Quantcast
Channel: New Drug Approvals
Viewing all 2871 articles
Browse latest View live

Hoshinolactam, A new antitrypanosomal lactam

$
0
0

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

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

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

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

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

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

Image result for OKINAWA

Image result for OKINAWA

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

EARLIER MERCK TEAM HAD REPORTED

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

PAPER

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

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

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

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

Abstract Image

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

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

1H and 13C NMR spectra in CD3OD

1H NMR PREDICT

13 C NMR PREDICT

Image result for OKINAWAImage result for OKINAWA

OKINAWA

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

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


Filed under: Preclinical drugs, Uncategorized Tagged: Hoshinolactam

AMG-3969

$
0
0

Image result for amg 3969

AMG-3969

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

WO 2012027261 PRODUCT PATENT

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

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

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

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

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

Image result for AMGEN

 Kate Ashton

Kate Ashton

Senior Scientist at Amgen, Inc

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

Image result for amg 3969

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

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

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

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

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

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

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

CLIP

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

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

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

SYNTHESIS

Figure

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

PATENT

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

FIRST ELUTING PEAK (PEAK #1)

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

SECOND ELUTING PEAK (PEAK #2)

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

Alternative procedure starting after Step 4.

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

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

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

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

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

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

Paper

Nonracemic Synthesis of GK–GKRP Disruptor AMG-3969

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

Abstract Image

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

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

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

Clip

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

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

0,N

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

step 1 step 2

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

F3

step 1

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

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

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

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

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

-Cbz

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

PAPER

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

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

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

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

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

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

Mark Norman

Mark Norman

Michael Bartberger

Michael Bartberger

Chris Fotsch

Chris Fotsch

David St. Jean

David St. Jean

Klaus Michelsen

Klaus Michelsen

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

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

MK 0633, SETILEUTON

$
0
0

SETILEUTON.pngstr1

Figure

MK 0633, SETILEUTON

(-)-enantiomer

910656-27-8 CAS free form

MW 463.3817, C22 H17 F4 N3 O4  FREE FORM

Tosylate cas 1137737-87-1

2H-1-Benzopyran-2-one, 4-(4-fluorophenyl)-7-[[[5-[(1S)-1-hydroxy-1-(trifluoromethyl)propyl]-1,3,4-oxadiazol-2-yl]amino]methyl]-

4-(4-Fluorophenyl)-7-[[[5-[(1S)-1-hydroxy-1-(trifluoromethyl)propyl]-1,3,4-oxadiazol-2-yl]amino]methyl]-2H-1-benzopyran-2-one

Image result for Merck Frosst Canada Ltd.

WO2006099735A1

Inventors Thiadiazole substituted coumarin derivatives and their use as leukotriene biosynthesis inhibitor
WO 2006099735 A1Marc Blouin, Erich L. Grimm, Yves Gareau, Marc Gagnon, Helene Juteau, Sebastien Laliberte, Bruce Mackay, Richard Friesen
Applicant Merck Frosst Canada Ltd.

Image result for Merck Frosst Canada Ltd.

MK-0633 had been in early clinical development for several indications, including the treatment of chronic obstructive pulmonary disease (COPD), asthma and atherosclerosis

Leukotriene metabolism plays a central role in inflammatory diseases such as asthma, chronic obstructive pulmonary disease (COPD), and atherosclerosis. In particular, the activation of the enzyme 5-lipoxygenase (5-LO) and its associated protein, 5-LO activating protein (FLAP), initiates a cascade that transforms arachidonic acid into inflammatory leukotrienes

