minutes, at which point LCMS analysis indicated >95%

conversion, and the desired product of -75% LCAP purity, with the remainder being unreacted starting material and 6/s-methylation products. The mixture was diluted with 200 mL Me-THF, and washed with 3 x 200 mL 1 : 1 H20:brine. The aqueous fractions were further extracted with 200 mL Me-THF, then the combined organic extracts were dried (Na2S04) and concentrated in vacuo. The resulting white solid was first triturated with 100 mL EtOAc, then with 50 mL THF, which provided (after drying) the title compound (1-1) of >95% LCAP. Purification to >99% LCAP is possible using Prep LCMS (Max-RP, 100 x 30 mm column; 30-60% CH3CN in 0.6% aqueous HCOOH over 8.3 min; 25 mL/min). lH NMR (400 MHz, DMSO): δ 1 1.69 (s, 1 H); 7.88 (d, J = 7.3 Hz, 1 H); 7.75 (s, 1 H); 7.62 (s, 1 H); 7.54 (s, 1 H); 6.67 (d, J = 7.3 Hz, 1 H); 5.17 (s, 2 H); 3.1 1 (s, 3 H). EXAMPLE 1A

3-Chloro-5-({ l-[(4-methyl-5-oxo-4,5-dihydro-lH-l ,2,4-triazol-3-yl)methyl]-2- (trifluoromethyl)-l ,2-dihydropyridin-3-yl}oxy)benzonitrile (1-1)

 

Figure imgf000045_0001

Step lA(a): 2-chloro-3-(3-chloro-5-iodophenoxy)-4-(trifluoromethyl)pyridine (1A-2)

 

Figure imgf000045_0002

A mixture of the 3-chloro-l-iodophenol (208 g; 816.0 mmol), 2-chloro-3-fluoro-

4-(trifluoromethyl)pyridine (155 g; 777.0 mmol) and K2CO3 (161 g; 1 165.0 mmol) in NMP (1.5 L) was held at 60°C for 2.5 hours, and then left at room temperature for 2 days. The mixture was then re-heated to 60°C for 3 hours, then cooled to room temperature. The mixture was then diluted with 4 L EtOAc and washed with 2 L water + 1 L brine. The combined organics were then washed 2x with 500 mL half brine then 500 mL brine, dried over MgS04 and concentrated to afford crude 1A-2. lH NMR (500 MHz, DMSO) δ 8.67 (d, J = 5.0 Hz, 1 H), 7.98 (d, J = 5.0 Hz, 1 H), 7.63-7.62 (m, 1 H), 7.42-7.40 (m, 1 H), 7.22 (t, J = 2.1 Hz, 1 H).

Step lA(b): 2-chloro-3-(3-chloro-5-iodophenoxy)-4-(trifluoromethyl)pyridine (1A-3)

 

Figure imgf000045_0003

To a suspension of 3-(3-chloro-5-iodophenoxy)-2-chloro-4- (trifluoromethyl)pyridine (1A-2; 421 g, 970 mmol) in t-BuOH (1 L) was added KOH (272 g, 4850 mmol) and the mixture was heated to 75°C for 1 hour, at which point HPLC analysis indicated >95% conversion. The t-BuOH was evaporated and the mixture diluted with water (7mL/g, 2.4L) and then cooled to 0°C, after which 12N HC1 (~240mL) was added until pH 5. This mixture was then extracted with EtOAc (20mL/g, 6.5L), back extracted with EtOAc 1 x 5mL/g (1.5L), washed 1 x water:brine 1 : 1 (l OmL/g, 3.2L), 1 x brine (lOmL/g, 3.2L), dried over MgS04, filtered and concentrated to afford a crude proudct. The crude product was suspended in MTBE (2.25 L, 7mL/g), after which hexanes (1 L, 3 mL/g) was added to the suspension over ten minutes, and the mixturen was aged 30minutes at room temperature. The product was filtered on a Buchner, rinsed with MTBE hexanes 1 :2 (2 mL/g = 640 mL), then hexanes

(640mL), and dried on frit to afford 1A-3. lH NMR (400 MHz, acetone-d6): δ 11.52 (s, 1 H); 7.63 (d, J = 7.01 Hz, 1 H); 7.50-7.48 (m, 1 H); 7.34-7.32 (m, 1 H); 7.09-7.07 (m, 1 H); 6.48 (d, J = 7.01 Hz, 1 H).

Step lA(c): 3-chloro-5-{[2-hydroxy-4-(trifluoromethyl)pyridin-3-yl]oxy}benzonitrile (1-4)

 

Figure imgf000046_0001

A solution of 3-(3-chloro-5-iodophenoxy)-4-(trifluoromethyl)pyridin-2-ol (1A-3; 190 g; 457 mmol) in DMF (914 mL) was degassed for 20 minutes by bubbling N2, after which CuCN (73.7 g; 823 mmol) was added, and then the mixture was degassed an additional 5 minutes. The mixture was then heated to 120°C for 17 hours, then cooled to room temperature and partitioned between 6 L MeTHF and 2 L ammonium buffer (4:3: 1 = NH4CI

sat/water/NH-iOH 30%). The organic layer washed with 2 L buffer, 1 L buffer and 1 L brine then, dried over MgS04 and concentrated. The crude solid was then stirred in 2.2 L of refluxing

MeCN for 45 minutes, then cooled in a bath to room temperature over 1 hour, aged 30 minutes, then filtered and rinsed with cold MeCN (2 x 400mL). The solid was dried on frit under N2 atm for 60 hours to afford title compound 1-4. lH NMR (400 MHz, DMSO): δ 12.71 (s, 1 H); 7.75 (s, 1 H); 7.63-7.57 (m, 2 H); 7.54 (s, 1 H); 6.49 (d, J = 6.9 Hz, 1 H).

Steps lA(d) and lA(e)

The title compound 1-1 was then prepared from compound 1-4 using procedures similar to those described in Steps 1(d) and 1(e) set forth above in Example 1.

New patent

WO-2014052171

Crystalline anhydrous Form II of doravirine, useful for the treatment of HIV-1 and HIV-2 infections. The compound was originally claimed in WO2008076223. Also see WO2011120133. Merck & Co is developing doravirine (MK-1439), for the oral tablet treatment of HIV-1 infection. As of April 2014, the drug is in Phase 2 trials.

References

  1.  Safety and Antiviral Activity of MK-1439, a Novel NNRTI, in Treatment-naïve HIV+ Patients. Gathe, Joseph et al. 20th Conference on Retroviruses and Opportunistic Infections. 3–6 March 2013. Abstract 100.
  2.  CROI 2013: MK-1439, a Novel HIV NNRTI, Shows Promise in Early Clinical Trials. Highleyman, Liz. HIVandHepatitis.com. 6 March 2013.

The next-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) doravirine (formerly MK-1439) showed potent antiretroviral activity and good tolerability in combination with tenofovir/FTC (the drugs in Truvada) in a dose-finding study presented at the 21st Conference on Retroviruses and Opportunistic Infections (CROI) last week in Boston.

NNRTIs are generally well tolerated and well suited for first-line HIV treatment, but as a class they are susceptible to resistance. Pre-clinical studies showed that Merck’s doravirine has a distinct resistance profile and remains active against HIV with common NNRTI resistance mutations including K103N and Y181C.

As reported at last year’s CROI, doravirine reduced HIV viral load by about 1.3 log in a seven-day monotherapy study. Doravirine is processed by the CYP3A4 enzyme, but it is neither a CYP3A4 inducer nor inhibitor, so it is not expected to have major drug interaction concerns.

Javier Morales-Ramirez from Clinical Research Puerto Rico reported late-breaking findings from a phase 2b study evaluating the safety and efficacy of various doses of doravirine versus efavirenz (Sustiva) for initial antiretroviral therapy.

This study included 208 treatment-naive people living with HIV from North America, Europe and Asia. More than 90% were men, 74% were white, 20% were black and the median age was 35 years. At baseline, the median CD4 cell count was approximately 375 cells/mm3 and 13% had received an AIDS diagnosis. Study participants were stratified by whether their viral load was above (about 30%) or below 100,000 copies/ml; median HIV RNA was approximately 4.5 log10.

Morales-Ramirez reported 24-week results from part 1 of the study, which will continue for a total of 96 weeks. In this part, participants were randomly allocated into five equal-sized arms receiving doravirine at doses of 25, 50, 100 or 200mg once daily, or else efavirenz once daily, all in combination with tenofovir/FTC.

At 24 weeks, 76.4% of participants taking doravirine had viral load below 40 copies/ml compared with 64.3% of people taking efavirenz. Response rates were similar across doravirine doses (25mg: 80.0%; 50mg: 76.2%; 100mg: 71.4%; 200mg: 78.0%). More than 80% of participants in all treatment arms reached the less stringent virological response threshold of <200 copies/ml.

Both doravirine and efavirenz worked better for people with lower pre-treatment viral load in an ad hoc analysis. For people with <100,000 copies/ml at baseline, response rates (<40 copies/ml) ranged from 83 to 89% with doravirine compared with 74% with efavirenz. For those with >100,000 copies/ml, response rates ranged from 50 to 91% with doravirine vs 54% with efavirenz.

Median CD4 cell gains were 137 cells/mm3 for all doravirine arms combined and 121 cells/mmfor the efavirenz arm.

Doravirine was generally safe and well tolerated. People taking doravirine were less than half as likely as people taking efavirenz to experience serious adverse events (3.0% across all doravirine arms vs 7.1% with efavirenz) or to stop treatment for this reason (2.4 vs 4.8%). Four people taking doravirine and two people taking efavirenz discontinued due to adverse events considered to be drug-related.

The most common side-effects were dizziness (3.6% with doravirine vs 23.8% with efavirenz), abnormal dreams (9.0 vs 7.1%), diarrhoea (4.8 vs 9.5%), nausea (7.8 vs 2.4%) and fatigue (6.6 vs 4.8%). Other central nervous system (CNS) adverse events of interest included insomnia (5.4 vs 7.1%), nightmares (1.2 vs 9.5%) and hallucinations (0.6 vs 2.4%). Overall, 20.5% of people taking doravirine reported at least one CNS side-effect, compared with 33.3% of people taking efavirenz.

People taking doravirine had more favourable lipid profiles and less frequent liver enzyme (ALT and AST) elevations compared with people taking efavirenz.

The researchers concluded that doravirine demonstrated potent antiretroviral activity in treatment-naive patients, a favourable safety and tolerability profile, and fewer drug-related adverse events compared with efavirenz.

Based on these findings, the 100mg once-daily dose was selected for future development and will be used in part 2 of this study, a dose-confirmation analysis that will enrol an additional 120 participants.

In the discussion following the presentation, Daniel Kuritzkes from Harvard Medical School noted that sometimes it takes longer for viral load to go down in people who start with a high level, so with further follow-up past 24 weeks doravirine may no longer look less effective in such individuals.

Reference

Morales-Ramirez J et al. Safety and antiviral effect of MK-1439, a novel NNRTI (+FTC/TDF) in ART-naive HIV-infected patients. 21st Conference on Retroviruses and Opportunistic Infections, Boston, abstract 92LB, 2014.

Merck Moves Doravirine Into Phase 3 Clinical Trials

Wednesday Mar 19 | Posted by: roboblogger | Full story: EDGE

Earlier this month, at the 21st Conference on Retroviruses and Opportunistic Infections , Merck indicated plans to initiate a Phase 3 clinical trial program for doravirine in combination with ARV therapy in the second half of 2014.

 

 


Filed under: Phase3 drugs, Uncategorized Tagged: doravirine, MK 1439

Cabotegravir, GSK 744 IN PHASE 2 FOR HIV INFECTION

$
0
0

GSK744.svg

Cabotegravir, GSK 744,

(3S,11aR)-N-(2,4-Difluorobenzyl)-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide

3S, 1 1 aR)- N-[(2,4-difluorophenyl)methyl]-2,3,5,7, 1 1 , 1 1 a-hexahydro-6-hydroxy-3- methyl-5,7- dioxo-oxazolo[3,2-a]pyrido[1 ,2-d]pyrazine-8-carboxamide

OTHER ISOMER

(3R,11 aS)-N-[(2,4-Diflυorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7, 11, 11a-hexahydro[1 ,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide

VIIV HEALTHCARE …INNOVATOR

  • GSK1265744, CAS 1051375-10-0, S-265744 LAP
  • C19-H17-F2-N3-O5
  • 405.3553

2D chemical structure of 1051375-10-0

GSK744 (also known as S/GSK1265744) is an investigational new drug under development for the treatment of HIV infection. It is anintegrase inhibitor, with a carbamoyl pyridone structure similar to dolutegravir. In investigational studies, the agent has been packaged into nanoparticles (GSK744LAP) conferring an exceptionally long half-life of 21–50 days following a single dose. In theory, this would make possible suppression of HIV with dosing as infrequently as once every three months.[1]

S-265744 LAP is in phase II clinical development at Shionogi-GlaxoSmithKline for the treatment of HIV infection. Phase III clinical trials had been ongoing for this indication; however, no recent development has been reported for this study.

The human immunodeficiency virus (“HIV”) is the causative agent for acquired immunodeficiency syndrome (“AIDS”), a disease characterized by the destruction of the immune system, particularly of CD4+ T-cells, with attendant susceptibility to opportunistic infections, and its precursor Al DS-related complex (“ARC”), a syndrome characterized by symptoms such as persistent generalized lymphadenopathy, fever and weight loss. HIV is a retrovirus; the conversion of its RNA to DNA is accomplished through the action of the enzyme reverse transcriptase. Compounds that inhibit the function of reverse transcriptase inhibit replication of HIV in infected cells. Such compounds are useful in the prevention or treatment of HIV infection in humans.

A required step in HIV replication in human T-cells is the insertion by virally-encoded integrase of proviral DNA into the host cell genome. Integration is believed to be mediated by integrase in a process involving assembly of a stable nucleoprotein complex with viral DNA sequences, cleavage of two nucleotides from the 3′ termini of the linear proviral DNA and covalent joining of the recessed 3′ OH termini of the proviral DNA at a staggered cut made at the host target site. The repair synthesis of the resultant gap may be accomplished by cellular enzymes. There is continued need to find new therapeutic agents to treat human diseases. HIV integrase is an attractive target for the discovery of new therapeutics due to its important role in viral infections, particularly HIV infections. Integrase inhibitors are disclosed in WO2006/116724.

(3S, 1 1 aR)- N-[(2,4-difluorophenyl)methyl]-2,3,5,7, 1 1 , 1 1 a-hexahydro-6-hydroxy-3- methyl-5,7- dioxo-oxazolo[3,2-a]pyrido[1 ,2-d]pyrazine-8-carboxamide, a compound of formula (I), also referred to as compound (I), has proven antiviral activity against human immunodeficiency virus (HIV).

Figure imgf000004_0001

The present invention features pharmaceutical compositions comprising the active ingredient (3S, 1 1 aR)- N-[(2,4-difluorophenyl)methyl]-2,3,5,7, 1 1 , 1 1 a-hexahydro-6-hydroxy-3- methyl-5,7- dioxo-oxazolo[3,2-a]pyrido[1 ,2-d]pyrazine-8-carboxamide, or a pharmaceutically acceptable salt thereof, suitable for administration once monthly or longer.

Methods for the preparation of a compound of formula (I) are described in WO 2006/1 16764, WO2010/01 1814, WO2010/068262, and WO2010/068253

WO 2006116764 

http://www.google.com/patents/WO2006116764A1?cl=en

Figure imgf000058_0001

[Chemical formula 68]  is  UNDESIRED ISOMER………..amcrasto@gmail.com

Figure imgf000122_0001

Example Z-1:

(3R,11 aS)-N-[(2,4-Diflυorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7, 11, 11a

-hexahydro[1 ,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide sodium salt.

 

Figure imgf000130_0002

(3R,11aS)-N-[(2,4-Diflυorophenyl)methyl]-3-methyl-5,7-dioxo-6-[(phenylmethyl)oxy]-2,

3,5,7,11,11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide. To a solution of 16a (409 mg, 0.87 mmol) in dichloroethane (20 mL) was added (2R)-2-amino-1-propanol (0,14 mL, 1.74 mmol) and 10 drops of glacial acetic acid.

The resultant solution was heated at reflux for 2 h. Upon cooling, Celite was added

to the mixture and the solvents removed in vacuo and the material was purified via

silica gel chromatography (2% CH3OH/CH2CI2 gradient elution) to give

(3R),11aS)-N-[(2,4-difluorophenyl)methyl]-3-methyl-5,7-dioxo-6- [(phenylmethyl)oxy]-2,

3,5,7, 1 l , 11a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazinc-8-carboxamide (396

mg, 92%) as a glass, JH NMR (CDCIo) δ 10.38 (m, 1 H), 8.42 (s, 1 H), 7,54-7,53 (m, 2

H), 7,37-7.24 (m, 4 H), 6.83-6,76 (m, 2 H), 5.40 (d, J = 10.0 Hz, 1 H), 5.22 (d, J = 10,0

Hz, 1 H), 5.16 (dd, J – 9,6, 6.0 Hz, 1 H), 4,62 (m, 2 H), 4.41 (m, 1 H), 4.33-4.30 (m, 2

H), 3.84 (dd, J= 12.0, 10.0 Hz, 1 H), 3.63 (dd, J= 8,4, 7.2 Hz, 1 H), 1.37 (d, J= 6.0 Hz,

3 H); ES+ MS: 496 (M+1).

b)

(3R, 11aS)-N-[(2,4-Difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7, 11, 1la

-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8vcarboxamide sodium salt. To a

solution of

(37?, 11aS)-N-[(2,4-difluo]-ophenyl)methyl]-3-methyl-5,7-dioxo-6- [(phenylmethyl)oxy] -2,

3,5,7,11,11 a-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide (396

mg, 0.80 mmol) in methanol (30 mL) was added 10% Pd/C (25 mg). Hydrogen was

bubbled through the reaction mixture via a balloon for 2 h. The resultant mixture

was filtered through Celite with methanol and dichloromethanc. The filtrate was

concentrated in vacuo to give

(3R, l] aS)-N-f(2,4-difliιorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7, υ , 11a- hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide as a pink tinted

white solid (278 mg, 86%), 1H NMR (ODCU) δ 11.47 (m, 1 H), 10.29 (m, 1 H), 8,32 (s,

1 H), 7.36 (m, 1 H), 6.82 (m, 2 H), 5.31 (dd, J – 9.6, 3.6 Hz, 1 H), 4.65 (m, 2 H),

4,47-4,38 (m, 3 H), 3.93 (dd, J= 12.0, 10.0 Hz, 1 H), 3,75 (m, 1 H), 1.49 (d, J= 5.6 Hz,

3 H); BS1 MS: 406 (M+ 1). The above material (278 mg, 0,66 mmol) was taken up

m cthanol (10 mL) and treated with 1 Nsodium hydroxide (aq) (0.66 mL, 0.66 mmol).

The resulting suspension was stirred at room temperature for 30 min, Ether was

added and the liquids were collected to provide the sodium salt of the title compound

as a white powder (291 mg, 99%).‘ 1H NMR (OMSO- do) δ 30.68 (m, 1 H), 7,90 (s, 1 H),

7.35 (m, 1 H), 7.20 (m, 1 H), 7,01 (m, 1 H), 5,20 (m, 1 H), 4,58 (m, I H), 4.49 (m, 2 H),

4.22 (m, 2 H), 3 74 (dd, J= 11.2, 10.4 Hz, 1 H), 3.58 (m, 1 H), 1.25 (d, J=- 4.4 Hz, 3 H)

DESIRED ISOMER………… ANY ERROR ………….amcrasto@gmail.com

Example Z-9-

(3£ 11aΛ^N-[(2.4-D-fluoroDhonyl)methyl] -6-hvdroxy-3-methyl-5.7-dioxo-2,3,5.7, n , 11 a

-hexahydro[1 ,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazino-8-carboxamide sodium salt.

 

Figure imgf000144_0001

The title compound was made in two steps using a similar process to that described

in example Z-I. 16a (510 mg, 1.08 mmol) and (2«5)-2-amino-1-propanol (0.17 mL, 2,17 mmol) were reacted in 1,2-dichloroethane (20 mL) with acetic acid to give

(3S, 11aR)-i\A[(2,4-diflιιorophenyl)methyl]-3-methyl-5,7-d.ioxo-6-[(phenylmethyl)oxy]-2,

3,5,7,11,1la-hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyrazine-8-carboxamide (500

mg, 93%). This material was hydrogenated in a second step as described in example

Z- I to give

3S, 11a R)-7N-[(2,4-Diiαuorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo-2,3,5,7, 11, 11a-

hexahydro[1,3]oxazolo[3,2-a]pyrido[1,2-d]pyraziine-8-carboxamide (386 mg, 94%) as a

tinted white solid. Η NMR (CDCL3) δ 11.46 (m, 1 H), 10.28 (m, 1 H), 8.32 (s, 1 H),

7.35 (m, 1 H), 6.80 (m, 2 H), 5.30 (dd, J = 10.0, 4.0 Hz, 1 H), 4.63 (m, 2 H), 4.48-4.37

(m, 3 H), 3.91 (dd, J = 12.0, 10.0 Hz, 1 H), 3.73 (m, 1 H), 1.48 (d, J – 6.0 Hz, 3 H);

ES 1 MS: 406 (M+ 1). This material (385 mg, 0.95 mmol) was treated with sodium

hydroxide (0,95 mL, 1.0 M, 0.95 mmol) m ethanol (15 mL) as described in example Z-1

to provide its corresponding sodrum sail (381 mg, 94%) as a white solid. 1H NMR

(DMSO- Λ) δ 10.66 (m, 1 PI), 7.93 (s, 1 H), 7.33 (m, 1 H), 7.20 (m, 1 H), 7.01 (m, 1 H),

5.19 (m, 1 H), 4.59 (m, 1 H), 4 48 (m, 2 H), 4.22 (m, 2 H), 3,75 (m, 1 H), 3.57 (m, 1 H),

1.24 (d, J= 5 6 Hz, 3 H).

…………………..

WO 2010068253 

http://www.google.com/patents/WO2010068253A1?cl=en

Example A

The starting material of Example A is compound 8, which is identical to formula (Ia). Thus, Example A depicts a process in providing an intermediate for the compound of formula 17 below which is isomeric to the compound ZZ-2 at page 237 of WO 2006/116764 to Brian Johns et al.

Figure imgf000030_0001

14

Figure imgf000030_0002
Figure imgf000030_0003

Example Aa After dissolution of mixture of 320 g of compound 8 (1.0 eq.) in 3.20 L of MeOH by heating, the solution was concentrated. To the residue, 1.66 L of MeCN, 5.72 mL of AcOH(0.1 eq.) and 82.6 g of (S)-2-Amino-propan-1-ol(1.1 eq.) were added and the mixture was heated to 70 °C, stirred at 70 0C for 4 h and concentrated. To the residue, 1.67 L of 2-propanol was added and the mixture was concentrated (twice). After cooling of the residue, filtration, washing with 500 mL of cold 2-propanol and drying provided 167 g of compound 14 (52% yield) as a crystal. 1H NMR(300 MHz1 CDCI3) δ 7.61-7.55 (m, 2H), 7.40-7.20 (m, 4H), 6.53 (d, J = 7.2, 1H), 5.46 (d, J = 10.5 Hz, 1H), 5.23 (d, J = 10.2 Hz, 1H), 5.20 (dd, J = 3.9, 9.6 Hz, 1H), 4.46- 4.34 (m, 1H)1 4.31 (dd, J = 6.6, 8.7 Hz, 1H)1 4.14 (dd, J = 3.9, 12.3 Hz1 1H)1 3.79 (dd, J = 9.9, 12.3 Hz1 1 H), 3.62 (dd, J = 6.9, 8.7 Hz1 1 H), 1.38 (d, J = 6.3 Hz1 3H).

Example Ab

To slurry of 156 g of compound 14 (1.0 eq.) in 780 ml_ of NMP was added 93.6 g of NBS(1.1 eq.) and the mixture was stirred at room temperature for 2.5 h. The reaction mixture was added to 3.12 L of H2O. Filtration, washing with 8.0 L of H2O and drying provided 163 g of compound 15 (84% yield) as a crystal.

1H NMR(300 MHz, DMSO-CT6) δ 8.37 (s, 1H), 7.55-7.50 (m, 2H), 7.42-7.25 (m, 3H), 5.34 (dd, J = 3.6, 9.9 Hz, 1H), 5.18 (d, J = 10.8 Hz, 1H), 5.03 (d, J = 10.5 Hz, 1H), 4.53 (dd, J = 3.6, 12.0 Hz, 1H)1 4.40-4.20 (m, 2H), 3.99 (dd, J = 9.9, 11.7 Hz1 1H), 3.64 (dd, J = 5.7, 8.1 Hz1 1 H)1 1.27 (d, J = 6.3 Hz1 3H).

Example Ac

Under carbon mono-oxide atmosphere, a mixture of 163 g of compound 15 (1.0 eq.), 163 mL of /-Pr2NEt(2.5 eq.), 68.4 ml_ of 2,4-difluorobenzylamine(1.5 eq.) and 22.5 g of Pd(PPh3)4(0.05 eq.) in 816 mL of DMSO was stirred at 90 0C for 7 h. After cooling, removal of precipitate, washing with 50 mL of DMSO and addition of 11.3 g of

Pd(PPh3)4(0.025 eq.), the reaction mixture was stirred at 90 0C for 2 h under carbon mono-oxide atmosphere again. After cooling, removal of precipitate and addition of 2.0 L of AcOEt and 2.0 L of H2O1 the organic layer was washed with 1.0 L of 1 N HCIaq. and 1.0 L of H2O (twice) and the aqueous layer was extracted with 1.0 L of AcOEt. The organic layers were combined and concentrated. Silica gel column chromatography of the residue provided 184 g of compound 16 (96% yield) as foam.

1H NMR(300 MHz, CDCI3) δ 10.38 (t, J = 6.3 Hz1 1H)1 8.39 (s, 1H)1 7.75-7.25 (m, 7H), 6.90-6.70 (m, 2H), 5.43 (d, J = 10.2 Hz, 1H), 5.24 (d, J = 10.2 Hz, 1H)1 5.19 (dd, J = 3.9, 9.9 Hz, 1H)1 4.63 (d, J = 6.0 Hz, 2H), 4.50-4.25 (m, 3H)1 3.86 (dd, J = 9.9, 12.3 Hz, 1H), , 3.66 (dd, J = 6.9, 8.4 Hz1 1 H), 1.39 (d, J = 6.0 Hz, 3H).