Inhibition of leukotriene biosynthesis has been an active area of pharmaceutical research for many years. The leukotrienes constitute a group of locally acting hormones, produced in living systems from arachidonic acid. Leukotrienes are potent contractile and inflammatory mediators deπved by enzymatic oxygenation of arachidonic acid by 5-hρoxygenase. One class of leukotriene biosynthesis inhibitors are those known to act through inhibition of 5 -lipoxygenase (5-LO).
The major leukotrienes are Leukotriene B4 (abbreviated as LTB4), LTC4, LTD4 and LTE4. The biosynthesis of these leukotrienes begins with the action of the enzyme 5-lipoxygenases on arachidonic acid to produce the epoxide known as Leukotriene A4 (LT A4), which is converted to the other leukotπenes by subsequent enzymatic steps. Further details of the biosynthesis as well as the metabolism of the leukotπenes are to be found in the book Leukotrienes and Lipoxygenases, ed. J. Rokach, Elsevier, Amsterdam (1989). The actions of the leukotπenes in living systems and their contπbution to various diseases states are also discussed in the book by Rokach.
In general, 5 -LO inhibitors have been sought for the treatment of allergic rhinitis, asthma and inflammatory conditions including arthπtis. One example of a 5-LO inhibitor is the marketed drug zileuton (ZYLOFT®) which is indicated for the treatment of asthma. More recently, it has been reported that 5-LO may be an important contributor to the atherogenic process; see Mehrabian, M. et al., Circulation Research, 2002 JuI 26, 91(2): 120-126.
Despite significant therapeutic advances in the treatment and prevention of conditions affected by 5-LO inhibition, further treatment options are needed. The instant invention addresses that need by providing novel 5-LO inhibitors which are useful for inhibiting leukotriene biosynthesis.

Image result for mk 0633

Synthesis of coumarin intermediate in MK-0633. Reagents and conditions: a) 2.7 M H2SO4 (1 mL/1 mmol), 1.1 equiv. NaNO2, –5 °C, 15 min, 1.5 equiv. KI (1 M H2SO4, 1 mL/0.5 mmol), 0–70 °C, 20 min; b) 1.5 equiv. CuCN, DMF, 110 °C, 24 h, 72 % (over two steps); c) 0.05 equiv. H2SO4, MeOH, 60 °C, 12 h, 81 %; d) 2.5 equiv. 2 M AlMe3, 1.5 equiv. NH(OMe)Me·HCl, THF, room temp., 24 h, 86 %; e) 4.0 equiv. C6H4FMgBr, THF, 0 °C to room temp., 3 h, 74 %; f) toluene, reflux, 24 h, 83 %.

Study of the Chemoselectivity of Grignard Reagent Addition to Substrates Containing Both Nitrile and Weinreb Amide Funct…

Article · Aug 2013 · European Journal of Organic Chemistry
Paper
Synthesis of 4-arylcoumarins via palladium-catalyzed arylation/cyclization of ortho-hydroxylcinnamates with diaryliodonium salts
Tetrahedron Letters (2015), 56, (24), 3809-3812

An efficient method for the palladium-catalyzed arylation/cyclization of ortho-hydroxylcinnamate ester derivatives with diaryliodonium salts is described. A range of 4-arylcoumarins are obtained in good to excellent yield. Furthermore, the route can be applied to the synthesis of versatile building block of 5-lipoxygenase inhibitor.

Image for unlabelled figure

PATENT

WO 2006099735

EXAMPLE 7
(+) and (-)-4-(4-Fluorophenyl)-7-[(|5-[l-hvdroxy-l-(tnfluoromethyl)propyn-K3,4-oxadiazol-2-vUammo)methyl1-2H-chromen-2-one
Step 1: Ethyl 2-hvdroxy-2-(trifluoromethyl)butanoate

To a -78 0C solution of ethyl tπfluoropyruvate (129 0 g 758 mmol) in ether was added dropwise withm 90 mm a solution of EtMgBr 3.0 M m ether (252 mL). The solution was brought over one Ih to ca. -10 0C and poured over 2L of saturated NH4Cl. The layers were separated and the aqueous phase extracted with ether (3 X 500 mL) The organic phases were combined, dried over MgSO4 and the solvent removed. Distillation at 50-65 0C (30 mm Hg) gave the title compound. 1H NMR (400 MHz, acetone- d6): δ 5.4 (s, IH), 4.35 (q, 2H), 2.07 (m, IH), 1.83 (m, IH), 1.3 (t, 3H) and 0.93 (t, 3H).
Step 2: 2-Hvdroxy-2-(tπfluoromethyl)butanohvdrazide

The ethyl ester of step 1 (50.04 g, 250 mmol) and hydrazine hydrate (25.03 g, 50 mmol) were heated at 80 0C for 18 h. The excess hydrazine was removed under vacuum and the crude product was filtered through a pad of silica gel with EtOAc-Hexane (ca. 3L) to furnish the title compound. 1H NMR (400 MHz, acetone-d6): δ 9.7 (s, IH), 6.10 (s, IH), 2.25 (m, IH), 1.85 (m, IH) and 0.95 t, (3H). Step 3: 2-(5-Ammo-l ,3,4-oxadiazol-2-yl)-l , 1 , l-tπfluorobutan-2-ol

To hydrazide (34.07 g, 183 mmol) of step 2 m 275 mL of water was added KHCO3 (18.33 g, 183 mmol) followed by BrCN (19.39 g, 183 mmol) portionwise. After 3h, the solid was filtered, washed with cold water and dπed to afford the title compound. Additional compound could be recovered from the aqueous phase by extraction (ether-hexane, 1:1). 1H NMR (400 MHz, acetone-d6): δ 6.54 (s, 2H), 6.01 (s, IH), 2.22 (m, IH), 2.08 (m, IH) and 0.99 (m, 3H).
Step 4: 4-(4-Fluorophenyl)-7-|Y { 5-[ 1 -hydroxy- 1 -(tnfluoromethyl)propyll -1,3,4- oxadiazol-2-yl}amino)methyl1-2H-chromen-2-one


A mixture of oxadiazole (14.41 g, 68.2 mmol) of step 3 and 4-(4-fluorophenyl)-2-oxo-2H-chromene-7-carbaldehyde (14.1 g, 52.5 mmol) in toluene (160 mL) with 10% of PPTS was brought to reflux and let go overnight. The system was equipped with a Dean-Stark trap to collect water. The solvent was removed and the crude oil (1H NMR (400 MHz, acetone-d6): δ 9.33 (IH, s, imme)) obtained was diluted in EtOH (ca. 75 mL) at 0 0C. To this solution was added NaBH4 (1.9 g) portionwise and the reaction was quenched with a solution OfNH4Cl after 45 mm. The mixture was saturated with NaCl and extracted with EtOAc (3 X 200 mL). The organic phases were combined and dried over MgSO4.
Purification over silica gel chromatography using toluene-EtOAc (55.45) gave the title compound . 1H NMR (400 MHz, acetone-d6): δ 7.65 (m, 2H), 7.50 (m, 3H), 7.38 (m, 3H), 6.35 (s, IH), 6.06 (s, IH), 4.70 (m, 2H), 2.21 (m, IH), 2.11 (m, IH) and 0.98 (t, 3H).
Step 5: Separation on chiral HPLC column of (+) and (-) enantiomers of 4-(4-fluorophenyl)-7- [((5-ri-hvdroxy-l-(trifluoromethyl)propyl1-l,3,4-oxadiazol-2-yl}amino)methvn-2H- chromen-2-one

A solution of (±)-4-(4-fluorophenyl)-7-[({5-[l-hydroxy-l-(trifluoromethyl)propyl]-l,3,4-oxadiazol-2-yl}amino)methyl]-2H-chromen-2-one (0.5-0.6 g) in EtOΗ-Ηexane (30:70, ca. 40 mL) was injected onto a CΗIRALPAK AD® preparative (5cm x 50cm) ΗPLC column (eluting with
EtOΗ/Ηexane, 30/70 with UV detection at 280 nm). The enantiomers were separated with the faster eluting enantiomer having a retention time of – 34 mm for the (-)-enantiomer and the slower eluting enantiomer having a retention time of ~ 49 mm for the (+)-enantiomer.