Example Ad

Under hydrogen atmosphere, a mixture of 184 g of compound 16 (1.0 eq.) and 36.8 g of 10%Pd-C in 3.31 L of THF and 0.37 L of MeOH was stirred for 3 h. After filtration of precipitate(Pd-C), washing with THF/MeOH(9/1 ) and addition of 36.8 g of 10% Pd-C, the mixture was stirred for 20 min under hydrogen atmosphere. After filtration of precipitate(Pd-C) and washing with THF/MeOH(9/1), the filtrate was concentrated. After 200 ml_ of AcOEt was added to the residue, filtration afforded crude solid of compound 17. The precipitates were combined and extracted with 4.0 L of CHCl3/MeOH(5/1). After concentration of the CHCI3ZMeOH solution and addition of 250 ml_ of AcOEt to the residue, filtration afforded crude solid of compound 17. The crude solids were combined and dissolved in 8.2 L of MeCN/H2O(9/1 ) by heating. After filtration, the filtrate was concentrated. To the residue, 1.5 L of EtOH was added and the mixture was concentrated (three times). After cooling of the residue, filtration and drying provided 132 g of compound 17 (88% yield) as a crystal. 1H NMR(300 MHz, DMSO-cfe) δ 11.47 (brs, 1H), 10.31 (t, J = 6.0 Hz, 1H), 8.46 (s, 1H), 7.40 (td, J = 8.6, 6.9 Hz, 1H), 7.24 (ddd, J = 2.6, 9.4, 10.6, 1H), 7.11-7.01 (m, 1H), 5.39 (dd, J = 4.1, 10.4 Hz, 1H), 4.89 (dd, J = 4.2, 12.3 Hz, 1H), 4.55 (d, J = 6.0 Hz, 2H), 4.40 (dd, J = 6.8, 8.6 Hz, 1H), 4.36-^.22 (m, 1H)1 4.00 (dd, J = 10.2, 12.3 Hz, 1H), 3.67 (dd, J = 6.7, 8.6 Hz, 1H), 1.34 (d, J = 6.3 Hz, 3H).

Example Ae

After dissolution of 16.0 g of compound 17 (1.0 eq.) in 2.56 L of EtOH and 0.64 L of H2O by heating, followed by filtration, 39 ml_ of 1N NaOHaq.(1.0 eq.) was added to the solution at 75 0C. The solution was gradually cooled to room temperature. Filtration, washing with 80 ml_ of EtOH and drying provided 13.5 g of compound 18 (80% yield) as a crystal.

1H NMR(300 MHz, DMSO-cfe) δ 10.73 (t, J = 6.0 Hz, 1H), 7.89 (s, 1H), 7.40-7.30 (m, 1H), 7.25-7.16 (m, 1H), 7.07-6.98 (m, 1H), 5.21 (dd, J = 3.8, 10.0 Hz, 1H), 4.58 (dd, J = 3.8, 12.1 Hz, 1H), 4.51 (d, J = 5.4 Hz, 2H), 4.3CM.20 (m, 2H), 3.75 (dd, J = 10.0, 12.1 Hz, 1H), 3.65-3.55 (m, 1H), 1.27 (d, J = 6.1 Hz, 3H).

………………

WO2010011814

http://www.google.st/patents/WO2010011814A1?cl=en&hl=pt-PT

Scheme 1

Figure imgf000016_0001

2a 2b

Scheme 2

Figure imgf000016_0002

Scheme 3

Figure imgf000016_0003

Scheme 4

phosphorylation

Figure imgf000017_0001

Figure imgf000017_0002

Scheme 5

Hydrogenolysis

Figure imgf000017_0003
Figure imgf000017_0004

Figure imgf000017_0005

The following examples are intended for illustratation only and are not intended to limit the scope of the invention in any way. Preparation 1 : (3S.11 af?VΛ/-r(2.4-DifluoroDhenvnmethyll-6-hvdroxy-3-methyl-5.7-dioxo- 2,3,5,7, 11 ,11 a-hexahydroM ,31oxazolor3,2-alpyridori ,2-c/1pyrazine-8-carboxamide sodium salt (compound 1 b, scheme 2).

I) MsCI, Et3N

Figure imgf000018_0001

2) DBU

P-1 P-2 P-3

Figure imgf000018_0002

a) Synthesis of 2-methyl-3-[(phenylmethvl)oxvl-4/-/-pvran-4-one (compound P-2). To a slurry of 2000 g of compound P-1(1.0 eq.) in 14.0 L of MeCN were added 2848 g of benzyl bromide(1.05 eq.) and 2630 g of K2CO3(1.2 eq.). The mixture was stirred at 80 0C for 5 h and cooled to 13°C. Precipitate was filtered and washed with 5.0 L of MeCN. The filtrate was concentrated and 3.0 L of THF was added to the residue. The THF solution was concentrated to give 3585 g of crude compound P-2 as oil. Without further purification, compound P-2 was used in the next step. 1H NMR(300 MHz, CDCI3) δ 7.60 (d, J = 5.7 Hz, 1 H), 7.4-7.3 (m, 5H), 6.37 (d, J = 5.7 Hz, 1 H), 5.17 (s, 2H), 2.09 (s, 3H).

b) Synthesis of 2-(2-hydroxy-2-phenylethyl)-3-[(phenylmethyl)oxy]-4H-pyran-4-one (compound P-3). To 904 g of the crude compound P-2 was added 5.88 L of THF and the solution was cooled to -60 0C. 5.00 L of 1.0 M of Lithium bis(trimethylsilylamide) in THF(1.25 eq.) was added dropwise for 2 h to the solution of compound 2 at -60 0C. Then, a solution of 509 g of benzaldehyde(1.2 eq.) in 800 ml. of THF was added at -60 0C and the reaction mixture was aged at -60 0C for 1 h. The THF solution was poured into a mixture of 1.21 L of conc.HCI, 8.14 L of ice water and 4.52 L of EtOAc at less than 2 0C.

The organic layer was washed with 2.71 L of brine (twice) and the aqueous layer was extracted with 3.98 L of EtOAc. The combined organic layers were concentrated. To the mixture, 1.63 L of toluene was added and concentrated (twice) to provide toluene slurry of compound P-3. Filtration, washing with 0.90 L of cold toluene and drying afforded 955 g of compound P-3 (74% yield from compound P-1 ) as a solid. 1H NMR(300 MHz, CDCI3) δ

7.62 (d, J = 5.7 Hz, 1 H), 7.5-7.2 (m, 10H), 6.38 (d, J = 5.7 Hz, 1 H), 5.16 (d, J = 11.4 Hz, 1 H), 5.09 (d, J = 11.4 Hz, 1 H), 4.95 (dd, J = 4.8, 9.0 Hz, 1 H), 3.01 (dd, J = 9.0, 14.1 Hz, 1 H), 2.84 (dd, J = 4.8, 14.1 Hz, 1 H).

c) Synthesis of 2-[(£)-2-phenylethenyl]-3-[(phenylmethyl)oxy]-4H-pyran-4-one (compound

P-4). To a solution of 882 g of compound P-3 (1.0 eq.) in 8.82 L of THF were added 416 g of Et3N(1.5 eq.) and 408 g of methanesulfonyl chloride(1.3 eq.) at less than 30 0C. After confirmation of disappearance of compound P-3, 440 ml. of NMP and 1167 g of DBU(2.8 eq.) were added to the reaction mixture at less than 30 0C and the reaction mixture was aged for 30 min. The mixture was neutralized with 1.76 L of 16% sulfuric acid and the organic layer was washed with 1.76 L of 2% Na2S03aq. After concentration of the organic layer, 4.41 L of toluene was added and the mixture was concentrated (tree times). After addition of 4.67 L of hexane, the mixture was cooled with ice bath. Filtration, washing with 1.77 L of hexane and drying provided 780 g of compound P-4 (94% yield) as a solid. 1H NMR(300 MHz, CDCI3) δ 7.69 (d, J = 5.7 Hz, 1 H), 7.50-7.25 (m, 10H), 7.22 (d, J = 16.2

Hz, 1 H), 7.03 (d, J = 16.2 Hz, 1 H), 6.41 (d, J = 5.7 Hz, 1 H), 5.27 (s, 2H). d) Synthesis of 4-oxo-3-[(phenylmethyl)oxy]-4H-pyran-2-carboxylic acid (compound P-5). To a mixture of 822 g of compound P-4 (1.0 eq.) and 1 1.2 g of RuCI3-nH2O(0.02 eq.) in 2.47 L of MeCN, 2.47 L of EtOAc and 2.47 L of H2O was added 2310 g of NalO4(4.0 eq.) at less than 25 0C. After aging for 1 h, 733 g of NaCIO2(S-O eq.) was added to the mixture at less than 25 0C. After aging for 1 h, precipitate was filtered and washed with 8.22 L of

EtOAc. To the filtrate, 1.64 L of 50% Na2S203aq, 822 ml. of H2O and 630 ml. of coc.HCI were added. The aqueous layer was extracted with 4.11 L of EtOAc and the organic layers were combined and concentrated. To the residue, 4 L of toluene was added and the mixture was concentrated and cooled with ice bath. Filtration, washing with 1 L of toluene and drying provided 372 g of compound P-5 (56% yield) as a solid. 1H NMR(300 MHz,

CDCI3) δ 7.78 (d, J = 5.7 Hz, 1 H), 7.54-7.46 (m, 2H), 7.40-7.26 (m, 3H), 6.48 (d, J = 5.7 Hz, 1 H), 5.6 (brs, 1 H), 5.31 (s, 2H).

e) Synthesis of 1-(2,3-dihydroxypropyl)-4-oxo-3-[(phenylmethyl)oxy]-1 ,4-dihydro-2- pyridinecarboxylic acid (compound P-6). A mixture of 509 g of compound P-5 (1.0 eq.) and

407 g of 3-amino-propane-1 ,2-diol(2.5 eq.) in 1.53 L of EtOH was stirred at 65 0C for 1 h and at 80 0C for 6 h. After addition of 18.8 g of 3-Amino-propane-1 ,2-diol(0.1 eq.) in 200 ml. of EtOH, the mixture was stirred at 80 0C for 1 h. After addition of 18.8 g of 3-amino- propane-1 ,2-diol (0.1 eq.) in 200 ml. of EtOH, the mixture was stirred at 80 0C for 30 min. After cooling and addition of 509 ml. of H2O, the mixture was concentrated. To the residue,

2.54 L of H2O and 2.54 L of AcOEt were added. After separation, the aqueous layer was washed with 1.02 L of EtOAc. To the aqueous layer, 2.03 L of 12% sulfuric acid was added at less than 12 0C to give crystal of compound P-6. Filtration, washing with 1.53 L of cold H2O and drying provided 576 g of compound P-6 (83% yield) as a solid. 1H NMR(300 MHz, DMSO-de) δ 7.67 (d, J = 7.5 Hz, 1 H), 7.5-7.2 (m, 5H), 6.40 (d, J = 7.5 Hz, 1 H), 5.07

(s, 2H), 4.2-4.0 (m, 1 H), 3.9-3.6 (m, 2H), 3.38 (dd, J = 4.2, 10.8 Hz, 1 H), 3.27 (dd, J = 6.0, 10.8 Hz, 1 H).

f) Synthesis of methyl 1-(2,3-dihydroxypropyl)-4-oxo-3-[(phenylmethyl)oxy]-1 ,4-dihydro-2- pyridinecarboxylate (compound P-7). To a slurry of 576 g of compound P-6 (1.0 eq.: 5.8% of H2O was contained) in 2.88 L of NMP were added 431 g of NaHCO3(3.0 eq.) and 160 ml. of methyl iodide(1.5 eq.) and the mixture was stirred at room temperature for 4 h. After cooling to 5 0C, 1.71 L of 2N HCI and 1.15 L of 20% NaClaq were added to the mixture at less than 10 0C to give crystal of compound 7. Filtration, washing with 1.73 L of H2O and drying provided 507 g of compound P-7 (89% yield) as a solid. 1H NMR(300 MHz, DMSO- cfe) δ 7.59 (d, J = 7.5 Hz, 1 H), 7.40-7.28 (m, 5H), 6.28 (d, J = 7.5 Hz, 1 H), 5.21 (d, J = 5.4 Hz, 1 H), 5.12 (d, J = 10.8 Hz, 1 H), 5.07 (d, J = 10.8 Hz, 1 H), 4.83 (t, J = 5.7 Hz, 1 H), 3.97 (dd, J = 2.4, 14.1 Hz, 1 H), 3.79 (s, 3H), 3.70 (dd, J = 9.0, 14.4 Hz, 1 H), 3.65-3.50 (m, 1 H), 3.40-3.28 (m, 1 H), 3.26-3.14 (m, 1 H).

g) Synthesis of methyl 1-(2,2-dihydroxyethyl)-4-oxo-3-[(phenylmethyl)oxy]-1 ,4-dihydro-2- pyridinecarboxylate (compound P-8). To a mixture of 507 g of compound P -7 (1.0 eq.) in

5.07 L of MeCN, 5.07 L of H2O and 9.13 g of AcOH(0.1 eq.) was added 390 g of NaIO4(1.2 eq.) and the mixture was stirred at room temperature for 2 h. After addition of 1.52 L of 10% Na2S2OsBq., the mixture was concentrated and cooled to 10 0C. Filtration, washing with H2O and drying provided 386 g of compound P-8 (80% yield) as a solid. 1H NMR(300 MHz, DMSO-d6) δ 7.62 (d, J = 7.5 Hz, 1 H), 7.42-7.30 (m, 5H), 6.33 (d, J = 6.0 Hz, 2H),

6.29 (d, J = 7.5 Hz, 1 H), 5.08 (s, 2H), 4.95-4.85 (m, 1 H), 3.80 (s, 3H), 3.74 (d, J = 5.1 Hz, 2H).

h) Synthesis of (3S, 11 aR)-3-methyl-6-[(phenylmethyl)oxy]-2,3, 1 1 ,1 1a- tetrahydro[1 ,3]oxazolo[3,2-a]pyrido[1 ,2-c/]pyrazine-5,7-dione (compound P-9). After dissolution of mixture of 320 g of compound P-8 (1.0 eq.) in 3.20 L of MeOH by heating, the solution was concentrated. To the residue, 1.66 L of MeCN, 5.72 ml. of AcOH(0.1 eq.) and 82.6 g of (S)-2-Amino-propan-1-ol(1.1 eq.) were added and the mixture was heated to 70 0C, stirred at 70 0C for 4 h and concentrated. To the residue, 1.67 L of 2-propanol was added and the mixture was concentrated (twice). After cooling of the residue, filtration, washing with 500 ml. of cold 2-propanol and drying provided 167 g of compound P-9 (52% yield) as a solid. 1H NMR(300 MHz, CDCI3) δ 7.61-7.55 (m, 2H), 7.40-7.20 (m, 4H), 6.53 (d, J = 7.2, 1 H), 5.46 (d, J = 10.5 Hz, 1 H), 5.23 (d, J = 10.2 Hz, 1 H), 5.20 (dd, J = 3.9, 9.6 Hz, 1 H), 4.46-4.34 (m, 1 H), 4.31 (dd, J = 6.6, 8.7 Hz, 1 H), 4.14 (dd, J = 3.9, 12.3 Hz, 1 H), 3.79 (dd, J = 9.9, 12.3 Hz, 1 H), 3.62 (dd, J = 6.9, 8.7 Hz, 1 H), 1.38 (d, J = 6.3 Hz, 3H).

i) Synthesis of (3 S, 1 1 aR)-8-bromo-3-methyl-6-[(phenylmethyl)oxy]-2,3, 11 ,11a- tetrahydro[1 ,3]oxazolo[3,2-a]pyrido[1 ,2-c/]pyrazine-5,7-dione (compound P-10). To slurry of 156 g of compound P-9 (1.0 eq.) in 780 ml. of NMP was added 93.6 g of NBS(1.1 eq.) and the mixture was stirred at room temperature for 2.5 h. The reaction mixture was added to 3.12 L of H2O. Filtration, washing with 8.0 L of H2O and drying provided 163 g of compound P-10 (84% yield) as a solid. 1H NMR(300 MHz, DMSO-d6) δ 8.37 (s, 1 H), 7.55- 7.50 (m, 2H), 7.42-7.25 (m, 3H), 5.34 (dd, J = 3.6, 9.9 Hz, 1 H), 5.18 (d, J = 10.8 Hz, 1 H), 5.03 (d, J = 10.5 Hz, 1 H), 4.53 (dd, J = 3.6, 12.0 Hz, 1 H), 4.40-4.20 (m, 2H), 3.99 (dd, J = 9.9, 1 1.7 Hz, 1 H), 3.64 (dd, J = 5.7, 8.1 Hz, 1 H), 1.27 (d, J = 6.3 Hz, 3H). j) Synthesis of (3S,1 1aS)-Λ/-[(2,4-difluorophenyl)methyl]-3-methyl-5,7-dioxo-6- [(phenylmethyl)oxy]-2,3,5,7, 11 ,1 1 a-hexahydro[1 ,3]oxazolo[3,2-a]pyrido[1 ,2-c/]pyrazine-8- carboxamide (compound P-11). Under carbon mono-oxide atmosphere, a mixture of 163 g of compound P-10 (1.0 eq.), 163 mL of /-Pr2NEt(2.5 eq.), 68.4 mL of 2,4- difluorobenzylamine(1.5 eq.) and 22.5 g of Pd(PPh3)4(0.05 eq.) in 816 mL of DMSO was stirred at 90 0C for 7 h. After cooling, removal of precipitate, washing with 50 mL of DMSO and addition of 1 1.3 g of Pd(PPh3)4(0.025 eq.), the reaction mixture was stirred at 90 0C for 2 h under carbon mono-oxide atmosphere again. After cooling, removal of precipitate and addition of 2.0 L of AcOEt and 2.0 L of H2O, the organic layer was washed with 1.0 L of 1 N HCIaq. and 1.0 L of H2O (twice) and the aqueous layer was extracted with 1.0 L of AcOEt.

The organic layers were combined and concentrated. Silica gel column chromatography of the residue provided 184 g of compound P-11 (96% yield) as foam. 1H NMR(300 MHz, CDCI3) δ 10.38 (t, J = 6.3 Hz, 1 H), 8.39 (s, 1 H), 7.75-7.25 (m, 7H), 6.90-6.70 (m, 2H), 5.43 (d, J = 10.2 Hz, 1 H), 5.24 (d, J = 10.2 Hz, 1 H), 5.19 (dd, J = 3.9, 9.9 Hz, 1 H), 4.63 (d, J = 6.0 Hz, 2H), 4.50-4.25 (m, 3H), 3.86 (dd, J = 9.9, 12.3 Hz, 1 H), 3.66 (dd, J = 6.9, 8.4 Hz,

1 H), 1.39 (d, J = 6.0 Hz, 3H).

k) Synthesis of (3S,1 1aR)-Λ/-[(2,4-difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo- 2,3,5,7, 11 ,11 a-hexahydro[1 ,3]oxazolo[3,2-a]pyrido[1 ,2-c/]pyrazine-8-carboxamide (compound 1a). Under hydrogen atmosphere, a mixture of 184 g of compound P-11 (1.0 eq.) and 36.8 g of 10%Pd-C in 3.31 L of THF and 0.37 L of MeOH was stirred for 3 h. After filtration of precipitate(Pd-C), washing with THF/MeOH(9/1 ) and addition of 36.8 g of 10% Pd-C, the mixture was stirred for 20 min under hydrogen atmosphere. After filtration of precipitate(Pd-C) and washing with THF/MeOH(9/1 ), the filtrate was concentrated. After 200 mL of AcOEt was added to the residue, filtration afforded crude solid of compound 1 a.

The precipitates were combined and extracted with 4.0 L of CHCI3/Me0H(5/1 ). After concentration of the CHCI3/MeOH solution and addition of 250 mL of AcOEt to the residue, filtration afforded crude solid of compound 1a. The crude solids were combined and dissolved in 8.2 L of MeCN/H2O(9/1 ) by heating. After filtration, the filtrate was concentrated. To the residue, 1.5 L of EtOH was added and the mixture was concentrated

(three times). After cooling of the residue, filtration and drying provided 132 g of compound 1a (88% yield) as a solid. 1H NMR(300 MHz, DMSO-d6) δ 11.47 (brs, 1 H), 10.31 (t, J = 6.0 Hz, 1 H), 8.46 (s, 1 H), 7.40 (td, J = 8.6, 6.9 Hz, 1 H), 7.24 (ddd, J = 2.6, 9.4, 10.6, 1 H), 7.11- 7.01 (m, 1 H), 5.39 (dd, J = 4.1 , 10.4 Hz, 1 H), 4.89 (dd, J = 4.2, 12.3 Hz, 1 H), 4.55 (d, J = 6.0 Hz, 2H), 4.40 (dd, J = 6.8, 8.6 Hz, 1 H), 4.36-4.22 (m, 1 H), 4.00 (dd, J = 10.2, 12.3 Hz, 1 H), 3.67 (dd, J = 6.7, 8.6 Hz, 1 H), 1.34 (d, J = 6.3 Hz, 3H).

I) Synthesis of (3S,1 1aR)-Λ/-[(2,4-difluorophenyl)methyl]-6-hydroxy-3-methyl-5,7-dioxo- 2,3,5,7, 11 ,11 a-hexahydro[1 ,3]oxazolo[3,2-a]pyrido[1 ,2-c/]pyrazine-8-carboxamide sodium salt (compound 1 b). After dissolution of 16.0 g of compound 1a (1.0 eq.) in 2.56 L of EtOH and 0.64 L of H2O by heating, followed by filtration, 39 ml. of 1 N NaOHaq.(1.0 eq.) was added to the solution at 75 0C. The solution was gradually cooled to room temperature. Filtration, washing with 80 ml. of EtOH and drying provided 13.5 g of compound 1b (80% yield) as a solid. 1H NMR(300 MHz, DMSO-d6) δ 10.73 (t, J = 6.0 Hz, 1 H), 7.89 (s, 1 H), 7.40-7.30 (m, 1 H), 7.25-7.16 (m, 1 H), 7.07-6.98 (m, 1 H), 5.21 (dd, J = 3.8, 10.0 Hz, 1 H), 4.58 (dd, J = 3.8, 12.1 Hz, 1 H), 4.51 (d, J = 5.4 Hz, 2H), 4.30-4.20 (m, 2H), 3.75 (dd, J = 10.0, 12.1 Hz, 1 H), 3.65-3.55 (m, 1 H), 1.27 (d, J = 6.1 Hz, 3H).

References

  1.  PrEP GSK744 Integrase Administered Monthly Perhaps Quarterly Prevents HIV-Infection in Monkeys. 20th Conference on Retroviruses and Opportunistic Infections. Atlanta, GA March 3–6, 2013.
  2. http://www.natap.org/2013/CROI/croi_38.htm

 

Cited Patent Filing date Publication date Applicant Title
WO2006116764A1 * Apr 28, 2006 Nov 2, 2006 Shionogi & Co Polycyclic carbamoylpyridone derivative having hiv integrase inhibitory activity
US6919351 * Oct 9, 2001 Jul 19, 2005 Merck & Co., Inc. Aza-and polyaza-naphthalenyl-carboxamides useful as HIV integrase inhibitors
WO2012018065A1 * Aug 4, 2011 Feb 9, 2012 Shionogi & Co., Ltd. Process for preparing compound having hiv integrase inhibitory activity
WO2012151361A1 May 3, 2012 Nov 8, 2012 Concert Pharmaceuticals Inc. Carbamoylpyridone derivatives
WO2013038407A1 * Sep 2, 2012 Mar 21, 2013 Mapi Pharma Ltd. Amorphous form of dolutegravir
US8552187 Dec 9, 2009 Oct 8, 2013 Shionogi & Co., Ltd. Processes and intermediates for carbamoylpyridone HIV integrase inhibitors
US8580967 Jul 23, 2009 Nov 12, 2013 Shionogi & Co., Ltd. Methyl 3-(benzyloxy)-1-(2,2-dihydroxyethyl)-4-oxo-1,4-dihydropyridine-2-carboxylate and processes for the preparation thereof
US8624023 Dec 8, 2009 Jan 7, 2014 Shionogi & Co., Ltd. Synthesis of carbamoylpyridone HIV integrase inhibitors and intermediates

Filed under: Phase2 drugs, Uncategorized Tagged: CARBOTEGRAVIR, GSK 744, hiv, phase 2

Vitamin A could prevent the spread of prostate cancer

$
0
0

Originally posted on lyranara.me:

Vitamin A could help treat and prevent the spread of prostate cancer, according to research published today (Monday, April 15th) in  Oncogenesis

Scientists funded by Yorkshire Cancer Research at the University of York have discovered that retinoic acid – a chemical made from vitamin A which is supplied in our diet by carrotsgreen vegetables and liver – can turn specific genes within prostate cancerstem cells back on, reducing the ability of the cancer to invade surrounding tissue.

The findings suggest that Vitamin A related compounds could be used to enhance clinical treatments for prostate cancer.

Professor Norman Maitland, Director of the YCR Cancer Research Unit in the Department of Biology at York, said: “Cancer arises from healthy cells going wrong. Certain controls can be turned off which allows the cancer to progress. For example, normal cells gain the ability to grow and invade the surrounding tissues.

View original 179 more words


Filed under: Uncategorized

How tumors become resistant to drugs, and how process can be reversed to inhibit cancer growth

$
0
0

Originally posted on lyranara.me:

Researchers at the Hebrew University of Jerusalem’s Faculty of Medicine have discovered a process whereby tumor cells become resistant to specific drugs, a finding that could significantly influence how anti-cancer drugs are administered and the development of a means for reversing the proliferation of malignant tumor growth. 

Researchers at the Hebrew University of Jerusalem’s Faculty of Medicine have discovered a process whereby tumor cells become resistant to specific drugs, a finding that could significantly influence how anti-cancer drugs are administered and the development of a means for reversing the proliferation of malignant tumor growth.