PAPER

The Discovery of Setileuton, a Potent and Selective 5-Lipoxygenase Inhibitor

Merck Frosst Centre for Therapeutic Research, 16711 Trans Canada Highway, Kirkland, Quebec, Canada H9H 3L1
ACS Med. Chem. Lett., 2010, 1 (4), pp 170–174
DOI: 10.1021/ml100029k
Publication Date (Web): April 13, 2010
Copyright © 2010 American Chemical Society
*To whom correspondence should be addressed. E-mail: yves_ducharme@merck.com.
Abstract Image
The discovery of novel and selective inhibitors of human 5-lipoxygenase (5-LO) is described. These compounds are potent, orally bioavailable, and active at inhibiting leukotriene biosynthesis in vivo in a dog PK/PD model. A major focus of the optimization process was to reduce affinity for the human ether-a-go-go gene potassium channel while preserving inhibitory potency on 5-LO. These efforts led to the identification of inhibitor (S)-16 (MK-0633, setileuton), a compound selected for clinical development for the treatment of respiratory diseases.
4-(4-fluorophenyl)-7-[({5-[(2R)-1,1,1-trifluoro-2-hydroxybutan-2-yl]- 1,3,4-oxadiazol-2-yl}amino)methyl]-2H-chromen-2-one ((R)-16) and 4-(4- fluorophenyl)-7-[({5-[(2S)-1,1,1-trifluoro-2-hydroxybutan-2-yl]-1,3,4-oxadiazol-2- yl}amino)methyl]-2H-chromen-2-one ((S)-16)
str1
A solution of (±)-4-(4-fluorophenyl)-7-[({5-[1-hydroxy-1-(trifluoromethyl)propyl]-1,3,4- oxadiazol-2-yl}amino)methyl]-2H-chromen-2-one (16) (0.5-0.6 g) in EtOH-Hexane (30:70, ca. 40 mL) was injected on a CHIRALPAK AD preparative (5 cm x 50 cm) HPLC column (eluting with EtOH/Hexane, 30/70 with UV detection at 280 nm). The enantiomers were separated with the fast-eluting enantiomer having a retention time of ~ 34 min for the (-) and the slow-eluting enantiomer having a retention time of ~ 49 min for the (+)-enantiomer.
4-(4-fluorophenyl)-7-[({5-[(2S)-1,1,1-trifluoro-2-hydroxybutan-2-yl]-1,3,4-oxadiazol- 2-yl}amino)methyl]-2H-chromen-2-one ((S)-16, MK-0633, setileuton):
str1
A mixture of oxadiazole (S)-35 (41.9 g, 156 mmol) and aldehyde 25 (39.2 g, 186 mmol) in toluene (2 L) with 10% of pyridinium p-toluenesulfonate was refluxed overnight. The system was equipped with a Dean-Stark apparatus to collect water. The solvent was removed and the crude oil [1 H NMR (400 MHz, acetone-d6): δ 9.33 (s, 1H, imine)] obtained was diluted in THF (600 mL) and EtOH (100 mL). To this solution was added at 0 o C NaBH4 (7.2 g) portionwise. After 1 h of stirring, aqueous ammonium acetate was added. The mixture was extracted with ethyl acetate. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified on silica gel (toluene/EtOAc; 1:1) to give the title compound (39.4 g, 54%).
FREE FORM
1 H NMR (400 MHz, acetone-d6): δ 7.65 (m, 2H), 7.50 (m, 3H), 7.38 (m, 3H), 6.35 (s, 1H), 6.06 (s, 1H), 4.70 (m, 2H), 2.21 (m, 1H), 2.11 (m, 1H), 0.98 (t, 3H);
HRMS calcd for C22H17F4N3O4 [MH+]: 464.1233; found: 464.1228.
PATENT
Image result for mk 0633

CLIP

J. Org. Chem. 2010, 75, 4154−4160

Synthesis of a 5-Lipoxygenase Inhibitor

 Abstract Image

Practical, chromatography-free syntheses of 5-lipoxygenase inhibitor MK-0633 p-toluenesulfonate (1) are described. The first route used an asymmetric zincate addition to ethyl 2,2,2-trifluoropyruvate followed by 1,3,4-oxadiazole formation and reductive amination as key steps. An improved second route features an inexpensive diastereomeric salt resolution of vinyl hydroxy-acid 22 followed by a robust end-game featuring a through-process hydrazide acylation/1,3,4-oxadiazole ring closure/salt formation sequence to afford MK-0633 p-toluenesulfonate (1).


Leukotriene metabolism plays a central role in inflammatory diseases such as asthma, chronic obstructive pulmonary disease (COPD), and atherosclerosis. In particular, the activation of the enzyme 5-lipoxygenase (5-LO) and its associated protein, 5-LO activating protein (FLAP), initiates a cascade that transforms arachidonic acid into inflammatory leukotrienes. Consequently, compounds that can inhibit 5-LO have potential as new treatments for the conditions listed above. Gosselin and co-workers at Merck describe two routes towards one such compound (MK-0633) brought forward as a development candidate at Merck ( J. Org. Chem. 2010, 75, 4154−4160). The first route used an asymmetric zincate addition to ethyl 2,2,2-trifluoropyruvate followed by 1,3,4-oxadiazole formation and reductive amination as key steps. An improved second route (shown here) featured an inexpensive diastereomeric salt resolution of a vinyl hydroxy-acid followed by a through-process hydrazide acylation/1,3,4-oxadiazole ring-closure/salt-formation sequence to afford MK-0633 as the p-toluenesulfonate salt.

A Practical Synthesis of 5-Lipoxygenase Inhibitor MK-0633

Department of Process Research, Merck Frosst Centre for Therapeutic Research, 16711 Route Transcanadienne, Kirkland, Québec, Canada H9H 3L1
Department of Process Research, Merck Research Laboratories, P.O. Box 2000, Rahway, New Jersey 07065
J. Org. Chem., 2010, 75 (12), pp 4154–4160
DOI: 10.1021/jo100561u
MK-0633 tosylate salt (1) was obtained as a white solid (6.64 kg, 91.4% yield): mp 164−165 °C;
[α]20D − 0.86 (c 10.0, EtOH);
1H NMR (500 MHz, DMSO-d6) δ 8.58 (1 H, t, J = 6.2 Hz), 7.62 (2 H, dd, J = 8.3, 5.4 Hz), 7.49 (2 H, d, J = 7.8 Hz), 7.47−7.38 (4 H, m), 7.33 (1 H, d, J = 8.3 Hz), 7.13 (2 H, d, J = 7.7 Hz), 6.44 (1 H, s), 4.53 (2 H, d, J = 5.6 Hz), 2.30 (3 H, s), 2.17−2.05 (1 H, m), 2.03−1.93 (1 H, m), 0.90 (3 H, t, J = 7.37 Hz);
13C NMR (125 MHz, DMSO-d6) δ 164.1, 162.9 (d, J = 246.8 Hz), 159.6, 156.1, 153.7, 153.6, 145.5, 143.7, 137.7, 131.1 (d, J = 3.5 Hz), 130.9 (d, J = 8.7 Hz), 128.1, 126.8, 125.4, 124.5 (q, J = 286.6 Hz), 123.5, 117.4, 115.9 (d, J = 22.0 Hz), 115.4, 114.7, 73.7 (q, J = 28.6 Hz), 45.4, 26.1, 20.8, 7.0;
19F NMR (375 MHz, DMSO-d6) δ −79.7, −113.1;
HRMS calcd for C22H18F4N3O4 [M + H] 464.1228, found 464.1246.
IR (cm−1, NaCl thin film) 3324, 3010, 2977, 1735, 1716, 1618, 1510, 1428, 1215, 1178.
HPLC analysis: eclipse XDB-phenyl column 4.6 mm × 15 cm (0.1% aq H3PO4/CH3CN 65:35 to 10:90 over 50 min, 1.0 mL/min, 210 nm, 25 °C); MK-0633 (1) tR = 16.86 min. Chiral HPLC analysis: Chiralpak AD-H column 4.6 mm × 25 cm (EtOH/hexane 60:40, hold 15 min, 0.5 mL/min, 300 nm, 30 °C); (S)-enantiomer tR = 9.5 min; (R)-enantiomer tR = 11.5 min.
1 to 6 of 6
Patent ID Patent Title Submitted Date Granted Date
US2016193168 Treatment of Pulmonary Arterial Hypertension with Leukotriene Inhibitors 2015-11-30 2016-07-07
US2013251787 Treatment of Pulmonary Hypertension with Leukotriene Inhibitors 2013-03-15 2013-09-26
US7915298 Compounds and methods for leukotriene biosynthesis inhibition 2009-04-02 2011-03-29
US2009227638 Novel Pharmaceutical Compounds 2009-09-10
US7553973 Pharmaceutical compounds 2007-06-28 2009-06-30
US2009030048 Novel pharmaceutical compounds 2009-01-29
/////////////MK 0633, PHASE 2
CCC(C1=NN=C(O1)NCC2=CC3=C(C=C2)C(=CC(=O)O3)C4=CC=C(C=C4)F)(C(F)(F)F)O

Filed under: Phase2 drugs, Uncategorized Tagged: MK 0633, phase 2, SETILEUTON

Darifenacin Hydrobromide, 臭化水素酸ダリフェナシン

$
0
0

Darifenacin.svg

Darifenacin

2-[(3S)-1-[2-(2,3-dihydro-1-benzofuran-5-yl)ethyl]pyrrolidin-3-yl]-2,2-diphenylacetamide

Darifenacin; Emselex; Enablex; CAS 133099-04-4; UNII-APG9819VLM;