Cancer has become one of the major challenges of biomedical research in the past decades, and is one of the leading causes of illness and death all over the world. While many drugs have been developed against cancer, doctors do not know in advance of treatment whether a patient might benefit from a particular drug. Thus…

View original 414 more words


Filed under: Uncategorized

NERATINIB, HKI 272, ..Puma presents positive results from phase II trial of its investigational drug PB272

$
0
0
File:Neratinib skeletal.svg
NERATINIB
(2E)-N-[4-[[3-chloro-4-[(pyridin-2-yl)methoxy]phenyl]amino]-3-cyano-7-ethoxyquinolin-6-yl]-4-(dimethylamino)but-2-enamide
 [(2E)-N-[4-[[3-chloro-4- [(pyridin-2-yl)methoxy]phenyl]amino]-3-cyano-7-ethoxyquinolin-6-yl]-4- (dimethylamino)but-2-enamide].
 (E)-N- {4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6- quinolinyl} -4-(dimethylamino)-2-butenamide 
FOR METASTATIC BREAST CANCER.PHASE 3
CAS 698387-09-6,
PFIZER …….INNOVATOR
HKI-272, HKI 272, Neratinib(HKI-272), Neratinib, HKI-272, 698387-09-6, HKI272, HKI 272, HKI-272,
HKI-272  
PB-272  
PF-0528767  
WAY-179272  
WAY-179272-B (maleate) 
Molecular Formula: C30H29ClN6O3
Molecular Weight: 557.04266
Puma Biotechnology, a development stage biopharmaceutical company, announced the presentation of positive results from the phase II clinical trial of Puma’s investigational drug PB272 (neratinib) for the neoadjuvant treatment of breast cancer(I-SPY 2 TRIAL) in an oral presentation at the American Association for Cancer Research (AACR) Annual Meeting 2014 in San Diego, California.
READ AT
Neratinib – малая молекула класса 6,7-дизамещенных-4-anilinoquinoline-3-карбонитрила -
ингибитор тирозинкиназы HER-2 с потенциальной противоопухолевой активностью.
Neratinib связывается с рецептором HER-2 необратимо, снижая аутофосфорилирование в клетках,
и направляя остаток цистеина в АТФ-связывающего кармана рецептора.
Обработка раковых клеток с этим агентом приводит к торможению передачи сигнала клеточного цикла и
в конечном счете уменьшает клеточную пролиферацию.
Neratinib ингибирует рецептор EGFR киназы и распространение EGFR-зависимых клеток.
Neratinib - small molecule 6,7-disubstituted class of 4-anilinoquinoline-3-carbonitrile -
inhibitor of the HER-2 tyrosine kinase with potential antitumor activity.
Neratinib binds to the receptor HER-2 irreversible, reducing autophosphorylation in cells
and directing the cysteine ​​residue in the ATP-binding pocket of the receptor.
Treatment of cancer cells with this agent leads to inhibition of signal transduction and cell cycle  ultimately reducescell proliferation.
Neratinib inhibit EGFR kinase receptor and distribution of EGFR-dependent cells.

Neratinib (HKI-272) is a tyrosine kinase inhibitor[1][2] under investigation for the treatment breast cancer[3] and other solid tumours.

It is in development for the treatment of early- and late-stage HER2-positive breast cancer.[4]

Like lapatinib and afatinib, it is a dual inhibitor of the human epidermal growth factor receptor 2 (Her2) and epidermal growth factor receptor (EGFR) kinases.[5]

Neratinib is a signal transduction pathway inhibitor and an irreversible inhibitor of HER-2 in early clinical trials for the treatment of advanced solid tumors in combination with paclitaxel. The company had also been developing the drug candidate for the treatment of non-small cell lung cancer (NSCLC); however, no recent development has been reported for the indication. In 2011, Pfizer discontinued development of the compound as monotherapy for the treatment of ErbB-2-positive breast cancer. A phase III clinical trial had been under way. Dana-Farber Cancer Institute is studying the compound for the treatment of patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer and brain metastases. Puma Biotechnology is conducting phase III trials for use as third-line treatment of HER2-positive metastatic breast cancer and phase II trials for the treatment of patients with HER2 activating mutations in Non-Small Cell Lung Cancer (as monotherapy or in combination with temsirolimus) as well as other solid tumors.

The drug candidate is a synthetic compound developed based on the chemical structure of EKB-569, an inhibitor of the epidermal growth factor receptor (EGFR) currently under clinical evaluation for the treatment of EGFR-positive tumors. In previous trials, neratinib inhibited kinase activity of HER-2 and EGFR by 50% while showing no effects on several serine-threonine kinases, and also inhibited the proliferation of two HER-2-positive breast cancer cell lines and a mouse fibroblast cell line transfected with the HER-2 oncogene. 

In 2011, the compound was licensed to Puma by Pfizer for global development and commercialization.

HKI-272 (neratinib) has been described for the treatment of neoplasms [US Patent 6,288,082]. Neratinib is a potent irreversible pan erbB inhibitor. Neratinib is an orally available small molecule that inhibits erbB-1 , erbB-2 and erbB-4 at the intracellular tyrosine kinase domains, a mechanism of action that is different from trastuzumab. Neratinib reduces erbB-1 and erbB-2 autophosphorylation, downstream signaling, and the growth of erbB-1 and erbB-2 dependent cell lines.

Preclinical data suggest that neratinib will have antitumor activity in erbB-1 – and/or erbB 2-expressing carcinoma cell lines, with cellular IC50 <100 nM [Rabindran SK, et al. Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase. Cancer Research. 2004;64(1 1 ):3958-65].

Neratanib is being developed by Puma Biotechnology. It will be included in the forthcoming I-SPY2 breast cancer trial.[6]

neratinib refers to HKI-272, which has the following core structure:

Figure imgf000005_0001

in its free base form. Optionally, a pharmaceutically acceptable salt or hydrate thereof may be used. The core structure represented above is a particular HKI-272 compound, called HKI-272 or neratinib, which has the chemical name [(2E)-N-[4-[[3-chloro-4- [(pyridin-2-yl)methoxy]phenyl]amino]-3-cyano-7-ethoxyquinolin-6-yl]-4- (dimethylamino)but-2-enamide]. Although currently less preferred, another HKI-272 compound may be used in the place of neratinib. “A HKI-272 compound” refers, in one embodiment, to a compound derived from the core structure of neratinib shown above

The preparation of HKI-272 compounds, of which neratinib is a species, are described in detail in US Patent Application Publication No. 2005/0059678, which is hereby incorporated by reference. See, also, US Patent Nos. 6,288,082, US Patent No. 6,002,008, US Patent No. 6,297,258 and US Patent Application Publication No. 2007/0104721 , which are hereby incorporated by reference. The methods described in these documents can also be used to prepare neratinib and/or the other HKI-272 and substituted 3-quinoline compounds used herein and are hereby incorporated by reference. In addition to the methods described in these documents, International Patent Publication Nos. WO-96/33978 and WO-96/33980, which are hereby incorporated by reference, describe methods that are useful for the preparation of these HKI-272 compounds. Although these methods describe the preparation of certain quinazolines, they are also applicable to the preparation of correspondingly substituted 3- cyanoquinolines and are hereby incorporated by reference.

The term “treating” or “treatment” refers to the administration of the neratinib to a subject to prevent or delay, to alleviate, or to arrest or inhibit development of the symptoms or conditions associated with neoplasms

(E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4- (dimethylamino)-2-butenamide is an irreversible inhibitor to Her-2 (also known as ErbB-2 or neu) kinase, a member of the epidermal growth factor receptor (EGFR) family. EGFR family members have been implicated in tumorigenesis and associated with poor prognosis in tumor types in humans. The structure of the (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano- 7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide in the form of a free base is shown below:

 

Figure imgf000009_0001

The compound (E)-N-{4-[3-chloro-4 J-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}- 4-(dimethylamino)-2-butenamide in the form of a free base is described in U.S. Patent No. 6,288,082. The compound is classified, based on the Biopharmaceutical Classification System, as a BCS Class IV compound (low water solubility and low permeability). The free base has low solubility in water, with a water solubility of about 1 μg/ml_ at about pH 7. The water solubility increases with decreasing pH as the compound becomes ionized. This compound is water soluble at gastrointestinal pH, and dissolution is not rate limiting.

Research on Chemical Intermediates, 2012, 09(22),6168
10.1007/s11164-012-0822-4
The Wittig–Horner reaction for the synthesis of neratinib

…………………

U.S. Patent No. 6,288,082

http://www.google.co.in/patents/US6288082

…………

WO2010048477A2

http://www.google.com/patents/WO2010048477A2?cl=en

U.S. Pat. No. 7,126,025 discloses certain novel 4-amino-2-butenoyl chlorides, processes for their preparation and their use as intermediates in the synthesis of pharmaceutically active protein kinase inhibitors, including but not limited to for example HKI-272 and EKB-569.

The sequence illustrated below and summarized in Scheme 1 describes one existing process for preparing HKI-272, (E)-Λ/-(4-(3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)-3- cyano-7-ethoxyquinolin-6-yl)-4-(dimethylamino)but-2-enamide in the form of the maleate salt, also known as Neratinib™.

 

Figure imgf000004_0001

1 95 eq (COCI)2, cat DMF

O

^

Step 5 OH 16 h HCI

 

Figure imgf000004_0002

Scheme 1

Figure imgf000004_0003

Scheme 2

Figure imgf000007_0001

Scheme 3. Formation of acid chloride with SOCI2 in DMAc and coupling with a substituted aniline.

SOCl2

/ Nv^-^’C02H HCI DMAc HCI

 

Figure imgf000010_0001

Scheme 4. Formation of the MW 638 impurity.

Figure imgf000010_0002

Example 4: Process 3

4-Dimethylaminocrotonoyl chloride hydrochloride and its coupling with 6-amino- 4-(3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)-7-ethoxyquinoline-3-carbonitrile (procedure with thionyl chloride and DMAc).

A suspension of 4-dimethylaminocrotonic acid (17.0 g, 97.5 mmol) in DMAc (170 ml_) was cooled to -15 0C under nitrogen atmosphere. Neat thionyl chloride (12.8 g, 7.83 mmol) was added to the slurry at a rate to maintain the temperature in the reactor in the range of -18 to -14 0C (moderate exotherm). The reaction mixture was held at -17 to -15 0C for 4 hrs. A solution of the aminoquinoline (36.2 g, 81.3 mmol) in DMAc (440 ml_) was added to the reactor maintaining the temperature in the -14 to -19 0C range. The resulting mixture was held for 18 hr at approximately -15 0C. At this point HPLC analysis showed residual aniline level at 2.5%. The thick suspension of the hydrochloride salt of the coupled product was quenched with water (200 ml_) maintaining the batch temperature between -5 and -16 0C. The pH of the resulting clear solution was adjusted to 1 1 with a 13% aqueous solution of NaOH (approx. 210 ml_ of the solution was added). The suspension was further diluted with water (350 ml_) and the solids were filtered on a polypropylene cloth filter. The cake was washed with water until neutral pH of the washes and dried first in the nitrogen flow on the filter and then on a tray in vacuum at 45 to 50 0C to afford crude (.=)-/\/-(4-(3-chloro-4-(pyridin-2-ylmethoxy)phenylamino)-3-cyano-7- ethoxyquinolin-6-yl)-4-(dimethylamino)but-2-enamide (42.0 g, 91 %) as a bright-yellow crystalline solid.

 

 

………………..

WO2004066919A2

http://www.google.com/patents/WO2004066919A2?cl=en

Reaction Scheme Example 1 :

SCHEME 1

 

Figure imgf000021_0001

(“)

 

Figure imgf000022_0001

6-(4-N,N-dimethylarninocrotonyt)amido- 4-(4-benzyioxy-3-chloro)arniπo-3-cyano- 7-ethoxyquiπoline, WAY-177820 C31H3[1CIN5θ3 MW 556.07

A suspension of 4-N,N-dimethylaminocrotonic acid hydrochloride in acetonitrile and a catalytic amount of DMF is cooled to 0-10° C. Oxalyl chloride (0.95 eq) is added dropwise and the mixture warmed to 25-30° C and stirred until the chlorinating agent is completely consumed. The light yellow solution is checked for complete consumption of oxalyl chloride by HPLC then cooled to 0-10° C. A cooled solution (0-10° C) of 4-[4-benzyloxy-3-chloro]amino-6-amino-3-cyano-7- ethoxyquinoline in NMP is added dropwise and the mixture is stirred until < 2% of the starting aniline remains. The mixture is added to saturated aqueous sodium bicarbonate, the yellow precipitates are filtered and washed with water. The wet solids are heated to reflux in acetonitrile and clarified hot to remove insolubles. The solution is cooled, the precipitated product filtered and washed with cold acetonitrile. The product is dried (40-50° C, 10 mm Hg, 24 hours) to obtain the final product. Reaction Scheme Example 2:

Figure imgf000023_0001

A solution of 4-N,N-dimethylaminocrotonic acid hydrochloride in tetrahydrofuran (THF) and a catalytic amount of dimethyiformamide (DMF) is cooled to 0-5s C. Oxalyl chloride (0.95 eq) is added dropwise and the mixture warmed to 25-302C and stirred until the chlorinating agent is completely consumed. The orange solution is checked for complete consumption of oxalyl chloride by high- pressure liquid chromatography (HPLC) then cooled to 0-52 C. A solution of 4-[4-(2- pyridylmethoxy)-3-chloro]amino-6-amino-3-cyano-7-ethoxyquinoline is added dropwise and the mixture is stirred until < 0.5% of the starting aniline remains. The reaction is quenched with water and the mixture warmed to 40s C. Aqueous sodium hydroxide is added to bring the pH to 10-11. The resulting precipitates are filtered hot and washed with water. The wet solids are heated to reflux (70-759 C) in acetonitrile:THF (1 :5:1) and the solution cooled slowly to room temperature. The product is filtered and washed with acetonitrile.THF. The product is dried (50e C, 10 mm Hg, 24 hours) to 80-85% yield.

Reaction Scheme Example 3:

Figure imgf000024_0001

4-Dirnethy!amino-but-2-enoic acid |4-(3-chloro-4-fluoro-phenylamino)-3-cvano-7- ethoxy-quinolin-6-vHamide

A. 4-(dimethylamino)-2-butenoyl chloride hydrochloride

A 1 L multi-neck flask equipped with agitator, thermometer, addition funnel, and nitrogen protection is charged with acetonitrile (0.67 kg, 0.85 L) followed by adding dimethylformamide (0.00086 kg, 0.91 mL, d=0.944 g/mL). At ambient temperature, is added 4-dimethylaminocrotonic acid hydrochloride (0.0709 kg) and the mixture stirred until homogeneous. Cool the reaction mixture to (0-10° C) and add oxalyl chloride (0.0473 kg, 0.0325 L, d = 1.45 g/mL) dropwise over (20 minutes) at (0-10° C) followed by a rinse with acetonitrile (0.02 kg, 0.03 L). The temperature (0-10°C) is maintained for about (20 minutes). The temperature of the reaction mixture is adjusted to (22-26° C) over (20 minutes) and maintained over (2 hours). The temperature of reaction mixture is adjusted to (40-45° C) and held for about (5 minutes). Cool the light suspension to about (20-25° C) and check for reaction completion by high-pressure liquid chromatography (HPLC). The reaction is complete when there is < 15 % of the starting material (4-dimethylaminocrotonic acid hydrochloride) present and/or < 2 % of oxalyl chloride (detected as the dimethyl oxalate).

B. 4-Dimethy!amino-but-2-enoic acid |4-(3-chloro-4-fluoro-phenylamino)-3-cyano-7- ethoxy-quinolin-6-yll-amide (crude)

A 3 L multi-neck flask equipped with agitator, thermometer, dip tube, and nitrogen protection is charged N-methyl pyrrolidinone (0.77 kg, 0.75 L, d=1.033 g/mL). At ambient temperature is added 4-[3-chloro-4-fluorophenyl]amino-6-amino-3-cyano-7- ethoxy quinoline (0.0748 kg). The reaction mixture is heated to 40-45° C and maintained for about (15 minutes). The reaction mixture is cooled to (0-10° C) and the light suspension of 4-(dimethylamino)-2-butenoyl chloride hydrochloride in CH3CN added via dip tube and positive nitrogen pressure, over (30-45 minutes) while maintaining the temperature (0-10° C) for at least (2 hours). Reaction completion is monitored by HPLC. The reaction is complete when there is < 2 % of the starting material (4-[3-chloro-4-fluorophenyl]amino-6-amino-3-cyano-7-ethoxy quinoline) present. To a 12 L multi-neck flask equipped with agitator, thermometer, dip tube, and nitrogen protection is charged with water (2.61 kg, 2.61 L) and sodium bicarbonate (0.209 kg) with stirring until a solution is obtained followed by cooling to (20-24° C) to which is transferred the reaction mixture above which contains < 2 % of the starting material (4-[3-chloro-4-fluorophenyl]amino-6-amino-3-cyano-7-ethoxy quinoline), via dip tube and positive nitrogen pressure, to the 12 L flask over about (45-60 minutes) while maintaining the temperature at (20-24° C). The temperature is maintained at (20-24° C) for at least (1 hour). Filter the reaction mixture on a Buchner funnel, rinse with water (3 x 0.40 kg, 3 x 0.40 L), and maintain suction until dripping stops. Dry the product in a vacuum oven at about (50° C) and about (10 mm Hg) for about (28-30 hours). The yield is 78.5 g (86%) at 79.7% strength and 12.3% total impurities.

4-Dimethylamino-but-2-enoic acid r4-(3-chloro-4-fluoro-phenylamino -3-cyano-7- ethoxy-quinolin-6-vn-amide (purified small scale)

First crop: A 6 L multi-neck flask equipped with agitator, condenser, temperature probe, and nitrogen protection is charged with acetonitrile (3.14 kg, 4.00 L) followed by adding 4-dimethylamino-but-2-enoic acid [4-(3-chloro-4-fluoro-phenylamino)-3-cyano-7- ethoxy-quinolin-6-yl]-amide (0.16 kg, 0.167 moles). Heat the mixture to (75-80° C) and hold it for (1 hour). Cool the mixture to (70-75° C) and filter on a pad of diatomaceous earth to remove inorganic salts. Wash the pad with acetonitrile (2 x 0.24 kg, 2x 0.30 L), preheated to (70-75° C). Concentrate the filtrate at (20-30 mm Hg) and a maximum temperature of (40-45° C) to a volume of ( 1.2 L). To the concentrate (slurry) add prefiltered tetrahydrofuran (0.53 kg, 0.60 L). Heat to (65-70° C) to obtain a complete solution. Cool the mixture to (40-45° C) over (0.3 hours). Add seeds and continue cooling to (20-25° C) over (1 hour). Hold at (20-25° C) for a minimum of (18 hours). Collect the solid on a Buchner funnel and wash the collected solid with a prefiltered and precooled at (0-5° C) mixture of acetonitrile/tetrahydrofuran (2/1 by volume) (2 x .06 kg, 2 x 0.08 L). Dry the product in a vacuum oven at (50° C) and (10 mm Hg) for (48 hours) to a loss on drying (LOD) of less than (0.5 %). All washes and concentrates (mother liquors) are saved for further purification.

Second crop:

A 3 L multi-neck flask equipped with agitator, temperature probe, nitrogen protection, and charge with the mother liquors and washes from above. Concentrate by distillation at (20-30 mm Hg) and a maximum temperature of (40-45° C) to a volume of (0.50 L). Collect the solid on a Buchner funnel and wash the solid with prefiltered acetonitrile (0.04 kg, 0.05 L). Dry the solid product in a vacuum oven at (50° C) and (10 mm Hg) for (18 hours). A 1 L multi-neck flask equipped with agitator, condenser, temperature probe, nitrogen protection and charge with prefiltered acetonitrile (0.47 kg, 0.60 L), and the collected solid is heated as a suspension to (70-75° C) over (0.5 hours). Add prefiltered tetrahydrofuran (0.03 kg, 0.03 L) to the suspension while maintaining the temperature at (70-75° C). Cool the solution to (40-45° C) and add seed crystals. Continue cooling to (20-25° C) over (1 hour) and hold for (2 hours). Collect the resulting solid on a Buchner funnel and wash the collected solid with a prefiltered and precooled to (5° C) mixture of acetonitrile/tetrahydrofuran (20/1 by volume) (2 x 0.02 kg, 2 x 0.03 L). Dry the collected solid in a vacuum oven at (50° C) and (10 mm Hg) for (24 hours) to an LOD of less than (0.5 %). The combined yield is 27.5 g + 30.5 g (73%) in 96.2-98.4% strength and 1.5-1.7% total impurities by high pressure liquid chromatography (HPLC).

4-Dimethylamino-but-2-enoic acid f4-(3-chloro-4-fluoro-phenylamino)-3-cvano-7- ethoxy-quinolin-6-vn-amide (purified larger scale)

Acetonitrile, practical (34.0 kg) and 4-dimethylamino-but-2-enoic acid [4-(3- chloro-4-fluoro-phenylamino)-3-cyano-7-ethoxy-quinolin-6-yl]-amide (2.69 kg crude, 1.53 kg at 100% strength) are charged to a purged (100 L) reactor. Acetonitrile, practical (2.0 kg) is used as rinse for funnel and vessel walls. The brown suspension is heated at 70 to 76° C using a jacket temperature not exceeding 85° C, then held at the latter temperature for a minimum of 45 minutes, not exceeding 60 minutes. The resulting suspension is then filtered on the warm-jacketed (70-76° C) 14″ Aurora filter, while maintaining the batch temperature at 70 to 76° C. The filtrates are collected by pump into a purged (100 L) receiver, while keeping their temperature below 50° C. The diatomaceous earth pad is then washed with warm (70 to 76° C) acetonitrile, practical (3 x 2.5 kg). The filtrates and washes in (100 L) receiver are cooled to 20 to 26° C, then transferred into a stainless steel drum. Acetonitrile, practical (2.0 kg) is used as rinse. After cleaning and purging both vessels, the contents of the stainless steel drum is transferred into the (100 L) receiver. Acetonitrile, practical (2.0 kg) is used as a rinse. The batch is heated at 70 to 76° C without exceeding jacket temperature of 85° C. The batch is filtered by pump through a .0 micron single cartridge filter, while maintaining the contents at 70 to 76° C. Warm (70-76° C) acetonitrile, practical (4.0 kg) is used as rinse for vessel, filters, pump and lines. The filtrate and rinse are collected and maintained below 50° C. The batch is adjusted to 10 to 16° C, then concentrated by vacuum distillation to 28 to 33 L volume: expected distillation temperature 20 to 30° C, distillate volume 32 to 37 L. The suspension is heated to 64 to 70° C without exceeding jacket temperature of 85° C. The resulting solution is cooled to 40 to 46° C, then seeded using 4-dimethylamino-but-2~enoic acid [4-(3-chloro-4-fluoro-phenylamino)-3-cyano- 7-ethoxy-quinolin-6-yl]-amide, purified (0.5 g). The mixture is cooled to 20 to 26° C over 1 hour, then held at the latter temperature for a minimum of 2 hours. The suspension is then cooled at -3 to 3° C over 1 hour, then held for a minimum of 1 hour. The solid product is collected on a 16″ Buchner, then washed with cold (0-5° C) acetonitrile-tetrahydrofuran (20-6 v/v) mixture (2 x 2.5 kg). The wet collected solid is recrystallized once more from acetonitrile-tetrahydrofuran (20-6 v/v) to desired purity. The material is dried in a vacuum oven first at 35 to 45° C (target 40° C) for 4 hours, liquid ring pump, then 45 to 55° C (target 50° C) for 4 hours. After high vacuum is applied at the latter temperature, until LOD <0.5% (90° C, 2 hours, full vacuum) and each of acetonitrile, tetrahydrofuran and 1-methyl-2-pyrrolidinone are below 0.2%. The purified drug substance is milled (Comil), then blended. The yield is 1.10 kg (70.1 %, corrected for starting material). The strength of the material is 98.3% and a total impurities of 1.27%.

 

………………….

N OXIDE

http://www.google.com/patents/US20130225594

EXAMPLE 19 Formula 57-Compound 19a

Figure US20130225594A1-20130829-C00247

 

19a: (E)-4-((4-((3-Chloro-4-(pyridin-2-ylmethoxy)phenyl)amino)-3-cyano-7-ethoxyquinolin-6-yl)amino)-N,N-dimethyl-4-oxobut-2-en-1-amine oxide

To a solution of compound A (200 mg, 0.36 mmol, 1.0 eq) in CH2Cl(20 mL) was added m-CPBA (74 mg, 0.43 mmol, 1.2 eq) and the resulting mixture was stirred at room temperature for 4 h. A saturated aqueous solution of NaHCO(20 mL) was then added and the organic layer was separated, dried over Na2SOand concentrated under reduced pressure. The residue was purified by preparative TLC (CH2Cl2/MeOH, 10/1, v/v) to give (E)-4-((4-((3-chloro-4-(pyridin-2-ylmethoxy)phenyl)amino)-3-cyano-7-ethoxyquinolin-6-yl)amino)-N,N-dimethyl-4-oxobut-2-en-1-amine oxide (20 mg, 10%) as a yellow solid.

LC-MS (Agilent): R3.03 min; m/z calculated for C30H29ClN6O[M+H]573.19. found 573.2.

1H NMR: (400 MHz, CD3OD) δ (ppm): 8.98 (s, 1H), 8.57 (m, 1H), 8.39 (s, 1H), 7.92 (td, J=7.2, 1.6 Hz, 1H), 7.72 (d, J=8.0 Hz, 1H), 7.39 (m, 1H), 7.36 (d, J=2.4 Hz, 1H), 7.28 (s, 1H), 7.24-7.13 (m, 3H), 6.74 (d, J=15.6 Hz, 1H), 5.29 (s, 2H), 4.32 (q, J=6.8 Hz, 2H), 4.20 (d, J=7.2 Hz, 2H), 3.28 (s, 6H), 1.57 (t, J=6.8 Hz, 3H).