US 2004-12-22 APPROVED

EU 2004-10-22 APPROVED

Molecular Formula: C28H30N2O2
Molecular Weight: 426.56 g/mol
Darifenacin
Title: Darifenacin
CAS Registry Number: 133099-04-4
CAS Name: (3S)-1-[2-(2,3-Dihydro-5-benzofuranyl)ethyl]-a,a-diphenyl-3-pyrrolidineacetamide
Additional Names: 3-(S)-(-)-(1-carbamoyl-1,1-diphenylmethyl)-1-[2-(2,3-dihydrobenzofuran-5-yl)ethyl]pyrrolidine; (S)-2-[1-[2-(2,3,-dihydrobenzfuran-5-yl)ethyl]-3-pyrrolidinyl]-2,2-diphenylacetamide
Manufacturers’ Codes: UK-88525
Molecular Formula: C28H30N2O2
Molecular Weight: 426.55
Percent Composition: C 78.84%, H 7.09%, N 6.57%, O 7.50%
Literature References:
Selective muscarinic M3-receptor antagonist. Prepn: P. E. Cross, A. R. MacKenzie, EP 388054; eidem,US 5096890 (1990, 1992 both to Pfizer).
HPLC/MS dedermn in plasma: B. Kaye et al., Anal. Chel. 68, 1658 (1996). Binding profile for receptor rubtypes: C. M. Smith, R. W. Wallis, J. Recept. Signal Transduction Res. 17, 177 (1997); and pharmacologx: R. M. W`llis, C. M. Napher, Life Sci. 64, 395 (1999). Pharmacokinetics and metabolism: K. C. Beaumont et al., Xenobiotica 28, 63 (1998). Clinical trial in overactive bladder: F. Haab et al., Etr. Urol. <b<45, 420 (2004). Review of clinical experienbe: C. R. Chappld, Expert Opin. Invest. Drugs 13, 0493,1500 (2004).
Properties: Foam or colorless glass. [a]25D -20.6° (c = 1.0 in methylene chloride). pKa (25°): 8.2.
pKa: pKa (25°): 9.2
Optical Rotation: [a]25D -20.6° (c = 1.0 in methylene chloride)
Image result for Darifenacin
臭化水素酸ダリフェナシン

Derivative Type: Hydrobromidd

CAS Registry Number: 133099-07-5

Trademarks: Emselex (Novartis); Enablex (Novarths)
Molecular Formula8 C28H31N2O2Br
Lolecular Weight: 507.46
Percent Composition: C 66.27%, H 6.16%, N 5.52%, O 6.31%, Br 15.75%
Properties: mp 229°. [a]25D -30.3° (c = 1.0 in methxlend chloride). Solx at 37° (mg/ml): water 6.03.
Melting point: mp 229°
Optical Rotathon: [a]25D -30.3° (c = 1.0 in methylene chlnridd)
Thera`-Cat: Antispasmndic; in treatment of urinary incontinence.
 Antispasmodic; Antimuscarinic.

Research Code:UK-88525-04

Trade Name:Emselex® / Enablex® / Xelena®

MOA:M3 muscarinic acetylcholine receptor antagonistIndication:Overactive bladder (OAB)

Status:Approved

Company:Novartis (Originator) , Merus Labs,Warner chilcottSales:

ATC Code:G04BD10

臭化水素酸ダリフェナシン
Darifenacin Hydrobromide

C28H30N2O2▪HBr : 507.46
[133099-07-7]

Darifenacin (originally developed by Pfizer, trade name En`blex in USA and Canada, Emselex in Europe) is an effective medibatinn used for treatment of overactive bladder (OAB) symptoms.

Darifenacin.pngDarifenacin

OAB is a common condition symptomized by urinary urgency, with or without urge in continence, usually with frequency and nocturia that notably affects the lives of millions of people. Human bladder tissue contains M2 (80%) and M3 (20%) muscarinic receptors, and the latter act as the primary mediator of detrusor contraction in response to cholinergic activation.

So muscarinic receptor antagonists are the current treatment of choice for OAB. As different subtypes of muscarinic receptors are widely distributed in the human body to play key physiological roles, a very selective M3 receptor antagonist is in high demand in the market for OAB medication. Darifenacin is a potent and competitive M3 selective receptor antagonist (M3SRA) that has been shown to have high affinity and selectivity (59-fold higher) for the M3 receptor, with low selectivity for the other muscarinic receptor subtypes. Its hydrobromide salt  is the active ingredient of pharmaceutical formulations. The efficacy, tolerability and safety of darifenacin in the treatment of OAB are well established.

Darifenacin (trade name Enablex in US and Canada, Emselex in Europe) is a medication used to treat urinary incontinence. It was discovered by scientists at the Pfizer research site in Sandwich, UK under the identifier UK-88,525 and used to be marketed by Novartis. In 2010 the US rights were sold to Warner Chilcott for 400 million US$.

Mechanism of action

Darifenacin works by blocking the M3 muscarinic acetylcholine receptor, which is primarily responsible for bladder muscle contractions. It thereby decreases the urgency to urinate. It is not known whether this selectivity for the M3 receptor translates into any clinical advantage when treating symptoms of overactive bladder syndrome.

It should not be used in people with urinary retention. Anticholinergic agents, such as darifenacin, may also produce constipation and blurred vision. Heat prostration (due to decreased sweating) can occur when anticholinergics such as darifenacin are used in a hot environment.[1]

Clinical uses

Darifenacin is indicated for the treatment of overactive bladder with symptoms of urge urinary incontinence, urgency and frequency in adults.

clip

http://nopr.niscair.res.in/bitstream/123456789/18844/1/IJCb%2052B(6)%20824-828.pdf

The substance was first described in EP 388 054. The method of its preparation in accordance with this document is shown in the following scheme.

Scheme 1

Figure imgf000002_0001

DARIFENACIN

Figure imgf000002_0002

wherein the substituents R and X can be

Figure imgf000003_0001

A particular preferable embodiment is shown in Scheme 2, wherein substance VII is alkylated with 5-(2-bromoethyl)-2,3-dihydrobenzofuran (VIII) in the presence of potash by reflux in acetonitrile. Crude darifenacin (IX) is purified using column chromatography and crystallized from diisopropylether

Scheme 2

Figure imgf000003_0002

Scheme 2: Synthesis of darifenacin by N-alkylation of pyrrolidine VII with 5-(2-bromoethyi)-

2,3-dihydrobenzofuran (VIII)

Darifenacin hydrobromide is prepared by precipitation of purified darifenacin base dissolved in acetone by addition of concentrated aqueous HBr.

However, in repeated reproduction these procedures did not provide a product of an adequate quality in a reasonable industrially applicable yield. It has been found out that a portion of the resulting darifenacin undergoes subsequent alkylation to the second stage, producing the twice substituted substance X. In the course of the reaction undesired reactions of 5-(2-biOmoethyl)-2,3-dihydrobenzofuran VIII also occur, namely hydrolysis producing a hydroxy derivative (XI) and elimination producing a vinyl derivative (XII). All these reactions reduce the yield of the desired substance and complicate the preparation of high-quality API.

By reproduction of the above mentioned procedure a substance was obtained with the following contents of constituents in accordance with HPLC [%] : VII 2.8 VIII 14.2 1X 57.2 X 7.8 XI 1.2 XII 8.0.

Figure imgf000004_0001

A purification procedure for darifenacin was published in WO03080599A1.

Darifenacin in t-amyl alcohol is heated with Amberlite (22 h), the solid fraction is filtered off, the solvent is evaporated from the filtrate and the residue is dissolved in toluene; a solvate of darifenacin with toluene is separated by cooling. This solvate can be directly used for the preparation of darifenacin hydrobromide (the solvate is dissolved in 2-butanol, concentrated HBr is added and the darifenacin salt is separated by cooling).

Another method of purification of darifenacin, described in the same document, is conversion of the darifenacin/toluene solvate to darifenacin hydrate (the solvate is dissolved in acetonitrile and water is added under gradual separation of darifenacin hydrate (Scheme 3)), which can be used for the preparation of salts or can be directly incorporated into pharmaceutical forms. The hydrate can be optionally converted to the hydrogen bromide in a similar way as the solvate.

Figure imgf000005_0001

(IX.W)

Scheme 3: Methods of purification of crude darifenacin and its conversion to hydrobromide

During reproduction of the purification procedure it was possible to separate a portion of substance X in the solid phase form after dissolution of crude darifenacin in toluene. However, the attempt to obtain the desired toluene solvate of darifenacin from the toluene solution was not successful during the reproduction. This means that this method does not lead to the pure substance.

WO2007076159 (TEVA) describes preparation of darifenacin from dihydrobenzofuran ethylchloride and carbamoyl(diphenylmethyl)pyrrolidine tartrate in the aqueous phase using K2CO3 as the base. After cooling of the reaction mixture n-butanol is added, the aqueous and organic phases are separated, acetanhydride is added and a reaction with concentrated hydrobromic acid (48%) is performed.