……………

http://www.google.fm/patents/EP1883631A1?cl=en

Scheme 2 and Scheme 3. Scheme 2

 

Figure imgf000028_0001

e-Acelamlno^chloro-S-cyano- 7-ethoxy quinoliπe C,4Hi2CIN2O2 +

MW 289.72

 

Figure imgf000028_0002

25 °C, 5 h 3-Chloro-4-(3-fluorobenzyl)oxy- anillne

C13Hi1CIFNO

Figure imgf000028_0003

MW 251.69

2 h

Figure imgf000028_0004

free base

 

Figure imgf000028_0005

Scheme 3

 

Figure imgf000029_0001

6-Acetamlno-4-chloro-3-cyanc~ 7-elhoxy qulnollne C,4H12CIN2O2 +

MW 28972

 

Figure imgf000029_0002

3-Chlorc-4-fluoronitrobenzene 2-Pyπdyl carblnol 3-Chloro-4-(3-pyndinylmethoxy) 3-Chloro-4-(2-pyrtdlnylmethewy)- C6H3CIFNO2 C6H7NO nitrobenzene anlllne

MW 17555 MW 109 13 C12H9CIN2O3 C12H11CIN2O d=1 1131 g/ml MW 26467 (EM 264) MW 23469

1 h

Figure imgf000029_0003

(HCI salt)

free base

maleate

 

Figure imgf000029_0004

Example 1

[0078] Synthesis of 3-chloro-4-(2-pyridylmethoxy)nitrobenzene

 

Figure imgf000030_0001

[0079] 2-pyridinyl carbinol (31.08 g, 1.05 eq) was dissolved in ACN (750 mL) and KOH flakes (85%) were added (20.6 g, 1.25 eq.). The resulting suspension was warmed to 35 °C. A solution of the 3-chloro-4-fluoronitrobenzene (50.0 g, 0.285 mol) in ACN (250 mL) was added at 35-40 °C. The mixture was held for 14 hours. The mixture was then cooled back to 20-25 °C, quenched with H2O (IL) and the resulting slurry filtered and washed with H2O (3 x 100 mL). The resulting product was isolated as a tan solid in 93% yield with a greater than 99.5% purity as determined by HPLC area. Example Ia

[0080] To accomplish the analogous synthesis of 3-chloro-4-(3-fluorobenzyloxy) nitrobenzene, 3-fluorobenzyl alcohol (0.30 kg, 2.39 mole, 1.05 eq) was dissolved in ACN (6.0 L) and to it was added potassium hydroxide flakes (85%) (0.16 kg, 1.25 eq). The resulting suspension was warmed to 35 0C. A solution of the 3-chloro-4-fluoronitrobenzene (0.40 kg, 2.28 mol) in ACN (2.0 L) was added at 35-40 °C. The mixture was held for 18 hours. The mixture was then cooled back to 20-25 °C, quenched with water (8 L) and the resulting slurry filtered and washed with water (2 x 0.40 L). The resulting product was dried at 45 °C, under 10 mm Hg pressure, for 25 hours to give 0.59 kg (92% yield). Example Ib

[0081] To prepare 4-(benzyloxy)3-chloronitrobenzene, benzyl alcohol (0.34 kg, 3.14 mole, 1.10 eq) was dissolved in acetonitrile (1.70 L) and to it was added potassium hydroxide flakes (85%) (0.24 kg, 1.50 eq). The resulting suspension was warmed to 25 0C. A solution of the 3- chloro-4-fluoronitrobenzene (0.50 kg, 2.85 mol, 1.0 eq) in acetonitrile (0.75 L) was added keeping the pot temperature < 45 0C. The mixture was held for 14 h. The mixture was then cooled back to 0-15 0C, quenched with water (2.5 L) and the resulting slurry was filtered and washed with water (2 x 0.50 L). The resulting product was dried at 50 0C, under 10 mm Hg pressure, for 24 hours to give 0.73 kg (97% yield). [0082] Experimental results for the reaction of Example 1 with different bases and solvents are shown in Table 1. The last three entries on Table 1 are large scale runs in which a 5% excess of pyridyl carbinol was used. Table 1 – Preparation of Nitroaryl Intermediate

 

Figure imgf000031_0001

NA = not applicable

RT = room temperature (20-25 °C)

Example 2

[0083] Preparation of 3-chloro-4-(2-pyridyhnethoxy)aniline from the nitrobenzene product of

Example 1 was accomplished with catalytic hydrogenation using platinum on carbon.

Figure imgf000032_0001

[0084] A typical hydrogenation was done using 6 volumes of THF, 2% by weight of 5%Pt/C (50% water wet), at 25 psi and at 25-30 0C for approximately 4-6 hours. The reaction is slightly exothermic and the temperature will rise to about 30-35 °C. Cooling is necessary to maintain the temperature below 30 0C.

[0085] As a specific example, a mixture of 3-chloro-4-(2-pyridylmethoxy)nitrobenzene (0.15 kg, 0.57 mole) and 2% (w/w) of 5% Pt/C (6.0 g) in tetrahydrofuran (0.90 L) was hydrogenated at 25 psi for at least 5 hours. The mixture was filtered through a celite pad and washed with tetrahydrofuran (0.60 L). The filtrate was distilled to a volume of about 0.75 L and ethanol (1.12 L) was added. Distillation was continued to a volume of about 0.75 L and ethanol (2.85 L) was added. The mixture may be used “as is” in the step of Example 3 below. Example 2 a

[0086] To accomplish an analogous synthesis of 3-chloro-4-(3-fluorobenzyloxy)aniline, zinc (0.464 kg) was added to a mixture of 3-chloro-4-(3-fluorobenzyloxy)nitrobenzene (0.40 kg, 1.42 mole) and ethanol (4.0 L). The mixture was heated to 40-50 °C. A solution of ammonium chloride (0.152 kg) in water (0.80 L) was added over 0.5 hour keeping the pot temperature at 40-50 °C. The mixture was stirred for 2 hours, filtered and washed with hot (40-50 °C) ethanol (2 x 0.40 L). The filtrate was distilled to a volume of about 0.80 L and 2- methyltetrahydrofuran (2.0 L) was added to dissolve the product. Water (0.80 L) and saturated brine (0.40 L) were added and the layers separated. The organic layer was washed with water (0.60 L), and distilled to a volume of about 0.40 L. Ethanol (2.0 L) was added and distillation continued to a volume of 1.2 L. Example 2b

[0087] To prepare 4-(benzyloxy)-3-chloroaniline, a mixture of 4-(benzyloxy)-3- chloronitrobenzene (0.325 kg, 1.23 mole, 1.0 eq) and 1% (w/w) of 5% Pt/C (3.25 g) in isopropanol (3.25 L) was hydrogenated at 25 psi for a minimum of 4.5 h. The mixture was filtered through a celite pad and washed with isopropanol (2.0 L). The filtrates were used as is in the next step.

[0088] Performing the hydrogenation in isopropyl alcohol (PA), methanol (MeOH), or ethanol

(EtOH) may result in the product being contaminated with late eluting impurity that partially precipitates out on standing in solution. It was found that performing the hydrogenation in a solvent where both the product and starting material are soluble, such as tetrahydrofuran

(THF), resulted in greater product purity and required much less solvent. Thus, THF is a preferred solvent for this step. Experimental results showing the effect of different reaction conditions are shown in Table 2. For the larger scale runs, the first aniline intermediate was not isolated (“NI”) before proceeding with the next step.

Table 2 – Hydrogenation to Form First Aniline Intermediate

 

Figure imgf000033_0001

* Solid impurities noted after reaction completion. ** percent by weight of starting material. Example 3

[0090] Following hydrogenation to form the first aniline intermediate, acid catalyzed coupling was performed to prepare 4~[3-chloro-4-(2-pyridylmethoxy)anilino]-3-cyano-7-ethoxy-6-N- acetylaminoquinoline, as shown below:

 

Figure imgf000034_0001

[0091] To perform the coupling reaction, the two reactants were heated together in alcohol at 65-78°C over 4-6 hours, yielding the product. The reaction begins as an amber slurry and thickens to a lighter beige slurry as it approaches completion. Upon scaling up from 75 g to 350 g, it proved necessary to add a catalytic amount (0.025 eq.) of methanesulfonic acid to initiate the reaction. As a specific example, 4-chloro-3-cyano-7-ethoxy-6-N- acetylaminoquinoline (0.141 kg, 0.49 mole) was added to the mixture of Example 2, followed by ethanol (0.037 L) to give a suspension. A catalytic amount of methanesulfonic acid (1.17 g) was added at 20-25 C. The resulting slurry was heated to 70-75 C and held for a minimum of 4 hours. Thickening of the slurry was evident after 1.5 hours. Following reaction completion, the mixture was cooled to room temperature and may be used “as is” in the telescoped reaction of Example 4 below. Example 3 a

[0092] To prepare 6-acetamido-4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7- ethoxyquinoline, ethanol (4.80 L) was added to the aniline solution followed by 4-chloro-3- cyano-7-ethoxy-6-N-acetylaminoquinoline (0.350 kg, 1.11 mole). A catalytic amount of methanesulfonic acid (2.0 ml) was added at 20-250C. The resulting suspension was heated to 70-750C and held for a minimum of 2 h. Thickening of the slurry was evident during this holding period. Following reaction completion, the mixture was used as is in the following telescoped reaction. Example 3 b

[0093] To prepare 6-acetainido-4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxy-quinoline, isopropanol (6.75 L) was added to the aniline solution followed by 4-chloro-3-cyano-7-ethoxy- 6-N-acetylaminoquinoline (0.277 kg, 0.96 mole, 0.78 eq). A catalytic amount of methane sulfonic acid (3.50 ml) was added at 20-250C. The resulting suspension was heated to 80-850C and held for a minimum of 10 hr. Thickening of the slurry was evident during this holding period. Following reaction completion, the mixture was cooled to 25-35 0C, filtered and the cake washed with isopropanol (3 x 0.25 L). The cake was used as is in the following telescoped reaction.

[0094] As solvents EtOH, DMF or other suitable solvent may be used. Experimental results obtained using different solvents and reaction conditions are shown in Table 3. Difficulty filtering the product of this step (noted in several entries on Table 3) was circumvented by not isolating the solid at this point, but telescoping the reaction with the next step. It has been found that on the order of 20 volumes of EtOH were necessary to achieve reasonable stirring, but that the reaction can proceed in only 10 volumes of DMF, without significant loss in purity. [0095] In Table 3, where the entry is labelled NI , the intermediate product was not isolated, but carried into the next reaction step. Table 3 – Coupling Reaction

 

Figure imgf000035_0001

 

Figure imgf000036_0002

NR = no reaction, NI = not isolated; ND = not determined; NA = not available

1. Carried through to the deprotection and generation of free base to give 69.5% overall yield.

2. The overall yield after the deprotection and generation of the free base is 76.1%

3. This reaction was not filtered at all but taken as slurry to the next step.

Example 4 – Deprotection

[0096] The deprotection of the quinoline intermediate formed by the coupling reaction using

2N HCl in water is preferred as noted in Table 4 below. As in the previous Examples, the intermediate product of this step is advantageously not isolated, but carried over as a wet cake into the next step.

[0097] Preparation of 4-[3-chloro-4-(2-pyridylmethoxy)anilino]-3-cyano-7-ethoxy-6- aminoquinoline hydrochloride.

 

Figure imgf000036_0001

[0098] The reaction mixture from the previous step (Example 3) was taken as is and to it was added 2.7N HCl (3.3L) in H2O (16.0 L). The slurry was heated to 700C and held for 19 hours. The resulting slurry was then filtered and rinsed with 1:1 EtOHTH2O (4 x 1.0 L). The product was isolated as a wet cake and carried through to the next step. A small sample was dried at this stage and analyzed. The HCl salt had a strength of 98.9%. Example 4a

[0099] To prepare 6-amino-4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7- ethoxyquinoline hydrochloride, the reaction mixture from the previous step was taken as is and to it was added ethanol (1.6 L) and concentrated hydrochloric acid (1.38 L) to bring the pH to 1-3. The suspension was held at 70-75 0C for a minimum of 2 h. After 1 h, the mixture thickens and ethanol (0.80 L) was added. After 2 h, water (6.80 L) was added, the mixture stirred for 1 h and then cooled to 35-45 0C and stirred overnight (12 h). The mixture was filtered and rinsed with 1 : 1 ethanol/water (2 x 0.84 L) at 35-45 0C. The product was isolated as a wet cake and carried through to the next step. Example 4b

[00100] To prepare 6-amino-4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7- ethoxyquinoline hydrochloride, the wet cake from the previous step was taken as is and to it was added a 2 N solution of concentrated hydrochloric acid (1.16 L) in methanol (5.84 L). The suspension was heated to 63-68 0C and held for a minimum of 30 h. The mixture was cooled to 20-300C, filtered and rinsed with methanol (2 x 0.30 L). The product was isolated as a wet cake and carried through to the next step. Table 4 – Deprotection

 

Figure imgf000037_0001
Figure imgf000038_0001

ND = not determined (the product was used in the next step as a wet cake) NA = not available SM= starting material

Example 5 – Preparation of free base

[0100] The 4-[3-chloro-4-(2-pyridylmethoxy)anilino]-3-cyano-7-ethoxy-6-aminoquinoline HCl salt was converted to the corresponding free base by treatment with 10% potassium carbonate (1.8 L) in MeOH (2.82 L). The mixture was stirred for a minimum of 2.5 hours and the pH was 9-10. The product was filtered, washed with 1:1 methanol/water (3 x 0.19 L) and dried (at 45-50 C at a pressure of 10 mm Hg, for 24 hours) to give 0.186 kg of product with an overall yield of 86% over 4 steps.

Figure imgf000039_0001

Example 5 a

[0101] To prepare 6-amino-4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7- ethoxyquinoline free base, the 6-amino-4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7- ethoxyquinoline hydrochloride salt was converted to its corresponding free base by treatment with 10% potassium carbonate (0.22 kg in 2.27 L water) in methanol (7.21 L) until pH was 10. The mixture was stirred for a minimum of 2 h. The beige suspension was filtered, washed with 1:1 methanol/water (2 x 0.84 L) and dried (45-50 0C, 10 mm Hg, 24 h) to give 0.51 kg of product with an overall yield of 99% over 4 steps. Example 5b

[0102] To prepare 6-amino-4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxyquinolme free base, the 6-amino-4-[4-(benzyloxy)-3-chloroamlino]-3-cyano-7-ethoxyqumoline hydrochloride salt was converted to its corresponding free base by treatment with 10% aqueous potassium carbonate (0.213 kg in 2.13 L) in methanol (6.40 L). The mixture was stirred for a minimum of 1.5 h keeping the pH at 9-10. The product was filtered, washed with water (2 x 0.50 L) and dried (50-60 0C, 10 mm Hg, 20 h) to give 0.347 kg of product with an overall yield of 82% over 4 steps.

Example 6 – Side Chain Coupling

[0103] An acid chloride of formula RV(C=O)-Cl, a mixed anhydride or an activated carboxylase R’ 2-(C=O)-LG derived from the corresponding carboxylic acid, may be used to couple a side chain at the 6 position to form a 6-amido-4-amino-3 cyanoquinoline. R’2 may be alkyl of 1-6 carbon atoms, which may be mono- or di-substituted with amino groups or cycloamino groups, or R’2 may be alkenyl of 2-6 carbon atoms which may be mono- or di- substituted with amino groups or cycloamino groups. [0104] Using the 2-step sequence shown below, an activated carboxylate is prepared in situ and coupled with the aniline. Although the acid chloride can be prepared in acetonitile, a better yield was obtained when the acid chloride was prepared in THF. In both cases, the aniline should be dissolved in NMP before amidation. It is believed that formation of product is better due to better solubility of the aniline in a THF/NMP mixture rather than in an ACN/NMP combination.

 

Figure imgf000040_0001

[0105] The amount of 4-N,N-dimethylaminocrotonic acid needed was 2 equivalents with respect to aniline. A slight undercharge of 1.95 eq of oxalyl chloride was added along with a catalytic amount (3 mol %) of DMF. The acid chloride was formed via the Vilsmeier intermediate. The completion test for the acid chloride reaction consists of quenching an aliquot of the reaction into ethanol and detecting by HPLC the crotonic acid ethyl ester. This method serves as a check to ensure complete consumption of oxalyl chloride. Excess oxalyl chloride will form diethyl oxalate when quenched in ethanol. [0106] The acid chloride is stable after holding for up to 5 hours at 0-10 °C, when decomposition begins. After 20 hours, complete decomposition takes place. If the acid chloride is allowed to warm, decomposition occurs and its effectiveness is diminished. [0107] The quality of the starting crotonic acid also plays a role in this coupling reaction, as commercially available crotonic acid may contain acetic acid. Acetic acid is detrimental to this reaction. 6-N-acetyl quinoline can be formed which is difficult to remove from the final product. The acetic acid can be removed by re-slurrying the crotonic acid in 4 volumes of isopropanol at room tempature, filtering and drying preferably to a level of less than 0.01%. [0108] It was found that the addition of the aniline solution in NMP to the acid chloride gave a better yield as compared to adding the acid chloride to the aniline. The addition is done keeping the temperature at 0-5 °C. The coupling reaction is slow and requires holding overnight at this temperature. It is not desirable to raise the reaction temperature as the stability of the acid chloride diminishes.

[0109] The reaction is quenched using aqueous sodium hydroxide at 40 °C and then filtered at that temperature. Quenching the reaction at 40 0C gives bigger crystals that are easily filterable. It was observed that filtration at 40 °C was faster than at room temperature. The product is recrystallized from a 1.5:1 mixture of acetonitrile:THF (15 volumes) at 70-75 0C. This in-process purification beneficially removes unreacted aniline. The recovery yields are typically greater than 85%.

[0110] To demonstrate a specific synthesis of (E)-N- {4-[3-chloro-4-(2- pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide, a solution of 4-N,N-dimethylaminocrotonic acid hydrochloride (186 g, 1.12 mol) in THF (1.88 L) and a catalytic amount of DMF (2 mL) was cooled to 0-5 °C. Oxalyl chloride (97 mL, 1.09 mol, 0.95 eq) was added dropwise over 45 minutes. The mixture was then warmed to 25-30 °C and stirred for 2 hours. The yellow solution was checked for complete consumption of oxalyl chloride by HPLC, then cooled to 0-5 0C.

[0111] When the reaction is deemed complete, a solution of 4-[4-(2-pyridylniethoxy)-3- chloro]amino-6-amino-3-cyano-7-ethoxyquinoline (250 g, 0.56 mol) in N-methyl-2- pyrolidinone (1.88 L) was added dropwise over 2 hours keeping the temperature at 0-5 °C. The mixture was stirred for at least 3 hours until less than about 2% of the starting aniline remains by HPLC, which takes about 3 hours.

[0112] Upon completion, the reaction was quenched with water (3.0 L), held for 30 minutes and then warmed to 40 °C. Aqueous sodium hydroxide (170 g in 1.25 L water) was added over 1.25 hours to bring the pH to 10-11. The mixture was stirred for an hour, then cooled to room temperature and held for 3 hours. The resulting precipitates were filtered and washed with water (100 mL) and heptane (100 mL). The wet solids were heated to reflux (70-75 °C) in acetonitrile:THF and the solution cooled over 3 hours to room temperature. The product was filtered and washed with cold acetonitrile:THF. The product was dried (40-50 0C, 10 mm Hg, 24 hours) to give 83% uncorrected yield. Example 6a

[0113] In an analogous synthesis of (E)-N- {4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3- cyano-7-ethoxy-6-qumolmyl}-4-(dimethylamino)-2-butenamide, a solution of 4-N5N- dimethylaminocrotonic acid hydrochloride (108 g, 0.65 mole) in tetrahydrofuran (1.13 L) and a catalytic amount of dimethylformamide (1.2 mL) was cooled to 0-5 °c. Oxalyl chloride (55 mL, 0.62 mole, 0.95 eq) was added dropwise over 50 min. The mixture was then warmed to 25-30 °c and stirred for 2 h then cooled to 0-5 °c. N-methyl-2-pyrrolidinone (0.225 L) was added over 25 min followed by a solution of 6-amino-4-[3-chloro-4-(3- fluorobenzyloxy)]anilino-3-cyano-7-ethoxy-quinoline (150 g, 0.32 mol) in N-methyl-2- pyrrolidinone (1.20 L) added dropwise over 2 hours keeping the temperature 0-5 . The mixture was stirred for at least about 3 hours, warmed to 10-15 °c and stirred for a further 12 hours. The mixture is cooled to 0-10 c, quenched by adding water (1.8 L) over 2 hours, and stirred for 30 minutes. The mixture is warmed to 40 °c. Aqueous sodium hydroxide (101 g in 0.75 L water) was added over 1 hour to bring the pH to 10-11. The mixture was stirred for an hour, filtered warm (40 °c) and washed with water (2 x 0.30 L) until the pH of the last wash was about 7. The wet solids were recrystallized by heating to reflux (70-75 °c) in 60:40 acetonitrile:tetrahydrofuran (2.25 L) and the solution cooled over 3 hours to room temperature. The product was filtered and washed with cold 60:40 acetonitrile:tetrahydrofuran (2 x 0.30 L). The product was dried (40-50 °c, 10 mm Hg, 16 h) to give 0.154 kg (83% yield). Example 6b

[0114] To prepare (E)-N- {4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxy-6-quinolinyl}- 4-(dimethylamino)-2-butenamide free base, a solution of 4-N,N-dimethylaminocrotonic acid hydrochloride (18.6 g, 112 mmole) in acetonitrile (295 ml) and a catalytic amount of dimethylformamide (0.25 mL) was cooled to 0-5 °c. Oxalyl chloride (9.3 mL, 106 mmole, 0.95

Op eq) was added dropwise over 5 min. The mixture was then warmed to 25-30 and stirred for 1-1.5 h then cooled to 0-10 °c. A solution of 6-amino-4-[4-(benzyloxy)-3-cliloroanilino]-3- cyano-7-ethoxy-quinoline (25 g, 56 mmole) in N-methyl-2-pyrrolidinone (175 ml) was added dropwise over 30 min keeping the temperature 0-10 °c. The mixture was stirred for a minimum of 1 h at 0-10 °c. After reaction completion, the mixture was quenched by dropwise addition to a solution of sodium bicarbonate (69.7 g in 870 ml water) over 30 mins. and stirred overnight while warming to room temperature. The mixture was filtered and washed with water (3 x 25 ml). The crude product was recrystallized in refluxing (80-82 °c) acetonitrile (570 ml). The product was dried (45-50 °c, 10 mm Hg, 28 h) to give 12.81 g (41% yield). 1H NMR : δ (DMSO-d6) 9.44 (s, IH, NH), 8.97 (s, IH, Ar), 8.44 (s, IH, Ar), 7.53-7.35 (m, 7H, Ar), 7.35- 7.10 (in, 2H, Ar), 6.78 (dt, IH, -CH2CH=CH-), 6.59 (d, IH, -CH2CH=CH-), 5.21 (s, 2H, OCH2Ph), 4.30 (q, 2H, OCH2CH3), 3.07 (s, 2H, NCH2), 2.18 (s, 6H, N(CHs)2), 1-47 (t, 3H, OCH2CH3).

[0115] Results obtained with different reaction procedures at different degrees of scale-up for synthesis of the 2-pyridylmethoxy analog are shown in Table 5. Table 5 – Side Chain Coupling

 

Figure imgf000043_0001
Figure imgf000044_0001

* TI = total impurities

[0116] Purificatiuon of the product is conducted by recrystallization in a suitable solvent followed by reslurrying with water followed by additional recrystallization, as necessary. As noted in Table 6, in the synthesis of the 2-pyridylmethoxy analog, several trials in different solvents did not result in the isolation of a single polymorphic form of the product. Table 6

 

Figure imgf000044_0002
Figure imgf000045_0001

Example 7 – Formation of Salt

[0117] The free base is hygroscopic and undergoes hydrolysis readily. Forming a salt of the compound, such as a fumarate or mesylate salt, stabilizes the molecule and renders the compound more soluble. The most preferred salt is a maleate salt, which has been found to be highly crystalline and to exist substantially as a single polymorph as shown by DSC thermogram in Fig. 1.

[0118] Recrystallizing the product in the presence of an acid has been found to yield a stable salt form of the product. Experimental results achieved utilizing different solvents for the recrystallization are set forth in Table 7. As seen in Table 7, an improvement is observed when n-propanol/water is used as the solvent system. A maleate salt is the most preferred, as it exists in a single polymorphic form. Table 7 – Recrystallization

 

Figure imgf000045_0002
Figure imgf000046_0001
Figure imgf000047_0001

Preparation of (E)-N- {4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6- quinolinyl} -4-(dimethylamino)-2-butenamide maleate, WAY- 179272-B

[0120] (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4- dimethylamino)-2-butenamide crude free base (0.1 kg, 0.159 mole) and maleic acid (0.019 kg, 0.164 mole) were dissolved at 40-50 in a 10% water/n-propanol mixture (1.20 L). The hot solution was clarified and cooled over 2 h to room temperature and held for 12-15 hr. The product was filtered and washed with 10% water/n-propanol (2 x 0.15 L). The product was dried (50 °c, 10 mm Hg, 24 h) to give 94.4 g (88% yield). DSC: 204 °c (single crystal form). 1H NMR : δ (DMSO-d6) 9.73 (s, IH, NH), 9.62 (s, IH, NH), 8.93 (s, IH, Ar), 8.60 (dd, IH, Ar), 8.50 (s, IH, Ar), 7.88 (dd, IH, Ar), 7.58 (d, IH, Ar), 7.40 (m, 3H, Ar), 7.24 (m, 2H, Ar), 6.75 (d, 2H, -CH=CH-), 6.03 (s, 2H, HOOC-CH=CH-COOH), 5.29 (s, 2H, OCH2PVr), 4.33 (q, 2H, OCH2CH3), 3.89 (s, 2H, NCH2), 2.76 (s, 6H, N(CH3)2), 1.47 (t, 3H, OCH2CH3). 13C NMR : δ (DMSO-d6) 168.0, 163.2, 156.9, 154.2, 153.2, 151.9, 151.3, 149.8, 148.5, 137.8, 136.5, 134.7, 133.4, 132.2, 128.0, 126.6, 124.9, 123.8, 122.3, 122.2, 117.9, 116.4, 115.1, 113.9, 109.5, 88.1, 72.0, 65.3, 57.8, 43.1, 14.9.

Example 7a

To prepare (E)-N- {4-[3-chloro-4-(3-fluorobenzyloxy)anilino]-3-cyano-7-ethoxy-6- quinolinyl}-4-(dimethylamino)-2-butenamide dimaleate,

 

(E)-N- {4-[3-chloro-4-(3- fluorobenzyloxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-dimethylamino)-2-butenamide crude free base (0.516 kg, 0.90 mole) and maleic acid (0.214 kg, 1.84 mole) were dissolved at 40-50 °c in a 6.5% water/n-propanol mixture (12.60 L). The hot solution was clarified, rinsed with 5% water/n-propanol (0.52 L) and n-propanol (2.0 L). The mixture was held at 45 for 3 hr, cooled over 2 h to room temperature and held overnight. The mixture was further cooled to 5-10 °c. The product was filtered and washed with cold 5% water/n-propanol (0.52 L). The product was dried (45 °c, 10 mm Hg, 16-24 h) to give 0.586 kg (81% yield). DSC: 184 °c (single crystal form). 1HNMR : δ (DMSO-d6) 9.77 (s, IH, NH), 8.95 (s, IH, Ar), 8.53 (s, IH, Ar), 7.49-7.16 (m, 8H, Ar), 6.78 (m, 2H, -CH=CH-), 6.15 (s, 4H, 2 x HOOC-CH=CH-COOH), 5.26 (s, 2H, OCH2PyT), 4.33 (q, 2H, OCH2CH3), 3.97 (dd, 2H, NCH2), 2.82 (s, 6H, N(CEb)2), 1.47 (t, 3H, OCH2CH3). 13C NMR : δ (DMS0-d6) 167.0, 163.8, 162.3, 160.6, 153.6, 152.2, 151.3, 150.8, 139.5, 139.4, 133.7, 133.2, 132.2, 131.8, 130.5, 130.4, 127.4, 126.1, 124.3, 123.3, 121.7, 116.9, 115.7, 114.8, 114.5, 114.4, 114.1, 113.8, 113.1, 108.1, 87.2, 69.5, 64.6, 56.9, 42.1, 14.2. Example 7b

[0122] To prepare (E)-N- {4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxy-6-quinolinyl}- 4-(dimethylamino)-2-butenamide maleate, (E)-N- {4-[4-(benzyloxy)-3-chloroanilino]-3-cyano- 7-ethoxy-6-quinolinyl}-4-dimethylamino)-2-butenamide crude free base (2.0 g, 3.6 mmole) and maleic acid (0.43 g, 3.7 mmole) were mixed at 40-50 c in a 10% water/n-propanol mixture (24 ml) for 2 hr. The mixture was cooled to ambient temperature, filtered and washed with 10% water/n-propanol (2 x 3 ml). The product was dried (40 °c, 10 mm Hg, 24 h) to give 0.32 g (13% yield). 1HNMR : δ (DMSO-d6) 9.75 (s, IH, NH), 8.95 (s, IH, Ar), 8.49 (s, IH, Ar), 7.49-7.37 (m, 7H, Ar), 7.23 (dd, 2H, Ar), 6.78 (s, 2H, -CH2CH=CH-), 6.06 (s, 2H, HOOC- CH=CH-COOH), 5.22 (s, 2H, OCH2Ph), 4.31 (q, 2H, OCH2CH3), 3.93 (s, 2H, NCH2), 2.79 (s, 6H, N(CH3)2), 1.46 (t, 3H, OCH2CH3).13C NMR : δ (DMSO-d6) 167.9, 163.1, 154.2, 153.3, 152.1, 151.3, 148.5, 137.3, 136.3, 134.5, 133.2, 132.3, 129.3, 129.2, 128.7, 128.3, 128.2, 128.0, 126.7, 124.9, 122.4, 117.9, 116.4, 115.2, 113.9, 109.5, 88.0, 71.1, 65.3, 57.7, 43.0, 15.0. [0123] (E)-N-{4-[4-(benzyloxy)-3-chloroanilino]-3-cyano-7-ethoxy-6-quinolinyl}-4- dimethylamino)-2-butenamide crude free base (2.0 g, 3.6 mmole) and maleic acid (0.43 g, 3.7 mmole) were mixed at 40-50 °c in a 10% water/n-propanol mixture (24 ml) for 2 hr. The mixture was cooled to ambient temperature, filtered and washed with 10% water/n-propanol (2 x 3 ml). The product was dried (40 °c, 10 mm Hg, 24 h) to give 0.32 g (13% yield). 1H NMR : δ (DMSO-d6) 9.75 (s, IH, NH), 8.95 (s, IH, Ar), 8.49 (s, IH, Ar), 7.49-7.37 (m, 7H, Ar), 7.23 (dd, 2H, Ar), 6.78 (s, 2H, -CH2CH=CH-), 6.06 (s, 2H, HOOC-CH=CH-COOH), 5.22 (s, 2H, OCH2Ph), 4.31 (q, 2H, OCH2CH3), 3.93 (s, 2H, NCH2), 2.79 (s, 6H, N(CH3)2), 1.46 (t, 3H, OCH2CH3). 13C NMR : δ (DMSO-d6) 167.9, 163.1, 154.2, 153.3, 152.1, 151.3, 148.5, 137.3, 136.3, 134.5, 133.2, 132.3, 129.3, 129.2, 128.7, 128.3, 128.2, 128.0, 126.7, 124.9, 122.4, 117.9,

116.4, 115.2, 113.9, 109.5, 88.0, 71.1, 65.3, 57.7, 43.0, 15.0.

 

……………….

http://www.google.com/patents/WO2009052264A2?cl=en

TABLE 1 1. STRUCTURES OF DEGRADATION PRODUCT AND PROCESS IMPURITIES

 

Figure imgf000025_0001

N-{4-[3-chloro-4-(2- (E)-4-({4-[3-chloro-4-(2- N -{4-[3-chloro-4-(2- pyrιdιnylmethoxy)anιlιno]-3-cyano-7- pyrιdιnylmethoxy)anιlιno]-3-cyano-7- pyrιdιnylmethoxy)anιlιno]-3-cyano-7-ethoxy- ethoxy-6-quιnolιnyl}acetamιde ethoxy-6-quιnolιnyl}amιno)-N,N,N- 6-quιnolιnyl}-N2,N2-dιmethylethanedιamιde trιmethyl-4-oxo-2-buten-1-amιnιum

Exact Mass 487 14 Exact Mass 544 16

Exact Mass 571 22

Process Impurity I Process Impurity J

 

Figure imgf000026_0001

SCHEME 1

 

Figure imgf000014_0001

The reaction of the free base and maleic acid occurs at an elevated temperature of from about 40 0C to about 60 0C, preferably between about 4O0C to about 5O0C. The ratio of watenn- propanol may vary, for example between about 1 :10 to about 1 :5, and the optimal ratio of watenn-propanol is about 1 :9. The water-alcohol solution may comprise from about 5% to about 20% by volume water and from about 80% to about 95% by volume alcohol. The alcohol may be n-propanol. In one embodiment, the water-alcohol solution comprises about 10% by volume water and about 90% by volume n-propanol. The volume of the solvent solution may be between about 8 to about 25 volumes, including about 10 to about 12 volumes. About 1.0-1.2 equivalents of maleic acid is used per equivalent of the free base, preferably about 1.03 equivalents of maleic acid per equivalent of the free base.

The resulting solution of the maleate salt may be clarified by filtration prior to cooling. The cooling step may be continued until the solution reaches a temperature of about 45°C or less, including a temperature of about 39°C or less, and more preferably to about 300C or less. In one embodiment, the solution is filtered after cooling to about room temperature, preferably from about 230C to about 25 0C. Typically, the maleate salt begins to crystallize out of solution once the temperature reaches 370C or below. The solution may be allowed to sit for at least 12 hours, preferably about 12 to about 15 hours at room temperature, and is then filtered and washed to recover the crystalline maleate salt product. The resulting filter cake may be washed with the same or a different water-alcohol solution to obtain the product. The product may be dried to obtain crystalline (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7- ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide maleate. At this point, the maleate salt product recovered and isolated is typically in the form of the monohydrate form of the maleate salt.

 

 

……………

http://www.google.com/patents/CN102731395A?cl=en

 The present invention relates to a process for preparing that imatinib (neratinib, HKI-272) is a new method for its preparation and its intermediates in the preparation to the application that imatinib

Figure CN102731395AD00071
Figure CN102731395AD00072

[0155] Example 14 (E)-N-(4 – (3 – chloro-4 – (2 – pyridyl) phenyl) amino] _3_ ethoxy-quinolin-6-cyano-_7_ – yl) -4 – dimethylamino-2 – butene amide

[0156]

Figure CN102731395AD00202

[0157] Compound of Example 13 (20mg, 0. 037mmol) was dissolved in DMF was added potassium carbonate (10mg, 0. 07mmol), dimethylamine hydrochloride (5mg, 0. 06mmol), at room temperature for I hour, after , the reaction mixture was dropped into water, stirred for 10 minutes, filtered, washed with water and dried to give the title compound 1511 ^ 75% yield.1HNMR (300MHz, DMS0_d6): δ I. 5 (t, 3H, J = 6 · 8,13. 8), 2. 2 (br s, 6H), 3. I (d, 2H, J = 3. 8 ), 4. 3 (q, 2H, J = 7. 0,14. 2), 5. 2 (s, 2H),

6. 6 (d, 1H, J = 15. 0), 6. 8 (m, 1H), 7. 1-7. 3 (m, 2H), 7. 3-7. 4 (m, 3H), 7. 6 (d, 1H, J = 3. 9),

7. 9 (d, 1H, J = 3. 9), 8. 5 (s, 1H), 8. 6 (d, 1H, J = 3. 9), 9. 0 (s, 1H), 9. 5 (s, 1H), 9. 6 (s, 1H). ESI-MS: [M + H] + = 557. 3.

GOING BACKWARDS…………………

[0152] Example 13 (E) -4 – bromo-N-(4 – (3 – chloro-4 – (2 – pyridyl) phenyl) amino] _3_ cyano _7_ ethoxyquin -6 – yl) -2 – butene amide

[0153]

Figure CN102731395AD00201

[0154] Example 12 Compound (100mg, 0. 2mmol) was suspended in carbon tetrachloride was added NBS (40mg,

O. 22mmol), benzoyl peroxide (2mg, 0. Olmmol), nitrogen, refluxed for 10 hours, the reaction solution directly mixed baby gel, silica gel column chromatography to obtain the title compound isolated 60mg, yield 51%. 1HnmrgoomHz, cdci3): δ i.6 (t, 3H, J = 6. 8,13. 7), 2. 0 (d, 2H, J = 6. 9), 4. 3 (q, 2H, J = 7. 2,13. 8), 5. 3 (s, 2H), 6. I (d, 1H, J =

15. 0), 7. 0 (m, 1H), 7. 2 (m, 1H), 7. 3 (s, 1H), 7. 4 (s, 1H), 7. 6 (d, 1H, J = 8. 2), 7. 8 (d, 1H, J =

7. 6), 8. 0 (s, 1H), 8. 5 (s, 1H), 8. 6 (d, 1H, J = 4. 7), 9. 2 (s, 1H). ESI-MS: [M + H] + = 594. I.

……………

Optimization of 6,7-disubstituted-4-(arylamino)quinoline-3-carbonitriles as orally active, irreversible inhibitors of human epidermal growth factor receptor-2 kinase activity
J Med Chem 2005, 48(4): 1107

http://pubs.acs.org/doi/full/10.1021/jm040159c

Abstract Image

 

(E)-N-{4-[3-Chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide (25o). 

This compound was prepared as a yellow solid (0.86 g, 85%) by the method described for 25g using 0.65 g (1.81 mmol) of 23 and 0.42 g (3.62 mmol) of 3-chloro-4-(2-pyridinylmethoxy)aniline: 

HRMS (ES+) m/z 557.205 89 (M + H)+1, Δ = −0.36 mmu; 

1H NMR (DMSO-d6) δ 9.62 (s, 1H), 9.49 (s, 1H), 8.96 (s, 1H),

8.60 (d, 1H, J = 3.9 Hz), 8.47 (s, 1H),

7.88 (t, 1H, J = 3.9 Hz), 7.58 (d, 1H, J = 3.9 Hz),

7.39−7.35 (m, 3H), 7.26 (d, 1H, J = 7.8 Hz),

7.19 (d, 1H, J = 8.1 Hz), 6.81−6.73 (m, 1H),

6.59 (d, 1H, J = 7.8 Hz), 5.28 (s, 2H),

4.30 (q, 2H, J = 6.9 Hz),

3.07 (d, 2H, J = 3.9 Hz),

2.17 (s, 6H),

1.46 (t, 3H, J = 3.9 Hz).

Anal. (C30H29ClN6O3·1.1H2O) C, H, N.

INTERPRETATION

1H NMR : δ (DMSO-d6)

9.44 (s, IH, NH),

8.97 (s, IH, Ar),

8.44 (s, IH, Ar),

7.53-7.35 (m, 7H, Ar),

7.35- 7.10 (in, 2H, Ar),

6.78 (dt, IH, -CH2CH=CH-),

6.59 (d, IH, -CH2CH=CH-),

5.21 (s, 2H, OCH2Ph),

4.30 (q, 2H, OCH2CH3),

3.07 (s, 2H, NCH2),

2.18 (s, 6H, N(CHs)2),

1-47 (t, 3H, OCH2CH3).

References

  1.  “Definition of neratinib – National Cancer Institute Drug Dictionary”. Retrieved 2008-12-01.
  2.  Rabindran SK, Discafani CM, Rosfjord EC, et al. (June 2004). “Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase”Cancer Res. 64 (11): 3958–65. doi:10.1158/0008-5472.CAN-03-2868PMID 15173008.
  3. ClinicalTrials.gov NCT00398567 A Phase 1/2 Study Of HKI-272 In Combination With Herceptin In Subjects With Advanced Breast Cancer
  4.  “Puma Acquires Global Rights to Pfizer’s Phase III Breast Cancer Drug Neratinib”.
  5.  Minami Y, Shimamura T, Shah K, et al. (July 2007). “The major lung cancer-derived mutants of ERBB2 are oncogenic and are associated with sensitivity to the irreversible EGFR/ERBB2 inhibitor HKI-272″. Oncogene 26 (34): 5023–7. doi:10.1038/sj.onc.1210292.PMID 17311002.
  6. http://www.reuters.com/article/idUSN1612347120100317 “Breast cancer study aims to speed drugs, cooperation” March 2010
  7. Sequist L.V., Besse B., Lynch T.J. and all; Neratinib, an Irreversible Pan-ErbB Receptor Tyrosine Kinase Inhibitor: Results of a Phase II Trial in Patients With Advanced Non-Small-Cell Lung Cancer., J. Clin. Oncol., 2010, May 17.  
    PubMed PMID: 20479403.
  8. Belani CP. The role of irreversible EGFR inhibitors in the treatment of non-small cell lung cancer: overcoming resistance to reversible EGFR inhibitors. Review.  Cancer Invest. 2010, 28(4), 413-423. Review. 
    PubMed PMID: 20307200.
  9. TSOU H-R ET AL: “Optimization of 6,7-Disubstituted-4-(arylamino)quinoline-3 -carbonitr iles as Orally Active, Irreverible Inhibitors of HEGFR-2 Kinase Activity” JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, US, vol. 48, 27 January 2005 (2005-01-27), pages 1107-1131, XP002414228 ISSN: 0022-2623 cited in the application
  10. Optimization of 6,7-disubstituted-4-(arylamino)quinoline-3-carbonitriles as orally active, irreversible inhibitors of human epidermal growth factor receptor-2 kinase activity
    J Med Chem 2005, 48(4): 1107
2-20-2009
Novel Combinational Use of Sulfonamide Compound
9-12-2008
Multi-Functional Small Molecules as Anti-Proliferative Agents
5-11-2007
Antineoplastic combinations with mTOR inhibitor,herceptin, and/or hki-272
11-31-2006
Methods of synthesizing substituted 3-cyanoquinolines and intermediates thereof
11-31-2006
Methods of synthesizing 6-alkylaminoquinoline derivatives
10-25-2006
Synthesis of 4-(amino)-2-butenoyl chlorides and their use in the preparation of 3-cyano quinolines
5-30-2012
Amide derivative for inhibiting the growth of cancer cells
9-21-2011
Maleate salts of (E)-N-{4-[3-chloro-4-(2-pyridinylmethoxy)anilino]-3-cyano-7-ethoxy-6-quinolinyl}-4-(dimethylamino)-2-butenamide and crystalline forms thereof
8-26-2011
COMPOUNDS THAT MODULATE EGFR ACTIVITY AND METHODS FOR TREATING OR PREVENTING CONDITIONS THEREWITH
5-7-2010
Antineoplastic Combinations of 4-Anilino-3-Cyanoquinolines and Capecitabine
4-30-2010
METHOD FOR PREDICTION OF THE EFFICACY OF VASCULARIZATION INHIBITOR
4-16-2010
METHOD FOR ASSAY ON THE EFFECT OF VASCULARIZATION INHIBITOR
3-19-2010
PHARMACEUTICAL COMPOSITIONS OF AN SRC KINASE INHIBITOR AND AN AROMATASE INHIBITOR
2-26-2010
Heterocyclic N-Oxides as Hypoxic Selective Protein Kinase Inhibitors
12-18-2009
Antineoplastic Combinations Containing HKI-272 and Vinorelbine
12-4-2009
ANTINEOPLASTIC COMBINATIONS WITH mTOR INHIBITOR, TRASTUZUMAB, AND/OR HKI-272

Filed under: Phase3 drugs, Uncategorized Tagged: Anthony crasto, HKI 272, NERATINIB, NEW DRUGS, organic chemistry, PROCESS, world drug tracker

Ambit Biosciences announces Phase 3 trial comparing quizartinib as monotherapy to chemotherapy regimens in relapsed/refractory acute myeloid leukemia (AML) patients with the FMS-like tyrosine kinase-3 (FLT3)-ITD mutation.

$
0
0
File:Quizartinib.svg
QUIZARTINIB
1-(5-(tert-Butyl)isoxazol-3-yl)-3-(4-(7-(2-morpholinoethoxy)benzo[d]imidazo[2,1-b]thiazol-2-yl)phenyl)urea

 N-(5-tert-butyl-isoxazol-3-yl)-N’-{ 4- [7-(2-morpholin-4-yl-ethoxy)imidazo [2, 1 -b] [ 1 ,3 ]benzothiazol-2-yl]phenyl } urea

 FOR acute myeloidLeukemia,
CAS 950769-58-1 (free base)      1132827-21-4 (2HCl)
Formula C29H32N6O4S
MW 560.7
Synonim AC220, AC-010220
ASP-2689
PATENTS
 U.S. Provisional Patent App. No. 60/743,543, filed March 17, 2006, U.S. Patent App. No. 11/724,992, filed March 16, 2007, and U.S. Patent App. Publication No. 2007/0232604, published October 4, 2007,
BioNews TexasAmbit initiates QUANTUM-R Phase 3 clinical trial of quizartinib in FLT3-ITD …News-Medical.net… the treatment of both newly diagnosed and relapsed FLT3-ITD positive and negative AML patients.
Both the U.S. Food and Drug Administration (FDA) and European Commission have granted orphan drug designation to quizartinib for the treatment of AML.AML, High Risk MDS Therapy
see

Quizartinib

Ambit Biosciences

 13 MAY 2013

Ambit Biosciences (NASDAQ:AMBI) is a biotech company that focuses on treatments that inhibit kinases, which are drivers for diseases such as cancer. Three drugs are in development, with the lead one being quizartinib — a Phase 2B trial treatment for acute myeloid leukemia. However, AMBI’s collaboration agreement with Astellas Pharma is set to expire in September, and if it is not replaced, it could mean a delay in Phase 3 trials for quizartinib. Keep in mind that AMBI generated $23.8 million in collaboration revenues last year.

Quizartinib (AC220) is a small molecule receptor tyrosine kinase inhibitor that is currently under development by Ambit Biosciencesfor the treatment of acute myeloid leukaemia. Its molecular target is FLT3, also known as CD135 which is a proto-oncogene.[1]

AC-220 is an angiogenesis inhibitor that antagonizes several proteins involved in vascularization. It was engineered by Ambit Biosciences using KinomeScan technology to potently target FLT3, KIT, CSF1R/FMS, RET and PDGFR kinases. Ambit is developing oral AC-220 in phase III clinical studies for the treatment of relapsed/refractory acute myeloid leukemia (AML) patients with the FMS-like tyrosine kinase-3 (FLT3)-ITD mutation. Early clinical trials are also ongoing for the treatment of advanced solid tumors, for the treatment of refractory or relapsed myelodysplasia, in combination with induction and consolidation chemotherapy for previously-untreated de novo acute myeloid leukemia, and as a maintenance therapy of AML following hematopoietic stem cell transplantation (HSCT). In 2009, orphan drug designation was received both in the U.S. and in the EU for the treatment of AML. In 2009, Ambit Biosciences and Astellas Pharma have entered into a worldwide agreement to jointly develop and commercialize the drug candidate for the treatment of cancer and non-oncology indications. This agreement was terminated in 2013.

Flt3 mutations are among the most common mutations in acute myeloid leukaemia due to internal tandem duplication of Flt3. The presence of this mutation is a marker of adverse outcome.

Quizartinib is a small molecule with potential anticancer activity. Quizartinib is a selective inhibitor of class III receptor tyrosine kinases, including FMS-related tyrosine kinase 3 (FLT3/STK1), stem cell factor receptor (SCFR / KIT), colony-stimulating factor 1 receptor (CSF1R/FMS) and platelet-derived growth factor receptors (PDGFRs .) Able to inhibition of ligand-independent cell proliferation and apoptosis. Mutations in FLT3 are the most frequent genetic alterations in acute myeloid leukemia (AML) and occur in approximately 30% of cases of AML.
Quizartinib представляет собой малую молекулу с потенциальной противораковой активностью. Quizartinib является селективным ингибитором класса III рецепторов тирозин киназ, в том числе FMS-связанных тирозинкиназы 3 (FLT3/STK1), фактор стволовых клеток рецепторов (SCFR / KIT), колониестимулирующий фактор 1 рецепторов (CSF1R/FMS)  и тромбоцитарный рецепторов фактора роста (PDGFRs). Способен к торможению лиганд-независимой клеточной пролиферации и апоптоза. Мутации в FLT3 являются наиболее частыми генетическими изменениями в остром миелобластном лейкозе (ОМЛ) и встречаются примерно в 30% случаев ОМЛ.

Mechanism

Specifically, Quizartinib selectively inhibits class III receptor tyrosine kinases, including FMS-related tyrosine kinase 3 (FLT3/STK1), colony-stimulating factor 1 receptor (CSF1R/FMS), stem cell factor receptor (SCFR/KIT), and platelet derived growth factor receptors (PDGFRs).

Mutations cause constitutive action of Flt3 leading to resulting in inhibition of ligand-independent leukemic cell proliferation and apoptosis.

Clinical trials

It had good results in a phase II clinical trial for refractory AML – particularly in patients who went on to have a stem cell transplant.[2]

………………………..

WO 2007109120 COMPD B1

EXAMPLE 3: PREPARATION OF N-(5-TERT-BUTYL-ISOXAZOL-3-YL)-N’-{4-[7-(2- MORPHOLIN-4-YL-ETHOXY)IMIDAZO[2,1 -B3[1 ,3]BENZOTHIAZOL-2-YL]PHENYL}UREA [Compound B1]

[00426] A. The intermediate 2-amino-1,3-benzothiazol-6-ol was prepared according to a slightly modified literature procedure by Lau and Gompf. J. Org. Chem. 1970, 35, 4103-4108. To a stirred solution of thiourea (7.6 g, 0.10 mol) in a mixture of 200 ml_ ethanol and 9 ml_ concentrated hydrochloric acid was added a solution of 1 ,4-benzoquinone (21.6 g, 0.20 mol) in 400 mL of hot ethanol. The reaction was stirred for 24 hours at room temperature and then concentrated to dryness. The residue was triturated with hot acetonitrile and the resulting solid was filtered and dried.

[00427] The free base was obtained by dissolving the hydrochloride salt in water, neutralizing with sodium acetate, and collecting the solid by filtration. The product (2-amino-1 ,3-benzothiazol-6-ol) was obtained as a dark solid that was pure by LCMS (M+H = 167) and NMR. Yield: 13.0 g (78 %). NMR (DMSOd6) £7.6 (m, 2H ), 6.6 (d, 1H).

[00428] B. To prepare the intermediate 2-(4-nitrophenyl)imidazo[2,1- b][1 ,3]benzothiazoI-7-ol, 2-amino-1 ,3-benzothiazol-6-ol, (20.0 g, 0.12 mol) and 2-bromo-4′-nitroacetophenone (29.3 g, 0.12 mol) were dissolved in 600 mL ethanol and heated to reflux overnight. The solution was then cooled to 00C in an ice-water bath and the product was collected by vacuum filtration. After drying under vacuum with P2O5 , the intermediate (2-(4- nitrophenyl)imidazo[2,1-_D][1,3]benzothiazol-7-ol) was isolated as a yellow solid. Yield: 17.0 g (46 %) NMR (DMSO-CT6) δ 10 (s, 1 H), 8.9 (s, 1H), 8.3 (d, 2H), 8.1 (d, 2H), 7.8 (d, 1 H), 7.4 (s, 1 H), 6.9 (d, 1 H). [00429] C. To make the 7-(2-morpholin-4-yl-ethoxy)-2-(4-nttro- phenyl)imidazo[2,1-£>][1 ,3]benzothiazo!e intermediate: 2-(4- nitrophenyl)imidazo[2,1-jb][1 ,3]benzothiazol-7-ol, (3.00 g, 9.6 mmol) was suspended in 100 mL dry DMF. To this mixture was added potassium carbonate (4.15 g, 30 mmol, 3 eq), chloroethyl morpholine hydrochloride (4.65 g, 25 mmol, 2.5 eq) and optionally tetrabutyl ammonium iodide (7.39 g, 2 mmol). The suspension was then heated to 900C for 5 hours or until complete by LCMS. The mixture was cooled to room temperature, poured into 800 mL water, and allowed to stand for 1 hour. The resulting precipitate was collected by vacuum filtration and dried under vacuum. The intermediate, (7-(2- morpholin-4-yl-ethoxy)-2-(4-nitro-phenyl)imidazo[2,1-jb][1 ,3]benzothiazole) was carried on without further purification. Yield: 3.87 g (95 %) NMR (DMSO-Cf6) δ 8.97 (s, 1 H), 8.30 (d, 2H), 8.0 (d, 2H), 7.9 (d, 1 H), 7.7 (s, 1 H), 7.2 (d, 1 H), 4.1 (t, 2H), 5.6 (m, 4H), 2.7 (t, 2H).

[00430] D. To make the intermediate 7-(2-morpholin-4-yl-ethoxy)-2-(4- amino-phenyl)!midazo[2, 1 -b][1 ,3]benzothiazole: To a suspension of 7-(2- morpholin-4-yl-ethoxy)-2-(4-nitro-phenyl)imidazo[2,1-ib][1 ,3]benzothiazole (3.87g, 9.1 mmol) in 100 ml_ isopropyl alcohol/water (3:1 ) was added ammonium chloride (2.00 g, 36.4 mmol) and iron powder (5.04 g, 90.1 mmol). The suspension was heated to reflux overnight with vigorous stirring, completion of the reaction was confirmed by LCMS. The mixture was filtered through Celite, and the filtercake was washed with hot isopropyl alcohol (150 ml_). The filtrate was concentrated to approximately 1/3 of the original , volume, poured into saturated sodium bicarbonate, and extracted 3 times with dichloromethane. The combined organic phases were dried over MgSO4 and concentrated to give the product as an orange solid containing a small amount (4-6 %) of starting material. (Yield: 2.75 g 54 %). 80% ethanol/water may be used in the place of isopropyl alcohol /water — in which case the reaction is virtually complete after 3.5 hours and oniy traces of starting material are observed in the product obtained. NMR (DMSO-d6) δ 8.4 (s, 1 H), 7.8 (d, 1 H), 7.65 (d, 1 H), 7.5 (d, 2H), 7.1 (d, 1 H), 6.6 (d, 2H), 4.1 (t, 2H)1.3.6 (m, 4H), 2.7 (t, 2H).

[00431] E. A suspension of 7-(2-morpholin-4-yl-ethoxy)-2-(4-amino- phenyl)imidazo[2,1-b][1 ,3]benzothiazole (4.06 g, 10.3 mmol) and 5-tert- butylisoxazole-3-isocyanate (1.994 g, 12 mmol) in toluene was heated at 120 0C overnight. The reaction was quenched by pouring into a mixture of methylene chloride and water containing a little methanol and neutralized with saturated aqueous NaHCO3 solution. The aqueous phase was extracted twice with methylene chloride, the combined organic extracts were dried over MgSO4 and filtered. The filtrate was concentrated to about 20 ml volume and ethyl ether was added resulting in the formation of a solid. The precipitate was collected by filtration, washed with ethyl ether, and dried under vacuum to give the free base. Yield: 2.342 g (41 %) NMR (DMSO-Cf6) £9.6 (br, 1H), 8.9 (br, 1H), 8.61 (s, 1H), 7.86 (d, 1 H), 7.76 (d, 2H), 7.69 (d, 1 H), 7.51 (d, 2H), 7.18 (dd, 1H), 6.52 (s, 1H), 4.16 (t, 2H), 3.59 (t, 4H), 3.36 (overlapping, 4H), 2.72 (t, 2H), 1.30 (s, 9H). NMR (CDCI3) £9.3 (br, 1H), 7.84 (m, 4H), 7.59 (d, 2H), 7.49 (d, 1 H), 7.22 (d, 1 H), 7.03 (dd, 1 H)1 5.88 (s, 1 H), 4.16 (t, 2H), 3.76 (t, 4H), 2.84 (t, 2H), 2.61 (t, 4H), 1.37 (s, 9H).

[00432] F. For the preparation of the hydrochloride salt, N-(5-tert-butyl- isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2, 1 - b][1 ,3]benzothiazol-2-yl]phenyI}urea hydrochloride, the free base was dissolved in a mixture of 20 ml methylene chloride and 1 ml methanol. A solution of 1.0 M HCI in ethyl ether (1.1 eq.) was added dropwise, followed by addition of ethyl ether. The precipitate was collected by filtration or centrifugation and washed with ethyl ether to give the hydrochloride salt. Yield: 2.44 g (98 %) NMR (DMSO-d6) £11-0 (br, 1 H), 9.68 (s, 1H), 9.26 (s, 1H), 8.66 (s, 1 H), 7.93 (d, 1H), 7.78 (m, 3H), 7.53 (d, 2H), 7.26 (dd, 1H), 6.53 (S, 1 H), 4.50 (t, 2H), 3.97 (m, 2H), 3.81 (t, 2H), 3.6 (overlapping, 4H)13.23 (m, 2H)1 1.30 (s, 9H).

[00433] G. Alternatively, Compound B1 may be made by taking the intermediate from Example 4B and reacting it with chloroethyl morpholine hydrochloride under conditions described in Step C. [00434] H . Λ/-(5-tert-butyl-isoxazol-3-yl)-Λ/’-{4-[5-(2-morpholin-4-yl- ethoxy)imidazo[2,1-6][1 ,3]benzothiazol-2-yl]phenyl}urea hydrochloride, a compound having the general formula (I) where R1 is substituted on the 5 position of the tricyclic ring, was prepared in the manner described in Steps A- F but using the cyciization product 2-amino-benzothiazol-4-ol with 2-bromo-4′- nitroacetophenone in Step A. 1H NMR (DMSO-d6) δ 11.6 (br, 1 H)1 9.78 (br, 1H), 9.56 (br, 1 H), 8.64 (s, 1H)1 7.94 (d, 2H), 7.70 (s, 1H)1 7.56 (d, 2H), 7.45 (t, 1 H), 7.33 (d, 1H), 6.54 (s, 1 H), 4.79 (t, 2H), 3.87 (m, 6H), 3.60 (m, 2H), 3.34 (m, 2H)1 1.30 (s, 9H); LC-MS: ESI 561 (M+H)+. [Compound B11] [00435] I. N-(5-tert-butyl-isoxazol-3-yl)-N’-{4-[6-(2-morpholin-4-yl- ethoxy)imidazo[2,1-b][1 ,3]benzothiazol-2-yl]phenyl}urea hydrochloride [Compound B12] was also prepared by first preparing the benzothiazole starting material, 5 methoxy-benzothiazol-2yl~amine: [00436] To prepare the 5-methoxy-benzothiazol-2-ylamine starting material: To a suspension of (3-methoxy-phenyl)-thiourea (1.822g, 10 mmol) in CH2CI2 (20 ml_) at 0 0C was added dropwise a solution of bromine (1.76 g, 11 mmol) in 10 ml of trichloromethane over a period of thirty minutes. The reaction was stirred for 3 hours at room temperature then heated to 3 hours to reflux for one hour. The precipitate was filtered and washed with dichloromethane. The solid was suspended in saturated NaHCOsand extracted with CH2CI2. The extract was dried over MgSO4 and concentrated to give a white solid (1.716 g, 95%).

………………….

WO 2011056939

N-(5-ieri-butyl- isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l-&][l,3]benzothiazol-2- yl]phenyl}urea (I), or a pharmaceutically acceptable salt, solvate, hydrate, or polymorph thereof. N-(5-ieri-Butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l- / ][!, 3]benzo

Figure imgf000004_0001

N- (5-ieri-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l- &][l,3]benzo-thiazol-2-yl]phenyl}urea (I), or a pharmaceutically acceptable salt, solvate, hydrate, or polymorph thereof, comprising any one, two, three, four, five, six, seven of the steps of:

(A) converting 2-amino-6-alkoxybenzothiazole (II), wherein R1 is a suitable phenolic hydroxyl protecting ;

Figure imgf000037_0001

(II)                                                                             (III)

(B) reacting 2-amino-6-hydroxybenzothiazole (III) with compound (IV), wherein X is a leaving group, to yield 2-(4-nitrophenyl)imidazo[2,l-b]benzothiazol-7-ol (V);

Figure imgf000037_0002

(C) reacting 2-(4-nitrophenyl)imidazo[2,l-b]benzothiazol-7-ol (V) with compound (VI), wherein X2 is a leaving group, to yield 7-(2-morpholin-4-yl-ethoxy)-2-(4- nitrophenyl)imidazo[ -b]benzothiazole (VII);

Figure imgf000037_0003

 

Figure imgf000037_0004

(D) reducing 7-(2-morpholin-4-yl-ethoxy)-2-(4-nitrophenyl)imidazo[2, 1- bjbenzothiazole (VII) to yield 7-(2-morpholin-4-yl-ethoxy)-2-(4- am

Figure imgf000038_0001

(E) converting 3-amino-5-£er£-butyl isoxazole (IX) to a 5-?er?-butylisoxazol-3- ylcarbamate derivative (X), wherein R2 is optionally substituted aryl, heteroaryl, alkyl, or cycloalkyl;

Figure imgf000038_0002

(IX)                                                              (X)

(F) reacting 7-(2-morpholin-4-yl-ethoxy)-2-(4-aminophenyl)imidazo[2,l- bjbenzothiazole (VIII) with a 5-£er£-butylisoxazol-3-ylcarbamate derivative (X) to yield N-(5-ieri-butyl-isoxazol-3-yl)-N'-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l- &][l,3]benzo-

 

Figure imgf000038_0003

(G) converting N-(5-ieri-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl- ethoxy)imidazo[2,l-&][l,3]benzo-thiazol-2-yl]phenyl}urea to an acid addition salt of N- (5-ieri-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l- b] [ 1 ,3]benzo-thiazol-2-yl]phenyl } urea.

[00128] In certain embodiments, provided herein are processes for the preparation of N-(5-ieri-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l- &][l,3]benzo-thiazol-2-yl]phenyl}urea, or a pharmaceutically acceptable salt, solvate, hydrate, or polymorph thereof, as depicted in Scheme 1, wherein R1, R2, X1, and X2 are defined herein elsewhere. In specific embodiments, provided herein are processes for the preparation of N-(5-ieri-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl- ethoxy)imidazo[2,l-&][l,3]benzo-thiazol-2-yl]phenyl}urea (I), or a pharmaceutically acceptable salt, solvate, hydrate, or polymorph thereof, comprising any one, two, three, four, five, six, seven, of the Steps A, B, C, D, E, F, and G, as depicted in Scheme 1.

Scheme 1 :

 

Figure imgf000039_0001

A. Preparation of 2-amino-6-hydroxybenzothiazole

Figure imgf000070_0001

1. Example A-l[00252] To a 1-L 3-necked round bottom flask fitted with a condenser, heating mantle, and mechanical stirrer was charged aqueous hydrobromic acid (48%, 632 mL, 5.6 mol, 10 equiv). 2-Amino-6-methoxybenzothiazole (100 g, 0.55 mol, 1 equiv) was added to the above flask over 15 minutes. The reaction temperature was raised slowly to reflux (105-110 °C). A clear dark brown colored solution was observed at about 80 °C. The reflux was continued at 105-110 °C for about 4 hr. The progress of the reaction was monitored by HPLC. When 2-amino-6-methoxybenzothiazole was less than 2%, the reaction was substantially complete.

[00253] The reaction mass was cooled to 0-5 °C and at this point precipitation of a solid was observed. The mixture was maintained at 0-5 °C for 0.5 hr and filtered, and the cake was pressed to remove HBr. The wet cake was transferred to a 2-L round bottom flask fitted with a mechanical stirrer. Saturated aqueous sodium bicarbonate solution (-1500 mL) was added slowly at ambient temperature, whereupon considerable frothing was observed. The pH of the solution was found to be about 6.5 to 7. The mixture was stirred for 0.5 hr at ambient temperature and filtered. The filter cake was washed with water (500 mL), dried on the filter and then under vacuum at 30-35 °C for 10-12 hr to give the product 2-amino-6-hydroxybenzothiazole (82 g, 89% yield, HPLC purity = 99%). JH NMR (DMSO-if6, 500 MHz): δ 7.12 (d, 1H), 7.06 (S, 2H, NH2), 7.01 (d, 1H), 6.64 (dd, 1H); MS (m/z) = 167.1 [M+ + 1].

[00254] Table: Summary of Plant Batches

 

Figure imgf000071_0001

[00255] HPLC chromatographic conditions were as follows: The column used was XTerra RP8, 250 X 4.6 mm, 5μ or equivalent. Mobile Phase A was buffer, prepared by mixing 3.48 g of dipotassium hydrogen phosphate in 1.0 L of water, and adjusting the pH to 9.0 with phosphoric acid. Mobile Phase B was methanol. The flow rate was 1.0 mL/minute. Detection was set at UV 270 nm. The injection volume was 20 μΐ^, and the sample was diluted with a diluent (Mobile Phase A : Mobile Phase B = 70:30). Test solution was prepared by weighing accurately about 25 mg of sample and transferring it into a 100 mL volumetric flask, dissolving with 20-30 mL of diluent, making up the volume to the mark with diluent, and mixing. The HPLC was performed by separately injecting equal volumes of blank and test solution, and recording the chromatogram for all injections. The purity was calculated by area normalization method.

[00256] Table: HPLC Method

 

Figure imgf000072_0001

2. Example A-2

[00257] 2-Amino-6-methoxybenzothiazole was reacted with hot aqueous HBr at a temperature of >70 °C for about 3 hours and then the clear solution was cooled to ambient temperature overnight. The precipitated solids were collected, dissolved in hot water and the pH was adjusted to between 4.5-5.5. The resultant solids were collected, dried and re-crystallized from isopropanol. Second crop material was collected. The solids were vacuum dried to give 2-amino-6-hydroxybenzothiazole.

[00258] The reaction progress was monitored by thin layer chromatography (TLC). The product was isolated as a white solid, with 99.4% purity (HPLC area %). JH NMR (300 MHz, DMSO-if6) was collected, which conformed to structure.

3. Example A-3

[00259] A 22-L 3-neck round bottom flask was equipped with a mechanical agitator, thermocouple probe, a reflux condenser, and a heating mantle. The flask was charged with hydrobromic acid (14 L, 123.16 mol, 13.10 equiv). Heating was initiated and 2- amino-6-methoxybenzothiazole was added (1.7 kg, 9.4 mol, 1.00 equiv) over 10 minutes with stirring. The heating of the reaction mixture was continued to reflux, and maintained (>107 °C) for approximately 5 hours. The reaction mixture turned into a clear solution between 75 °C and 85 °C. The reaction progress was monitored by TLC until no starting material was observed (A -0.5 mL reaction mixture aliquot was diluted with -0.5 mL water as a clear solution, neutralized with sodium acetate to pH -5 and extracted with 1 mL dichloromethane. The organic layer was spotted: 5%

methanol/dichloromethane; Rf (product) = 0.35; Rf (starting material) = 0.40).

[00260] The reaction mixture was cooled to – 20 °C (overnight). White solids precipitated. The solids were filtered on a polypropylene filter and pressed to remove as much hydrobromic acid from the solids as possible to facilitate the subsequent pH adjustment step. The slightly wet crude product was dissolved in hot (50 °C) water (5 L). The clear solution was filtered to remove any insoluble material present, and the solids were washed with 50 °C water. The filtrate was cooled to 10 °C. The cooled filtrate was neutralized with sodium acetate (- 1.0 kg) to pH 4.5 to 5.5 with vigorous stirring. A thick white solid precipitated. The solids were collected by filtration, and washed with cool (-10 °C) water (2 x 1.0 L) and air dried.

[00261] The wet crude product was slurried in hot (50 °C) isopropanol (3 L) briefly and allowed to stand in a cool room (-5 °C) overnight. The solids were collected by filtration and washed with methyl ferf-butylether (2 x 500 mL). The solids were dried in a vacuum oven overnight (<30 mm Hg) at 30 °C (first crop). Yield: 1068 g (68%), white solid. HPLC: 99.4% (area). JH NMR (300 MHz, DMSO- ) conformed to structure.

[00262] The organic filtrate was collected in a total volume of 1.0 L, cooled to 10 °C. The off-white solids were precipitated and collected by filtration. The solids were dried in a vacuum oven overnight (<30 mm Hg) at 30 °C (second crop). Yield: 497 g (32%), off-white solid. HPLC: 99.8% (area).

[00263] The overall yield combining the first crop and the second crop was 1565 g, (99%).

B. Preparation of 2-(4-nitrophenyl)imidazo[2,l-b]benzothiazol-7-ol

 

Figure imgf000073_0001

1. Example B-l[00264] A 3-L 3-neck round bottom flask fitted with a condenser, a heating mantle, and a mechanical stirrer was charged with H-butanol (1.5 L), followed by 2-amino-6- hydroxybenzothiazole (75 g, 0.45 mol, 1.0 equiv), 2-bromo-4′-nitroacetophenone (121 g, 0.50 mol, 1.1 equiv), and sodium bicarbonate (41.6 g, 0.50 mol, 1.0 equiv). The reaction temperature was gradually raised to reflux and maintained at reflux (110-115 °C) for 2-3 hr. During the temperature increase, the reaction mass turned into a clear solution and then immediately changed into an orange colored suspension at 65-75 °C. The progress of the reaction was monitored by HPLC analysis every 1 hr (reaction mass sample was submitted to QC). When the level of 2-amino-6-hydroxybenzothiazole was less than 2%, the reaction was substantially complete.

[00265] The reaction mass was slowly cooled to 50-60 °C and then further cooled to 0-5 °C and stirred for 15 min. The precipitated solids were collected by filtration, and dried on the filter. The wet cake was transferred in to a 1-L round bottom flask, and water (600 mL) was added. The suspension was stirred for 0.5 hr and filtered, and the solid was dried on the filter. The wet cake was again taken in to a 1-L round bottom flask and stirred with acetone (200 mL). The slurry was filtered and washed with acetone (2 X 100 mL), and the solid was dried on the filter, unloaded and further dried in a vacuum oven at ambient temperature to give the product 2-(4-nitrophenyl)imidazo[2,l- b]benzothiazol-7-ol (V) (120 g, 85.7% yield, HPLC purity = 98.7%). JH NMR (DMSO- d6, 500 MHz): δ 9.96 (s, 1H, OH), 8.93 (s, 1H), 8.27 (d, 2H), 8.06 (d, 2H), 7.78 (d, 1H), 7.38 (d, 1H), 6.97 (dd, 1H); MS (m/z) = 312 [M+ + 1].

[00266] Table: Summary of Plant Batches

 

Figure imgf000074_0001

* Input of 2-amino-6-hydroxybenzothiazole (III)

[00267] HPLC chromatographic conditions were as follows: The column used was XTerra RP8, 250 X 4.6 mm, 5μ or equivalent. Mobile Phase A was buffer, prepared by mixing 3.48 g of dipotassium hydrogen phosphate in 1.0 L of water, and adjusting the H to 9.0 with phosphoric acid. Mobile Phase B was methanol. The flow rate was 1.0 mL/minute. Detection was set at UV 235 nm. The injection volume was 10 μΐ^. The blank was prepared by transferring 200 μΐ. of DMSO and 200 μΐ. of 2M NaOH into a 10 mL volumetric flask, making up the volume to the mark with methanol, and mixing. The test solution was prepared by weighing accurately about 10 mg of sample and transferring it into a 50 mL volumetric flask, dissolving with 1 mL of DMSO and 1 mL of 2M NaOH, sonicating to dissolve, making up the volume to the mark with methanol, and mixing. The HPLC was performed by separately injecting equal volumes of blank and test solution, and recording the chromatogram for all injections. The purity was calculated by area normalization method.

[00268] Table: HPLC Method

 

Figure imgf000075_0001

2. Example B-2

[00269] A 50-L 3-neck round bottom flask was equipped with a mechanical agitator, a thermocouple probe, a reflux condenser, and a heating mantle. The flask was charged with 2-amino-6-hydroxybenzothiazole (1068 g, 6.43 mol, 1.0 equiv) and ethanol (200 proof, 32.0 L), and the suspension was stirred for 10 minutes. 2-Bromo-4- nitroacetophenone (1667 g, 6.49 mol, 1.01 equiv) was added in one portion. The reaction mixture was heated to reflux (78 °C). The reflux was maintained for approximately 25 hours, resulting in a yellow suspension. The reaction progress was monitored by TLC (20% methanol/ethyl acetate; Rf (product) = 0.85; Rf (starting material) = 0.30). TLC indicated -50% 2-amino-6-hydroxybenzothiazole after 24 hours of reflux. Tetrabutylammonium iodide (10 g) was added and reflux was maintained for an additional 12 hours. TLC indicated -50% starting material still present. Coupling was seen to occur at both the thiazole and the amine.

[00270] The reaction mixture was cooled to 0-5 °C. The solids were collected by filtration, and the yellow solid was washed with ethanol (200 proof, 2 X 1.0 L) and diethyl ether (2 X 1.5 L). The solids were dried in a vacuum oven (<10 mm Hg) at 40 °C. Yield: 930 g (46%), yellow solid. HPLC: 99.5% (area). JH NMR (300 MHz, DMSO-i¾) conformed to structure.

3. Example B-3

[00271] The reaction of Step B was carried out on multiple runs, varying solvents, temperature, and base. The results were summarized in the table below. The product (V) was isolated as yellow or green solids, with 1H NMR consistent with the structure and a purity of greater than about 98% by HPLC analysis.

[00272] Table: Reaction Condition Screening

 

Figure imgf000076_0002

TBAI = Tetrabutylammonium Iodide

C. Preparation of 7-(2-morpholin-4-yl-ethoxy)-2-(4- nitrophenyl)imidazo[2, 1 -bjbenzothiazole

Figure imgf000076_0001

1. Example C-l

[00273] To a 2000-L glass-lined (GL) reactor was charged DMF (298 kg), and the agitator was started. Under a nitrogen blanket, the reactor was charged with 2-(4- nitrophenyl)imidazo[2,l-&]benzothiazol-7-ol (36.8 kg, 118.2 mol, 1.0 equiv), 4-(2- chloroethyl)morpholine hydrochloride (57.2-66.0 kg, 307.3-354.6 mol, 2.6-3.0 equiv), tetrabutylammonium iodide (8.7 kg, 23.6 mol, 0.2 equiv) and potassium carbonate (49.0 kg, 354.6 mol, 3.0 equiv). The resulting yellow suspension was heated and stirred at 90 + 5 °C for at least 15 minutes, then the temperature was increased to 110 + 5 °C. The mixture was stirred for at least 1 hour and then sampled. The reaction was deemed complete if 2-(4-nitrophenyl) imidazo[2,l-&]benzothiazol-7-ol was <1% by HPLC. If the reaction was not complete, the heating was continued and the reaction sampled. If the reaction was incomplete after 6 hours, additional 4-(2-chloroethyl)morpholine hydrochloride may be charged. In general, additional charges of 4-(2- chloroethyl)morpholine hydrochloride had not been necessary at the given scale.

[00274] The reactor was cooled to 20 + 5 °C and charged with water (247 kg) and acetone (492 kg). The mixture was agitated at 20 + 5 °C for at least 1 hour. The product (VII) was isolated by filtration or centrifuge, and washed with water and acetone, and then dried in a vacuum oven at 45 °C to constant weight to give a yellow solid (46.2 kg, 92% yield, HPLC purity = 97.4% by area). JH NMR (300 MHz, DMSO- ) conformed to structure.

2. Example C-2

[00275] 2-(4-Nitrophenyl)imidazo[2, l-b]benzothiazol-7-ol, 4-(2-chloroethyl)- morpholine hydrochloride, potassium carbonate, and tetrabutylammonium iodide were added to N,N-dimethylformamide forming a yellow suspension that was heated at a temperature of >50 °C for over 3 hours. The reaction was cooled and the solids were collected, slurried into water, filtered, slurried into acetone, filtered and washed with acetone to give yellow solids that were dried under vacuum to give 7-(2-morpholin-4-yl- ethoxy)-2-(4-nitrophenyl)imidazo[2,l-b]benzothiazole.

[00276] The reaction progress was monitored by thin layer chromatography (TLC). The product was isolated as a yellow solid, with 99% purity (HPLC area %), and a water content of 0.20%. Infrared (IR) spectrum was collected, which conformed to structure.

3. Example C-3

[00277] A 50-L 3-neck round bottom flask was equipped with a mechanical agitator, a thermocouple probe, a drying tube, a reflux condenser, and a heating mantle. The flask was charged with 2-(4-nitrophenyl)imidazo [2,l-&]benzothiazol-7-ol (1.770 kg, 5.69 mol, 1.0 equiv), N,N-dimethylformamide (18.0 L), 4-(2-chloroethyl)morpholine hydrochloride (2.751 kg, 14.78 mol, 2.6 equiv), potassium carbonate (2.360 kg, 17.10 mol, 3.0 equiv), and tetrabutylammonium iodide (0.130 kg, 0.36 mol, 0.06 equiv) with stirring. The resulting yellow suspension was heated to about 90 °C to 95 °C, maintaining the temperature for approximately 5 hours. The reaction was monitored by TLC until no starting material was observed (20% methanol / ethyl acetate; Rf (product) = 0.15; Rf (starting material) = 0.85).

[00278] The reaction mixture was cooled to -10 °C, and the yellow solids were collected by filtration on a polypropylene filter pad. The solids were slurried in water (2 X 5 L) and filtered. The crude wet product was slurried in acetone (5 L), filtered, and the solids were rinsed with acetone (2 X 1.5 L). The solids were dried in a vacuum oven (<10 mm Hg) at 45 °C. Yield: 2.300 kg (95%), yellow solid. TLC: R/ = 0.15 (20% methanol / EtOAc). HPLC: 95.7% (area). JH NMR (300 MHz, DMSO-i¾) conformed to the structure.

[00279] Table: Yields from multiple batch runs

 

Figure imgf000078_0001

4. Example C-4

[00280] To a reactor were added 2-(4-nitrophenyl)imidazo [2,l-&]benzothiazol-7-ol (1.0 kg), 4-(2-chloroethyl)morpholine hydrochloride (1.6 kg), tetrabutylammonium iodide (0.24 kg), and potassium carbonate (1.3 kg, anhydrous, extra fine, hydroscopic). N,N-Dimethylformamide (DMF) (8.6 L) was added into the reactor. The DMF used had water content of no more than 0.05% w/w. The mixture was stirred for between 15 and 30 minutes to render a homogeneous suspension, which was heated to between 85 °C and 95 °C and stirred at between 85 °C and 95 °C for 15 to 30 minutes. The mixture was then heated to between 105 °C and 120 °C and stirred at between 105 °C and 120 °C (e.g. , 115 °C) until the reaction was complete (as determined by HPLC to monitor the consumption of starting material). In some embodiments, if necessary (e.g. , if after 6 hours the reaction was not complete as indicated by HPLC analysis), additional 4-(2- chloroethyl)morpholine hydrochloride (0.03 kg) may be added and the reaction mixture stirred at between 105 °C and 120 °C (e.g. , 115 °C) until reaction completion.

[00281] The reaction mixture was cooled to between 20 °C and 30 °C (e.g. , over a period of 3 hours). To another reactor was added deionized water (7.6 L) and acetone (15 L). The mixture of water and acetone was then added into the reaction mixture while maintaining the temperature at between 20 °C and 30 °C. The mixture was then stirred for 1 to 2 hours at a temperature of between 20 °C and 30 °C. The mixture was filtered, and the solid was washed with deionized water (e.g. , about 45x deionized water) until pH of washes was below 8. The solid was then washed with acetone (9.7 L). The solid was dried under vacuum at a temperature of less than 50 °C until the water content by Karl-Fischer was less than 0.30% w/w and TGA curve showed a mass loss of less than 0.30% w/w at about 229 °C (sampling approximately every 6 hours). The desired product was obtained in about 89% yield having about 99% purity by HPLC.

5. Example C-5

[00282] To a reactor were added 2-(4-nitrophenyl)imidazo [2, l-&]benzothiazol-7-ol (1.0 kg), 4-(2-chloroethyl)morpholine hydrochloride (1.6 kg), and potassium carbonate (1.3 kg, anhydrous, extra fine, hydroscopic). N,N-Dimethylformamide (DMF) (8.6 L) was added into the reactor. The DMF used had water content of no more than 0.05% w/w. The mixture was stirred for between 15 and 30 minutes to render a homogeneous suspension, which was heated to between 95 °C and 120 °C (e.g. , between 100 °C and 105 °C) and stirred at between 95 °C and 120 °C (e.g. , 105 °C) until the reaction was complete (as determined by HPLC to monitor the consumption of starting material). In some embodiments, if necessary (e.g. , if after 6 hours the reaction was not complete as indicated by HPLC analysis), additional 4-(2-chloroethyl)morpholine hydrochloride (0.03 kg) and potassium carbonate (0.024 kg) may be added and the reaction mixture stirred at between 100 °C and 120 °C (e.g. , 105 °C) until reaction completion.

[00283] The reaction mixture was cooled to between 60 °C and 70 °C over a period of at least 60 minutes. Industrial water (6 L) was added to the reactor. The reaction mixture was cooled to between 20 °C and 30 °C. Acetone (6 L) was added to the reactor. The mixture was stirred for 1 to 2 hours at a temperature of between 20 °C and 30 °C. The mixture was filtered, and the solid was washed with industrial water (e.g. , about 45 x industrial water) until pH of washes was below 8. The solid was then washed with acetone (9.7 L). The solid was dried under vacuum at a temperature of less than 50 °C, until the water content by Karl-Fischer was less than 0.30% w/w and TGA curve showed a mass loss of less than 0.30% w/w at about 229 °C (sampling approximately every 6 hours).

6. Example C-6

[00284] To a reactor is added 2-(4-nitrophenyl)imidazo [2, l-&]benzothiazol-7-ol (1.0 kg), 4-(2-chloroethyl)morpholine hydrochloride (1.6 kg), and potassium carbonate (1.3 kg, anhydrous, extra fine, hydroscopic). N,N-Dimethylformamide (DMF) (8.6 L) is added into the reactor. The DMF has a water content of no more than 0.05% w/w. The mixture is stirred for between 15 and 30 minutes to render a homogeneous suspension, which is heated to between 95 °C and 120 °C (e.g. , between 100 °C and 105 °C) and stirred at between 95 °C and 120 °C (e.g. , 105 °C) until the reaction is complete (as determined by HPLC to monitor the consumption of starting material). In some embodiments, if necessary (e.g. , if after 6 hours the reaction is not complete as indicated by HPLC analysis), additional 4-(2-chloroethyl)morpholine hydrochloride (0.03 kg) and potassium carbonate (0.024 kg) may be added and the reaction mixture stirred at between 100 °C and 120 °C (e.g. , 105 °C) until reaction completion.

[00285] The reaction mixture is cooled to between 20 °C and 30 °C (e.g. , over a period of 3 hours). To another reactor is added deionized water (7.6 L) and acetone (15 L). The mixture of water and acetone is then added into the reaction mixture while maintaining the temperature at between 20 °C and 30 °C. The mixture is then stirred for 1 to 2 hours at a temperature of between 20 °C and 30 °C. The mixture is filtered, and the solid is washed with deionized water (e.g. , about 45x deionized water) until pH of washes is below 8. The solid is then washed with acetone (9.7 L). The solid is dried under vacuum at a temperature of less than 50 °C until the water content by Karl-Fischer is less than 0.30% w/w and TGA curve shows a mass loss of less than 0.30% w/w at about 229 °C (sampling approximately every 6 hours). D. Preparation of 7-(2-morpholin-4-yl-ethoxy)-2-(4- aminophenyl)imidazo [2, 1 -bjbenzothiazole

 

Figure imgf000081_0001

[00286] To a 200-L high pressure (HP) reactor was charged a slurry of 7-(2- morpholin-4-yl-ethoxy)-2-(4-nitrophenyl)imidazo [2,l-&]benzothiazole (VII) (7.50 kg, 17.7 mol, 1.0 equiv) in methanol (30 kg). The container was rinsed with additional methanol (10 kg) and the rinse was charged to the reactor. The reactor was then charged with THF (67 kg) and methanol (19 kg). The contents were agitated and the reactor was flushed with nitrogen by alternating nitrogen and vacuum. Vacuum was applied to the reactor and Raney Ni catalyst (1.65 kg, 0.18 wt. equiv) was charged through a sample line. Water (1 kg) was charged through the sample line to rinse the line. The reactor was flushed with nitrogen by alternating nitrogen and vacuum. The reactor was then vented and heated to 50 °C. The reactor was closed and pressurized with hydrogen gas to 15 psi keeping the internal temperature below 55 °C. The reactor was vented and re- pressurized a total of 5 times, then pressurized to 150 psi with hydrogen gas. The contents were agitated at 50 °C for at least 4 hours. At this point a hydrogen uptake test was applied: The reactor was re-pressurized to 150 psi and checked after 1 hour. If a pressure drop of more than 5 psi was observed, the process was repeated. Once the pressure drop remained < 5 psi, the reactor was vented and sampled. The reaction was deemed complete when 7-(2-morpholin-4-yl-ethoxy)-2-(4-nitrophenyl)imidazo [2,1- 6]benzothiazole (VII) was < 0.5% by HPLC.

[00287] The reactor was flushed with nitrogen as shown above. The 200-L HP reactor was connected to the 2000-L GL reactor passing through a bag filter and polish filter. The bag filter and polish filter were heated with steam. The 200-L HP reactor was pressurized (3 psi nitrogen) and its contents were filtered into the 2000-L reactor. The filtrates were hot. The 200-L reactor was vented and charged with THF (67 kg) and methanol (59 kg), the reactor agitated, and filtered into the 2000-L GL reactor.

[00288] A total of 6 reductions (46.2 kg processed) were carried out and the combined batches were concentrated by vacuum distillation (without exceeding an internal temperature of 40 °C) to a volume of -180 L. The reactor was cooled to 20 °C and charged with heptane (250 kg) and again vacuum distilled to a volume of -180 L. The reactor was charged with heptane (314 kg) and agitated at 20 °C for at least 1 hour, and then the product was isolated by centrifugation or collection on a Nutsche filter, washing with heptanes (2-5 kg per portion for centrifugation, followed by a 10-20 kg heptanes rinse of the reactor; or 94 kg for Nutsche filtration, rinsing the reactor first). The cake was blown dry, transferred to a vacuum oven and dried to constant weight maintaining a temperature < 50 °C to give the desired product (VIII) (34.45 kg, 80% yield, HPLC purity = 97.9%).

2. Example D-2

[00289] 7-(2-Morpholin-4-yl-ethoxy)-2-(4-nitrophenyl)imidazo[2,l-b]benzothiazole was dissolved into methanol and THF and placed in a hydrogenator. Raney nickel was added and the vessel was pressurized with hydrogen and stirred for >24 hours. The reaction mixture was concentrated to a thick paste and diluted with methyl ferf-butyl ether. The resulting solids were filtered and washed with methyl ferf-butyl ether and dried under vacuum to give 7-(2-morpholin-4-yl-ethoxy)-2-(4-aminophenyl) imidazo [2, 1 -bjbenzothiazole.

[00290] The reaction progress was monitored by thin layer chromatography (TLC). The product was isolated as a yellow solid, with 99% purity (HPLC area %). IR was collected, which conformed to structure.

3. Example D-3

[00291] Into a 5-gallon autoclave, 7-(2-morpholin-4-yl-ethoxy)-2-(4-nitrophenyl) imidazo[2,l-&]benzothiazole (580 g, 1.37 mol, 1.0 equiv), THF (7.5 L), methanol (7.5 L, AR) and -55 g of decanted Raney nickel catalyst were added. The reaction vessel was purged with nitrogen (3 X 50 psi) and hydrogen (2 X 50 psi), with stirring briefly under pressure and then venting. The autoclave was pressurized with hydrogen (150 psi). The mixture was stirred and the hydrogen pressure was maintained at 150 psi for over 24 hours with repressurization as necessary. The reaction progress was monitored by TLC (10% methanol / chloroform with 1 drop ammonium hydroxide; Rf (product) 0.20; Rf (SM) 0.80). The reaction was substantially complete when the TLC indicated no starting material present, typically after 24 hours of stirring at 150 psi. The hydrogenation was continued at 150 psi for a minimum of 4 hours or until completion if starting material was still present after the initial 4 hours.

[00292] The reaction mixture was filtered through a Buchner funnel equipped with a glass fiber filter topped with a paper filter. Unreacted starting material was removed together with the catalyst. The filtrate was concentrated to a total volume of 0.5 L, and the residue was triturated with methyl ferf-butyl ether (0.5 L). The resultant solids were collected by filtration, and washed with methyl ferf-butyl ether (0.3 L) (first crop).

[00293] The filtrate was concentrated to dryness and the residue was diluted with methyl ferf-butyl ether (2 L). The resultant solids were collected by filtration, washing with methyl ferf-butyl ether (0.5 L) (second crop).

[00294] The solids were dried in a vacuum oven (<10 mm Hg) at 25 °C. Yield: 510 g (95%), beige solid. TLC: R/ 0.2 (10% methanol / chloroform with one drop of ammonium hydroxide). HPLC: 99.0% (area). JH NMR (300 MHz, DMSO-i¾) conformed to the structure.

[00295] Table: Yields from multiple batch runs

 

Figure imgf000083_0001

4. Example D-4

[00296] The reaction of Step D was carried out in multiple runs under various conditions, such as, e.g. , varying catalyst loading, concentration of reactant, reaction temperature, and/or workup procedures. The results are summarized in the table below.

 

Figure imgf000083_0002

Description Run # l Run # 2 Run # 3 Run # 4 Run # 5Rxn Temp (°C) RT RT RT RT RT

Rxn Time (Hr) 24 hr 24 hr 24 hr 24 hr 24 hr

Filtered the Filtered the solution

Filtered the Filtered the Filtered the

solution through through celite. The solution through solution through solution through

celite, washed celite filter cake celite, celite, celite,

with THF, refluxed in THF concentrated, concentrated, concentrated,

concentrated, washed with hot solvent exchanged solvent exchanged solvent exchanged

Work Up solvent exchanged THF, concentrated, with heptane, with heptane, with heptane,

with heptane, solvent exchanged stirred the solids stirred the solids stirred the solids

stirred the solids with heptane, stirred and filtered and filtered and filtered

and filtered the solids and washed with washed with washed with

washed with filtered washed with heptane heptane heptane

heptane heptane

Produce (VIII) 1.9 g 3.88 g 1.11 g 2.6 g 4.4 g

Yield 88% 83.4% 56 94.6%

HPLC purity 95.6% 77.5% 91% 93.8%

 

Figure imgf000084_0001

5. Example D-5

[00297] To a pressure reactor under nitrogen atmosphere was added a slurry of Raney Nickel in water (0.22 kg) (e.g. about 0.14 kg dry catalyst in water) and the charging line was rinsed with deionized water (0.13 L). To another reactor (Reactor B) were added methanol (5.05 L) and 7-(2-morpholin-4-yl-ethoxy)-2-(4-nitrophenyl)imidazo [2, 1- &]benzothiazole (1.0 kg), and the mixture was stirred for between 15 and 30 minutes to render a homogenous suspension. The suspension was transferred to the pressure reactor. Reactor B was washed with methanol (4.88 L) and the wash was transferred to the pressure reactor. Tetrahydrofuran (10.1 L) was added to the pressure reactor.

Hydrogen was charged to the pressure reactor to a pressure of between 2.0 bar and 3.0 bar. The reactor was heated to a temperature of between 45 °C and 55 °C. Hydrogen was then charged to the pressure reactor to a pressure of between 6.0 bar and 7.0 bar. The mixture was stirred at a temperature of between 45 °C and 55 °C (e.g. , 50 °C), while maintaining the hydrogen pressure between 6.0 bar and 7.0 bar until reaction completion (as determined by HPLC to monitor the consumption of starting material).

[00298] The mixture was filtered while maintaining the temperature at between 35 °C and 50 °C. The pressure reactor and the filter were washed with a mixture of THF (10.1 L) and methanol (9.93 L) preheated to a temperature of between 45 °C and 55 °C (e.g. , 50 °C). The combined filtrate was concentrated to a volume of between 2.4 L and 2.8 L under vacuum at a temperature of no more than 40 °C, and a precipitate was formed. Methanol (7.5 L) was added. The slurry was cooled to a temperature of between 5 °C and -5 °C (e.g. , over 3 hours) and stirred for at least 1 hour (e.g. , for 3 hours) while maintaining the temperature at between 5 °C and -5 °C. The suspension was filtered. The solid was washed with methanol (2 X 1.2 L). The solid was then dried under vacuum at a temperature of less than 50 °C until the methanol and THF contents were each less than 3000 ppm as analyzed by GC (e.g. , less than 1500 ppm). The desired product was obtained in about 88.5% yield having about 99% purity by HPLC.

E. Preparation of phenyl 5-£er£-butylisoxazol-3-ylcarbamate

Figure imgf000085_0001

[00299] The jacket temperature of a 200-L glass-lined (GL) reactor was set to 20 °C. To the reactor was charged 5-ieri-butylisoxazole-3-amine (15.0 kg, 107.0 mol, 1.0 equiv), then K2C03 (19.5 kg, 141.2 mol, 1.3 equiv) and anhydrous THF (62 kg).

Agitation was started and then phenyl chloroformate (17.6 kg, 112.4 mol, 1.05 equiv) was charged. The charging line was rinsed with additional anhydrous THF (5 kg). The reaction was agitated at 20 + 5 °C for at least 3 hours then sampled. The reaction was deemed complete if 5-£er£-butylisoxazole-3-amine was < 2% by HPLC. If the reaction was not complete after 6 hours, additional K2CO3 and phenyl chloroformate may be added to drive the reaction to completion.

[00300] Once complete, the reaction was filtered (Nutsche). The filter was rinsed with THF (80 kg). The filtrate was vacuum distilled without exceeding an internal temperature of 40 °C until -50 L remained. Water (188 kg) and ethanol (45 L) were charged, and the mixture was agitated for at least 3 hours with a jacket temperature of 20 °C. The resulting solid was isolated by centrifugation or collection on a Nutsche filter, rinsed with water (2-5 kg for each centrifugation portion; 30 kg for Nutsche filtration) and blow-dried. The solid was then dried to constant weight in a vacuum oven (45 °C) to give the desired product (19.4 kg, 92% yield, HPLC purity = 97.4%). On an 800 g scale, 1559 g of the desired product (98% yield) was obtained with a 99.9% HPLC purity. JH NMR (DMSO-i¾) δ 11.17 (s, 1H); 7.4 (m, 2H); 7.2 (m, 3H); 1.2 (s, 9H). LCMS (M+H)+ 261.

F. Preparation of N-(5-ieri-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl- ethoxy)imidazo[2, 1 -b] [ 1 ,3 ]benzothiazol-2-yl]phenyl } urea

 

Figure imgf000086_0001

1. Example F-l

[00301] The jacket of a 2000-L GL reactor was set to 20 °C and the reactor was charged with 7-(2-morpholin-4-yl-ethoxy)-2-(4-aminophenyl)imidazo[2,l- &]benzothiazole (26.7 kg, 67.8 mol, 1.0 equiv), 3-amino-5-?-butylisoxazole phenyl carbamate (19.4 kg, 74.5 mol, 1.1 equiv), DMAP (0.5 kg, 4.4 mol, 0.06 equiv), and DCM (anhydrous, 260 kg). Agitation was started, triethylamine (1.0 kg, 10.2 mol, 0.15 equiv) was charged followed by additional DCM (5 kg) through the charging line. The reaction was heated to reflux (-40 °C) and agitated for at least 20 hours with complete dissolution observed followed by product crystallizing from solution after -30 minutes. The reaction was sampled and deemed complete when HPLC analysis showed a ratio of compound (VIII) to compound (I) < 1%.

[00302] The reactor was cooled to 0 °C and stirred for at least 2 hours. The content of the reactor were isolated by centrifuge. Each portion was rinsed with 2-3 kg of cold (0 °C) DCM and spun dry for at least 5 minutes with a 10 psi nitrogen purge. For the final portion, the reactor was rinsed with 10 kg of cold (0 °C) DCM and transferred to the centrifuge where it was spun dry for at least 5 minutes with a 10 psi nitrogen purge. The combined filter cakes were transferred to a vacuum tray dryer and dried to constant weight at 50 °C and at least >20 inches of Hg to give the desired product (I) (35.05 kg, 92% yield, HPLC purity = 98.8%). Phenol was the major impurity detected (0.99%); and three other impurities (<0.10%) were detected. JH NMR (300 MHz, DMSO- ) conformed to structure.

2. Example F-2

[00303] A variety of solvents were used in the reaction of Step F to optimize for better yields and purity profiles. The contents of the symmetrical urea impurity (XI) were compared and summarized in the table below:

 

Figure imgf000087_0001

http://www.google.com/patents/WO2011056939A1?cl=en   SE THIS FOR DELETED CLIPS

 

Figure imgf000090_0001

 

Figure imgf000090_0002

4. Example F-4

[00305] To Reactor A under a nitrogen atmosphere was added 7-(2-morpholin-4-yl- ethoxy)-2-(4-aminophenyl)imidazo[2,l-&]benzothiazole (1 kg) and DMAP (0.02 kg). To Reactor B under a nitrogen atmosphere was added 3-amino-5-?-butylisoxazole phenyl carbamate (0.73 kg) and DCM (5.6 L). The DCM used had a water content of less than 0.05 % w/w. The mixture in Reactor B was stirred until dissolution. The solution was transferred into Reactor A (the solution can be filtered into Reactor A to remove any insoluble impurities in the carbamate starting material), and the mixture was stirred in Reactor A. Reactor B was washed with DCM (0.8 L) and the wash was transferred into Reactor A. Reactor A was washed with DCM (0.9 L). To Reactor A was added triethylamine (0.1 L) and the charging vessel and lines were rinsed with DCM (0.1 L) into Reactor A. The mixture was then heated to reflux and stirred at reflux until reaction completion (as determined by HPLC to monitor the consumption of starting material).

[00306] The reaction mixture was cooled (e.g. , over 2 to 3 hours) to a temperature of between -5 °C and 5 °C (e.g. , 0 °C). The mixture was then stirred for 2 to 3 hours at a temperature of between -5 °C and 5 °C (e.g. , 0 °C). The suspension was filtered. The solid was washed with cool DCM (2 X 1.5 L) (pre-cooled to a temperature of between -5 °C and 5 °C). The solid was dried under vacuum at a temperature of less than 45 °C until the DCM content was less than 1000 ppm (e.g., below 600 ppm) as analyzed by GC. The desired product was obtained having about 99% purity by HPLC.

G. Preparation of N-(5-ieri-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl- ethoxy)imidazo[2, l-b] [1 ,3]benzothiazol-2-yl]phenyl }urea dihydrochloride

 

Figure imgf000091_0001

1. Example G-l

[00307] The jacket of a 2000-L GL reactor was set to 20 °C and the reactor was charged with N-(5-ieri-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo [2, 1-&][1, 3]benzothiazol-2-yl]phenyl}urea (35.0 kg, 62.4 mol, 1.0 equiv) followed by methanol (553 kg). Agitation was started and the reaction mixture was heated to reflux (-65 °C). Concentrated aqueous HC1 (15.4 kg, 156.0 mol, 2.5 equiv) was charged rapidly (<5 minutes) and the charge line was rinsed into the reactor with methanol (12 kg). Addition of less than 2.0 equivalents of HC1 normally resulted in the formation of an insoluble solid. The reaction mixture was heated at reflux for at least 1 hour. Upon HC1 addition, the slurry dissolved and almost immediately the salt started to crystallize, leaving insufficient time for a polish filtration.

[00308] The reactor was cooled to 20 °C and the product was isolated by filtration (Nutsche) rinsing the reactor and then the cake with methanol (58 kg). The solid was then dried in a vacuum oven (50 °C) to constant weight to give the desired

dihydrochloride salt (35 kg, 89% yield, HPLC purity = 99.94%). JH NMR (300 MHz, DMSO-i¾) conformed to structure.

2. Example G-2

[00309] Concentrated HC1 was added to a suspension of N-(5-ieri-butyl-isoxazol-3- yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l-&][l,3]benzothiazol-2- yl]phenyl}urea in warm methanol forming a solution that slowly began to precipitate. The reaction mixture was refluxed for over 2 hours and then stirred overnight at ambient temperature. The dihydrochloride salt was collected and dried under vacuum.

3. Example G-3

[00310] A 50-L 3-neck round bottom flask was equipped with a mechanical agitator, a thermocouple probe, a nitrogen inlet, a drying tube, a reflux condenser, an addition funnel, and a heating mantle. The flask was charged with N-(5-ieri-butyl-isoxazol-3-yl)- N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l-&][l,3]benzothiazol-2-yl]phenyl}urea (775 g, 1.38 mol, 1.0 equiv) and MeOH (40 L, AR). The resulting off-white suspension was heated to reflux (68 °C). A clear solution did not form. HC1 (37% aqueous) (228 mL, 3.46 mol, 2.5 equiv) was added over 5 minutes at 68 °C. The reaction mixture turned into a clear solution and then a new precipitate formed within approximately 3 minutes. Heating was continued at reflux for approximately 5 hours. The reaction mixture was allowed to cool to ambient temperature overnight. The off-white solids were collected by filtration on a polypropylene filter, washing with MeOH (2 X 1 L, AR). [00311] Two lots of material prepared in this manner were combined (740 g and 820 g). The combined solids were slurried in methanol (30 L) over 30 minutes at reflux and allowed to cool to the room temperature. The solids were collected by filtration on a polypropylene filter, rinsing with methanol (2 X 1.5 L). The solids were dried in a vacuum oven (<10 mm Hg) at 40 °C. Yield: 1598 g (84%), off-white solid. HPLC: 98.2% (area). MS: 561.2 (M+l)+JH NMR (300 MHz, DMSO-i¾) conformed to the structure. Elemental Analysis (EA): Theory, 54.97 %C; 5.41 %H; 13.26 %N; 5.06 %S; 11.19 %C1; Actual, 54.45 %C; 5.46 %H; 13.09 %N; 4.99 %S; 10.91 %C1.

4. Example G-4

[00312] Into a 50-L 3-neck round bottom flask equipped with a mechanical stirrer, a heating mantle, a condenser and a nitrogen inlet, were charged N-(5-ieri-butyl-isoxazol- 3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l-&][l,3]benzothiazol-2- yl]phenyl}urea (1052.4 g, 1.877 mol, 1.0 equiv) and methanol (21 L). The reactor was heated and stirred. At an internal temperature of > 50 °C, cone. HC1 (398.63 mL, 4.693 mol, 2.5 equiv) was charged over 5 minutes through an addition funnel. During the addition, the mixture changed from a pale yellow suspension to a white suspension. The internal temperature was 55 °C at the conclusion of the addition. The mixture was heated to reflux for 1 hour, then heating was discontinued and the mixture was allowed to cool to room temperature. The mixture was filtered in two portions, and each filter cake was washed with methanol (2 X 1 L), transferred to trays and dried in a vacuum oven (45 °C) to constant weight. The dried trays were combined to produce 1141.9 g of the salt (96% yield, 99.1 % HPLC purity, 10.9% chloride by titration).

 

H. Analytical Data

1. N-(5-ieri-butyl-isoxazol-3-yl)-N’-{ 4-[7-(2-morpholin-4-yl- ethoxy)imidazo[2, l-&] [l ,3]benzothiazol-2-yl]phenyl}urea

dihydrochloride

[00314] A batch of about 30 grams of N-(5-ieri-butyl-isoxazol-3-yl)-N’- {4-[7-(2- morpholin-4-yl-ethoxy)imidazo[2, l-&] [l ,3]benzothiazol-2-yl]phenyl}urea

dihydrochloride was prepared using the methods described herein. This lot was

prepared in accordance with the requirements for production of clinical Active

Pharmaceutical Ingredients (APIs) under GMP conditions. The analytical data for this batch was obtained, and representative data were provided herein. [00315] Summary of analytical data for the dihydrochloride salt.

 

Figure imgf000095_0001

………………………

http://www.google.com/patents/WO2009038757A2?cl=en

 EXAMPLE 1. SYNTHESIS OF N-(5-TERT-BUTYL-ISOXAZOL-3-YU- N>-{4-f7-(2-MORPHOLIN-4- YL-ETHOXY)IMID AZO[2,1- BlH,31BENZOTHIAZOL-2-YL|PHENYLiUREA ("COMPOUND Bl")

[00357] A. The intermediate 2-amino-l,3-benzothiazol-6-ol was prepared according to a slightly modified literature procedure by Lau and Gompf: J. Org. Chem. 1970, 35, 4103- 4108. To a stirred solution of thiourea (7.6 g, 0.10 mol) in a mixture of 200 mL ethanol and 9 mL concentrated hydrochloric acid was added a solution of 1 ,4-benzoquinone (21.6 g, 0.20 mol) in 400 mL of hot ethanol. The reaction was stirred for 24 hours at room temperature and then concentrated to dryness. The residue was triturated with hot acetonitrile and the resulting solid was filtered and dried.

[00358] The free base was obtained by dissolving the hydrochloride salt in water, neutralizing with sodium acetate, and collecting the solid by filtration. The product (2- amino-l,3-benzothiazol-6-ol) was obtained as a dark solid that was pure by LCMS (M+H = 167) and NMR. Yield: 13.0 g (78 %). NMR (DMSO-^) Sl.6 (m, 2H), 6.6 (d, IH). [00359] B. To prepare the 2-(4-nitrophenyl)imidazo[2,l-b][l,3]benzothiazol-7-ol intermediate, 2-amino-l,3-benzothiazol-6-ol (20.0 g, 0.12 mol) and 2-bromo-4′- nitroacetophenone (29.3 g, 0.12 mol) were dissolved in 600 mL ethanol and heated to reflux overnight. The solution was then cooled to O0C in an ice-water bath and the product was collected by vacuum filtration. After drying under vacuum with P2O5 , the intermediate (2- (4-nitrophenyl)imidazo[2,l-£][l,3]benzothiazol-7-ol) was isolated as a yellow solid. Yield: 17.0 g (46 %) NMR (DMSO-(I6) δ 10 (s, IH), 8.9 (s, IH), 8.3 (d, 2H), 8.1 (d, 2H), 7.8 (d, IH), 7.4 (s, IH), 6.9 (d, IH).

[00360] C. To make the 7-(2-morpholin-4-yl-ethoxy)-2-(4-nitro-phenyl)imidazo[2,l-

6][l,3]benzothiazole intermediate: 2-(4-nitrophenyl)imidazo[2,l-6][l,3]benzothiazol-7-ol,

NYI-4144519vl 84 (3.00 g, 9.6 mmol) was suspended in 100 mL dry DMF. To this mixture was added potassium carbonate (4.15 g, 30 mmol, 3 eq), chloroethyl morpholine hydrochloride (4.65 g, 25 mmol, 2.5 eq) and optionally tetrabutyl ammonium iodide (7.39 g, 2 mmol). The suspension was then heated to 900C for 5 hours or until complete by LCMS. The mixture was cooled to room temperature, poured into 800 mL water, and allowed to stand for 1 hour. The resulting precipitate was collected by vacuum filtration and dried under vacuum. The intermediate, (7-(2-morpholin-4-yl-ethoxy)-2-(4-nitro-phenyl)imidazo[2, 1 - b][\, 3]benzothiazole) was carried on without further purification. Yield: 3.87 g (95 %) NMR (DMSO-d6) δ 8.97 (s, IH), 8.30 (d, 2H), 8.0 (d, 2H), 7.9 (d, IH), 7.7 (s, IH), 7.2 (d, IH), 4.1 (t, 2H), 5.6 (m, 4H), 2.7 (t, 2H).

[00361] D. To make the intermediate 7-(2-morpholin-4-yl-ethoxy)-2-(4-amino- phenyl)imidazo[2,l-b][l,3]benzothiazole: To a suspension of 7-(2-morpholin-4-yl-ethoxy)- 2-(4-nitro-phenyl)imidazo[2,l -b][\ , 3]benzothiazole (3.87g, 9.1 mmol) in 100 mL isopropyl alcohol/water (3:1) was added ammonium chloride (2.00 g, 36.4 mmol) and iron powder (5.04 g, 90.1 mmol). The suspension was heated to reflux overnight with vigorous stirring, completion of the reaction was confirmed by LCMS. The mixture was filtered through Celite, and the filtercake was washed with hot isopropyl alcohol (150 mL). The filtrate was concentrated to approximately 1/3 of the original volume, poured into saturated sodium bicarbonate, and extracted 3 times with dichloromethane. The combined organic phases were dried over MgSO4 and concentrated to give the product as an orange solid containing a small amount (4-6 %) of starting material. (Yield: 2.75 g 54 %). 80% ethanol/water may be used in the place of isopropyl alcohol /water – in which case the reaction is virtually complete after 3.5 hours and only traces of starting material are observed in the product obtained. NMR (DMSO-Λfc) δ 8.4 (s, IH), 7.8 (d, IH), 7.65 (d, IH), 7.5 (d, 2H), 7.1 (d, IH), 6.6 (d, 2H), 4.1 (t, 2H), 3.6 (m, 4H), 2.7 (t, 2H).

[00362] E. A suspension of 7-(2-morpholin-4-yl-ethoxy)-2-(4-amino- phenyl)imidazo[2,l-b][l,3]benzothiazole (4.06 g, 10.3 mmol) and 5-tert-butylisoxazole-3- isocyanate (1.994 g, 12 mmol) in toluene was heated at 120 0C overnight. The reaction was quenched by pouring into a mixture of methylene chloride and water containing a little methanol and neutralized with saturated aqueous NaHCO3 solution. The aqueous phase was extracted twice with methylene chloride, the combined organic extracts were dried over

NYI-4144519vl 85 MgSO4 and filtered. The filtrate was concentrated to about 20 ml volume and ethyl ether was added resulting in the formation of a solid. The precipitate was collected by filtration, washed with ethyl ether, and dried under vacuum to give the free base of Compound B 1. Yield: 2.342 g (41 %) NMR (DMSO-J6) £9.6 (br, IH), 8.9 (br, IH), 8.61 (s, IH), 7.86 (d, IH), 7.76 (d, 2H), 7.69 (d, IH), 7.51 (d, 2H), 7.18 (dd, IH), 6.52 (s, IH), 4.16 (t, 2H), 3.59 (t, 4H), 3.36 (overlapping, 4H), 2.72 (t, 2H), 1.30 (s, 9H). NMR (CDCl3) £9.3 (br, IH), 7.84 (m, 4H), 7.59 (d, 2H), 7.49 (d, IH), 7.22 (d, IH), 7.03 (dd, IH), 5.88 (s, IH), 4.16 (t, 2H), 3.76 (t, 4H), 2.84 (t, 2H), 2.61 (t, 4H), 1.37 (s, 9H).

6.2 EXAMPLE 2. ALTERNATIVE SYNTHESIS QF N-(5-TERT-BUTYL- ISOXAZQL-3- YL)-N -{4-[7-q-MORPHOLIN-4- YL- ETHOXYUMID AZOf2,l-BUl,31BENZOTHIAZOL-2- YLIPHENYLIUREA ("COMPOUND Bl")

[00363] A. To a suspension of the intermediate 2-(4-Nitrophenyl)imidazo[2,l- b][l,3]benzothiazol-7-ol from Example IB (2.24 g, 7.2 mmol) in ethanol (40 mL) was added SnCl2 1H2O (7.9Og, 35 mmol) and heated to reflux. Concentrated HCl was added to the reaction mixture and the precipitate formed gradually. The reaction mixture was heated to reflux for 20 hours and then allowed to cool to room temperature. The solution was poured into ice and neutralized with 10% NaOH and adjusted to approximately pH 6. The organic phase was extracted three times with ethyl acetate (80 mL x 3). Extracts were dried over MgSθ4 and concentrated to give a yellow solid. (1.621 g, 80%). The solid was recrystallized from methanol to give a pure product (1.355 g, 67%).

[00364] B. To a suspension of the intermediate from Step 2A (0.563 g, 2 mmol) in toluene (30 mL) was added 5-tert-butylisoxazole-3-isocyanate (0.332g, 2 mmol) and heated to reflux overnight. LC-MS analysis showed presence of the intermediate but no trace of 5- tert-butylisoxazole-3-isocyanate and an additional 0.166 g of the isocyanate was added. The reaction was again heated to reflux overnight. Completion of reaction was verified by LC- MS. The solvent was removed and the resulting mixture was dissolved in methanol which was removed to give the second intermediate as a solid.

[00365] The mixture was dissolved in CH2Cl2 (150 mL) and washed with saturated

NaHCO3. The organic layer was dried over MgSO4, concentrated, and purified by silica gel chromatography three times, first using a methanol/CH2Cl2 gradient, the second time using a

NYI-4144519vl 86 hexane/ethyl acetate gradient followed by a methanol/ethyl acetate gradient, and a third time using a methanol/CH2Cl2 gradient.

[00366] C. To a suspension of the intermediate from Step 2B (0.1 10 g, 0.25 mmol) in

THF (5mL) was added Ph3P (0.079g, 0.3 mmol), diisopropylazodicarboxylate (0.06 Ig, 0.3 mmol) and 4-morpholinoethanol (0.039 g, 0.3 mmol). The reaction mixture was stirred at room temperature overnight. Completion of the reaction was verified by LC-MS. The solvent was removed and the final product was purified using silica gel chromatography, with methanol in CH2Cl2 (0.030g, 21%).

6.3 EXAMPLE 3. BULK SYNTHESIS OF N-(5-TERT-BUTYL- ISOXAZOL-3-YL)-N’-f4-[7-(2-MORPHOLIN-4-YL- ETHOXY^IMID AZO[2α-BUlJlBENZOTHIAZOL-2- YLlPHENYLiUREA ("COMPOUND Bl")

[00367] A multi-step reaction scheme that was used to prepare bulk quantities of

Compound Bl is depicted in FIG. 66a and FIG. 66b, and is described further below. [00368] Step 1 : Preparation of 2- Amino-6-hydroxybenzothiazole (Intermediate 1). 2-

Amino-6-methoxybenzothiazole is reacted with hot aqueous HBr for about 3 hrs and then the clear solution is cooled to ambient temperature overnight. The precipitated solids are collected, dissolved in hot water and the pH is adjusted to between 4.5-5.5. The resultant solids are collected, dried and recrystallized from Isopropanol. Second crop material is collected. The solids are vacuum dried to give Intermediate 1.

[00369] Step 2: Preparation of 2-(4-Nitrophenyl) imidazo [2J-b]benzothiazol-7-ol

(Intermediate 2). 2-Amino-6-hydroxybenzothiazole, 2-Bromo-4-nitroacetophenone and absolute Ethanol are added together and heated to reflux for approximately 24 hours. Tetrabutylammonium iodide is added and the reaction is refluxed an additional 12 hours. The resulting yellow suspension is cooled and the solids collected and washed with Ethanol and Diethyl ether. The solids are dried under vacuum to give Intermediate 2. [00370] Step 3: Preparation of 7-(2-Morpholin-4-yl-ethoxy)-2-(4-nitrophenyl) imidazo

[2,1-b] benzothiazole (Intermediate 3). Intermediate 2, 4-(2-Chloroethyl)morpholine hydrochloride, Potassium carbonate and Tetrabutylammonium iodide are added to N,N- Dimethylformamide forming a yellow suspension that is heated for over 3 hours. The reaction is cooled and the solids are collected, slurried into water, filtered, slurried into

NYl-4 l4451′)v l 87 acetone, filtered and washed with Acetone to give yellow solids that are dried under vacuum to give Intermediate 3.

[O0371] Step 4: Preparation of 7-(2-Moφholin-4-yl-ethoxy)-2-(4-aminophenyl) imidazo [2,1 -b] benzothiazole (Intermediate 4). Intermediate 3 is dissolved into Methanol and THF and placed in a Hydrogenator. Raney Nickel is added and the vessel is pressurized with Hydrogen and stirred for >24 hrs. The reaction mixture is concentrated to a thick paste and diluted with Methyl tert-butyl ether. The resulting solids are filtered and washed with Methyl tert-butyl ether and dried under vacuum to give Intermediate 4. [O0372] Step 5: Preparation of {[5-(tert-Butyl) isoxazol-3-vnatnino}-N-{4-r7-(2- morpholin-4-yl-ethoxy)(4-hvdroimidazolo[2J-blbenzothiazol-2-yl)]phenyl|carboxamide (Compound Bl). 3 -Amino- 5 -tert-butyl isoxazole in Methylene chloride is added to a vessel containing toluene which is cooled to approx 0 0C. Triphosgene is then added and the reaction mixture is cooled to below -15 0C. Triethylamine is added, followed by Intermediate 4. The mixture is heated to distill off the Methylene chloride and then heated to over 60 0C for over 12 hours and cooled to 50-60 °C. The resulting solids are filtered, washed with Heptane, slurried with 4% sodium hydroxide solution, and filtered. The solids are then washed with Methyl tert-butyl ether followed by Acetone and dried under vacuum to give Compound Bl.

6.4 EXAMPLE 4. EXAMPLES OF PREPARATION OF COMPOUND Bl HCL SALT

[00373] Example A: For the preparation of a hydrochloride salt of Compound Bl5 N-

(5-tert-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,l- b][l,3]benzothiazol-2-yl]phenyl}urea hydrochloride, the free base was dissolved in a mixture of 20 ml methylene chloride and 1 ml methanol. A solution of 1.0 M HCl in ethyl ether (1.1 eq.) was added dropwise, followed by addition of ethyl ether. The precipitate was collected by filtration or centrirugation and washed with ethyl ether to give a hydrochloride salt of Compound Bl. Yield: 2.44 g (98 %) NMR (DMSO-^) S X 1.0 (br, IH), 9.68 (s, IH), 9.26 (s, IH), 8.66 (s, IH), 7.93 (d, IH), 7.78 (m, 3H), 7.53 (d, 2H), 7.26 (dd, IH), 6.53 (s, IH), 4.50 (t, 2H), 3.97 (m, 2H), 3.81 (t, 2H), 3.6 (overlapping, 4H), 3.23 (m, 2H), 1.30 (s, 9H). [00374] Example B: Concentrated HCl is added to a suspension of Compound Bl in warm methanol forming a solution that slowly begins to precipitate. The reaction mixture is

NYI-4144519vl 88 refluxed for over 2 hrs and then stirred overnight at ambient temperature. The HCl salt is collected and dried under vacuum.

[00375] Example C: Materials: {[5-(tert-Butyl) isoxazol-3-yl]amino}-N-{4-[7-(2- morpholin-4-yl-ethoxy)(4-hydroimidazolo[2,l-6]benzothiazol-2-yl)] phenyl }carboxamide (775 g, 1.38 mol, 1.0 eq); HCl 37% aqueous (288 mL, 3.46 mol, 2.5 eq); Methanol (MeOH, AR) (40L). Procedure: (Step 1) Equipped a 5OL 3-neck round bottom flask with a mechanical agitator, thermocouple probe, Nitrogen inlet, drying tube, reflux condenser, addition funnel and in a heating mantle. (Step 2) Charged the flask with {[5-(tert-Butyl) isoxazol-3-yl] amino}-N-{4-[7-(2-morpholin-4-yl-ethoxy)(4-hydroimidazolo[2,l- b]benzothiazol-2-yl)] phenyl jcarboxamide (775g) and MeOH, AR (40L). Heat the resulting off-white suspension to reflux (680C). A clear solution did not form. (Step 3) Added HCl (37% aqueous) (228 mL) over 5 minutes at 68°C. The reaction mixture turned into a clear solution and then a new precipitate formed within approximately 3 minutes. Continued heating at reflux for approximately 5 hours. Allowed the reaction mixture to cool to ambient temperature overnight. (Step 4) Collected the off-white solids by filtration onto a polypropylene filter, washing the solids with MeOH, AR (2 x 1 L). (Step 5) Combined two lots of material prepared in this manner (74Og and 82Og). Slurried the combined solids in Methanol (30 L) over 30 minutes at reflux and cool to the room temperature. (Step 6) Collected the solids by filtration onto a polypropylene filter, rinsing with Methanol (2 x 1.5L). (Step 7) Dried the solids in a vacuum oven (<10mniHg) at 400C. Yield: 1598 g (84%), off-white solid; HPLC: 98.2% (area); MS: 561.2 (M+l); IH NMR: conforms (300 MHz, DMSO-d6); Elemental Analysis (EA): Theory = 54.97 %C; 5.41 %H; 13.26 %N; 5.06 %S; 11.19 %C1; Actual = 54.45 %C; 5.46 %H; 13.09 %N; 4.99 %S; 10.91 %C1.

NYl-4I44519v! 89 [00376] Examples of Compound Bl HCl salt synthesis

 

Figure imgf000091_0001

[00377] Example D: In a 50-L 3-neck round bottom flask equipped with a mechanical stirrer, heating mantle, condenser and nitrogen inlet was charged Compound Bl (1052.4 g, 1.877 mol, 1.00 equiv.) and methanol (21 L). The reactor was heated and stirred. At an internal temperature > 50 0C, cone. HCl (398.63 mL, 4.693 mol, 2.5 equiv.) was charged over 5 minutes through an addition funnel. With the addition, the reaction changed from a pale yellow suspension to a white suspension. The internal temperature was 55 0C at the conclusion of the addition. The reaction was heated to reflux for 1 hour, then heating discontinued and the reaction allowed to cool to room temperature. The reaction was filtered in two portions, each filter cake washed with methanol (2 x 1 L), transferred to trays and dried in a vacuum oven (45 0C) to constant weight. The dried trays were combined to produce 1141.9 g, 96% yield, 99.1 % HPLC purity, 10.9% chloride by titration.

Solid Forms Comprising the HCl Salt of Compound Bl 6.6.2.1 Preparation of Solid Forms

 

Figure imgf000103_0001

6.6.2.2 Cold Precipitation Experiments

 

Figure imgf000103_0002

NYl-4144519vl 102 6.6.2.3 Slurry Experiments

 

Figure imgf000104_0001

NYI-41445 l9vl 103 6.6.2.4 Additional Preparation of Solid Forms Comprising the HCI Salt of Compound Bl

Figure imgf000105_0001

NYl-4144519v l 104

Figure imgf000106_0001

NYM 144519vl 105

Figure imgf000107_0001

N Y l -4 1 4 4 5 1 9 v l 1 0 6

Figure imgf000108_0001

NYI-4I44519vi 107

Figure imgf000109_0001

N V I 4 1 4 4 5 1 9 1 0 8

Figure imgf000110_0001

“Abbreviations in Table: CC = crash cool, CP = crash precipitation, EtOAc = ethyl acetate, FE = fast evaporation, VD = vapor diffusion, IPA = isopropanol, MEK = methyl ethyl ketone (2-butanone), RE = rotary evaporation, RT = room (ambient) temperature, SC = slow cool, SE = slow evaporation, THF = tetrahydrofuran, TFE = 2,2,2=trifluoroethanol.

6.6.2.5 Scale-up Experiments of Involving Crystal Forms Comprising the HCl Salt of Compound Bl

 

Figure imgf000110_0002

NYI-4144519v l 109

Figure imgf000111_0001

Abbreviations in Table: CC = crash cool, CP = crash precipitation, EtOAc = ethyl acetate, FE = fast evaporation, IPA = isopropanol, MEK = methyl ethyl ketone (2-butanone), RE = rotary evaporation, RT = room (ambient) temperature, SC = slow cool, SE = slow evaporation, THF = tetrahydrofuran, TFE = 2,2,2=trifluoroethanol.

 

……………………

Identification of N-(5-tert-butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,1-b][1,3]benzothiazol-2-yl]phenyl}urea dihydrochloride (AC220), a uniquely potent, selective, and efficacious FMS-like tyrosine kinase-3 (FLT3) inhibitor
J Med Chem 2009, 52(23): 7808

http://pubs.acs.org/doi/full/10.1021/jm9007533

Abstract Image

N-(5-tert-Butyl-isoxazol-3-yl)-N′-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,1-b][1,3]benzothiazol-2-yl]phenyl}urea Dihydrochloride (7): General Procedure D
A suspension of 2-(4-aminophenyl)-7-(2-morpholin-4-yl-ethoxy)imidazo[2,1-b][1,3]benzothiazole (19c) (4.06 g, 10.3 mmol) and 5-tert-butyl-isoxazole-3-isocyanate (5) (1.994 g, 12 mmol) in toluene (60 mL) was heated at 120 °C overnight. The reaction was quenched with a mixture of dichloromethane and water containing a little methanol, and the mixture was neutralized with saturated aqueous NaHCO3. The aqueous phase was extracted twice with dichloromethane, and the combined organic extracts were dried over MgSO4 and filtered. The filtrate was concentrated to a volume of about 20 mL and ethyl ether was added, resulting in the formation of a solid. The precipitate was collected by filtration, washed with ethyl ether, and dried under vacuum to give the free base of 7 (2.342 g, 41%).
 1H NMR (DMSO-d6) δ 9.6 (br, 1H), 8.9 (br, 1H), 8.61 (s, 1H), 7.86 (d, J = 8.9 Hz, 1H), 7.76 (d, J = 8.0 Hz, 2H), 7.69 (d, J = 1.3 Hz, 1H), 7.51 (d, J = 8.0 Hz, 2H), 7.18 (dd, J = 1.3 and 8.9 Hz, 1H), 6.52 (s, 1H), 4.16 (t, J = 5.7 Hz, 2H), 3.59 (t, J = 4.2 Hz, 4H), 3.36 (overlapping, 4H), 2.72 (t, J = 5.7 Hz, 2H), 1.30 (s, 9H).
General Procedure E for Preparation of Hydrochloride Salt
The free base was dissolved in a mixture of dichloromethane (20 mL) and methanol (1 mL). A solution of 1.0 M HCl in ethyl ether (1.1 equiv for all compounds except 7, for which 2.5 equiv were used) was added dropwise, followed by addition of ethyl ether. The precipitate was collected by filtration to give
N-(5-tert-butyl-isoxazol-3-yl)-N′-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,1-b][1,3]benzothiazol-2-yl]phenyl}urea dihydrochloride (7) (2.441 g, 98%).
1H NMR (DMSO-d6) δ 11.0 (br, 1H), 9.68 (s, 1H), 9.26 (s, 1H), 8.66 (s, 1H), 7.93 (d, J = 8.9 Hz, 1H), 7.78 (m, 3H), 7.53 (d, J = 8.7 Hz, 2H), 7.26 (dd, J = 2.4 and 8.9 Hz, 1H), 6.53 (s, 1H), 4.50 (t, J = 4.1 Hz, 2H), 3.97 (m, 2H), 3.81 (t, J = 12.1 Hz, 2H), 3.6 (overlapping, 4H), 3.23 (m, 2H), 1.30 (s, 9H). LC-MS (ESI) m/z 561 (M + H)+.
Anal. (C29H32N6O4S·2HCl) C, H, N. C: calcd 54.97; found 54.54. H: calcd 5.22; found 5.87. N: calcd 13.26; found 13.16.

References

  1.  Chao, Qi; Sprankle, Kelly G.; Grotzfeld, Robert M.; Lai, Andiliy G.; Carter, Todd A.; Velasco, Anne Marie; Gunawardane, Ruwanthi N.; Cramer, Merryl D.; Gardner, Michael F.; James, Joyce; Zarrinkar, Patrick P.; Patel, Hitesh K.; Bhagwat, Shripad S. (2009). “Identification of N-(5-tert-Butyl-isoxazol-3-yl)-N’-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,1-b][1,3]benzothiazol-2-yl]phenyl}urea Dihydrochloride (AC220), a Uniquely Potent, Selective, and Efficacious FMS-Like Tyrosine Kinase-3 (FLT3) Inhibitor”. Journal of Medicinal Chemistry 52 (23): 7808–7816.
  2.  Drug Tames Refractory AML. ASH Dec 2012
  3. NMR……….http://file.selleckchem.com/downloads/nmr/S152601-AC-220-HNMR-Selleck.pdf
  4. HPLC………http://file.selleckchem.com/downloads/hplc/S152601-AC-220-HPLC-Selleck.pdf

Figure


Filed under: Phase3 drugs, Uncategorized Tagged: orphan, quizartinib

Chinese Dodder Seeds, Tu Si Zi, Semen Cuscutae, 菟絲子 

$
0
0

Chinese Dodder Seeds ( Tu Si Zi ) 菟絲子 , also known as Beggarweed, Cuscutae, Devil’s Guts, Dodder Of Thyme, Hellweed, Lesser Dodder, Scaldweed, Strangle Tare, Tu Si Zi, Tu Sizi. Cuscuta epithymum; Cuscuta chinensis. It belong to the “Convolvulaceae” family.

Chinese Dodder Seeds ( Tu Si Zi ) 菟絲子 has a sweet, pungent and neurtal properties. It is use for treating the kidney and liver.

Chinese Dodder Seeds ( Tu Si Zi ) 菟絲子 Chinese Herbs Articles was created to help cleanse and rejuvenate your body enable you to<br /><br /><br /><br /><br /><br />
stay younger and healthier with chinese herbal recipes.” width=”348″ height=”272″ /></p>
<h3 style=

What does it do?
Property:
In the term of traditional Chinese medicine (TCM),
Tu Si Zi is acrid, sweet, neutral.
The channels Tu Si Zi influences are Kidney, Liver.Action:
In the term of TCM, Tu Si Zi:
1: Tonifies Kidneys, Augments Yin, Secures Jing and Reserves Urine.
2: Tonifies Kidneys and Liver, Improves Vision.
3: Benefits Spleen and Kidneys, Stops Diarrhea.
4: Calms the Fetus.

Usage:

• tonify kidneys, strengthen yin, secures essence, reserves urine.
• tonify liver, improves vision.
• strengthen spleen, stops diarrhea.
• calms fetus, habitual/threatened miscarriage.

 Other Use:

Orally, dodder is used for urinary tract, spleen, and hepatic disorders.

Cuscuta chinensis.Dodder.Dodder seed extract Pharmacological Actions.

Botanical Basic Data of Cuscuta chinensis(Dodder).:

Dodder Seed Extract Cuscuta chinensis Extract photo picture imageBotanical Source:Cuscuta chinensis(The ripe seed of Cuscuta chinensis Lam.,an annual voluble parasitic herb of the family Convolvulaceae).
Latin Name: Semen Cuscutae
Family: Convolvulaceae.
Common Name: Cuscuta seed, Chinese Dodder seed,Huang Si,Huang Teng Zi,Dou Ji Sheng.Huang Shan Teng.Wu Gen Cao,Wu Niang Teng,Huang Shan Si,Lao Ya Si,Huang Si Teng.
Scientific Name: Cuscuta chinensis Lam
Pin Yin Name: Tu Si Zi
Dodder Seed Extract Cuscuta chinensis Extract photo picture image
Cuscuta Classification in China:(1).Cuscuta chinensis Lam.;(2).Cuscuta australis R.Br.;(3).Cuscuta campestris Yunker;
Pinyin Name: Tu si zi,Also called Chinese Dodder Seed.

Pin yin description:tu is a character for this herb derived from the character meaning rabbit; si means silk, and zi means seeds, the part used; this plant is a parasitic weed that sets up a mat of hair-like fibers at its base and then rapidly sends fibrous stems upward; thus Tu Si refers to the quality of these fibers like silky rabbit hair; a common name for dodders in the West, based on the undesirable weed-like nature of these plants, is Devil’s Hair.
Part use:Dodder seed,Aerial parts.(whole plants are harvested in autumn when the seeds are ripe, and then threshed after dried to get the seeds)

Synoms:Dodder,love vine,strangleweed,devil’s-guts,goldthread,pull-down,devil’s-ringlet,hellbine,hairweed,devil’s-hair,Beggarweed, Cuscutae, Devil’s Guts, Dodder Of Thyme, Hellweed,Lesser Dodder,Scaldweed,Strangle Tare,Tu Si Zi,Tu Sizi,Cuscuta epithymum,Cuscuta chinensis and hailweed.

Habitat:Dodder grows throughout Europe, Asia, and southern Africa. Dodder prefers coastal and mountainous regions, and is gathered in summer.In China,mainly distributed in Jiangsu,Liaoning, Jilin, Hebei, Shandong and Henan provinces of China.
Taste:Pungent, Sweet,It is sweet in taste, warm in nature and manifests its therapeutic actions in the liver, kidney and spleen meridians.
Dodder Seed Extract Cuscuta chinensis Extract photo picture image

Constitutents:Dodder contains flavonoids (including kaempferol and quercitin) and hydroxycinnamic acid.

Cuscuta , or Dodder plant, is a parasitic vine that wraps around other plants for nourishment.The ripe seed of Cuscuta chinensis Lam.; an annual voluble parasitic herb of the family Convolvulaceae.Cuscuta seed is used in China for kidney deficiency. Cuscuta has a high content of flavonoids and has strong antioxidant properties. Cuscuta seed has been found in studies to have positive effects on sperm health and motility, and invigorates the reproductive system.

The plant growns near seashores.Slim stems spread out,twist and yellow color,no leaf.flower blossom fascination on axil.flower bud and small bud squama shape,caylx shape cup,5 divide,white crown,bell shape,double length of calyx.The flowers are hermaphrodite (have both male and female organs).Stamen flower flat short,squama grow on base,shape square roundness,2 room germen. Capsule shape flat ball.Seed 2~4,florescence July to September,fruit august to october. It can grow in semi-shade (light woodland) or no shade and requires moist soil.

Dodder is distributed in most parts of China. It is collected in autumn when the seed is ripe, dried in the sun and used unprepared or boiled after removal of impurities.


Filed under: CHINESE HERBS Tagged: Chinese Dodder Seeds, 菟絲子, Tu Si Zi
Viewing all 2025 articles
Browse latest View live