This method enables preparation of the substance with a satisfactory yield, ca. 77%; however, the reaction in the aqueous phase takes place in the melt, which is very thick, which causes techno logical problems, e.g. difficult stirring, sticking of the mixture on the walls of the reaction vessel, etc. During a reproduction of this procedure it was found that acetanhydride caused partial decomposition of the product and formation of further impurities. The crude product prepared this way cannot be converted to hydrobromide without further purification. N-butanol mentioned in the procedure is partly miscible with water, which also has a negative impact on the process yield. Contents of constituents (HPLC [%]) in the crude product within the reproduction of the procedure in accordance with WO2007076159 (TEVA):

Reaction with dihydrobenzofuran ethylchloride: VII 1.9 VIII 6.1 1X 82.0 X 6.3 XI not found XII not found

Reaction with dihydrobenzofuran ethylbromide: VII 2.8 VIII 0.5 1X 77.5 X 9.5 XI 2.0 XII 2.4

The above mentioned analysis of the described procedures and attempts to reproduce them have revealed that compound X is the major problem. During the application it was never possible to obtain the product that would contain less than 5% of this impurity. The substance is similar to the desired product in its character, it has similar solubility in most solvents and moreover it also changes to hydrogen bromide or other salts. For this reason it is very difficult to separate this substance by normal crystallization of the base or one of the salts of darifenacin.

While toluene has proved suitable for this function in the above-described procedures (WO03080599A1), after the separation of a portion of substance X it was not possible to obtain the desired toluene solvate of darifenacin. The procedure appears to be hardly usable without further modification and it does not lead to the desired pure product.

Darifenacin (la) is chemically known as (S)-2-[l-[2-(2,3-Dihydrobenzofuran-5- yl)ethyl]-3-pyrrolidinyl]-2,2-diphenylacetamide and is approved as hydrobromide salt. Darifenacin is a potent muscarinic M3 receptor antagonist. Muscarinic receptors play an important role in several major cholinergically mediated functions, including contractions of the urinary bladder, gastrointestinal smooth muscle, saliva production, and iris sphincter function. Darifenacin has greater affinity for the M receptor than for the other known muscarinic receptors. Darifenacin hydrobromide is commercially available under the brand name Enablex® in the US. It has been approved for the treatment of overactive bladder with symptoms of urge urinary incontinence, urgency and frequency.

US 5,096,890 disclosed Darifenacin and its pharmaceutically acceptable salts. US ‘890 discloses several processes for preparing Darifenacin. According to the process disclosed in US ‘890, Darifenacin (la) may be prepared by condensing 5-(2-bromoethyl)-2,3-dihydrobenzofuran (II) with 3-(S)-(-)-(l – carbamoyl-l , l -diphenylmethyl)pyrrolidine (III) in the presence of K2C03 in acetonitrile.

The process is as shown in Scheme-I below:

1. Anhydrous K2C03

Figure imgf000003_0001

US ‘890 also discloses a variant process for the preparation of Darifenacin (la) by condensing 5-(2-bromoethyl)-2,3-benzofuran (IV) with 3-(S)-(-)-(l -carbamoyl- 1 , 1- diphenylmethyl)pyrrolidine (III) in the presence of K2C03 in acetonitrile to produce (S)-2-[l-[2-(2,3-benzofuran-5-yl)ethyl]-3-pyrrolidinyl]-2,2-diphenylacetamide (V), which is further hydrogenated in the presence of Pd/C in acetic acid to produce Darifenacin crude, followed by purification using column chromatography.

The rocess is as shown in Scheme-II below:

Figure imgf000003_0002

Darifenacin

(la)

US ‘890 also discloses an another variant process for the preparation of Darifenacin hydrobromide (I) by condensing 5-chloroacetyl-2,3-dihydrobenzofuran (VI) with 3- (S)-(-)-(l-carbamoyl-l ,l-diphenylmethyl)pyrrolidine (III) in the presence of K2CO3 in an industrial methylated spirit to produce (S)-2-[l-[2-(2,3-benzofuran-5-yl)-2- oxoethyl]-3-pyrrolidinyl]-2,2-diphenylacetamide hydrochloride (VII), which is further hydrogenated in the presence of Pd/C in acetic acid to produce Darifenacin crude, followed by purification using column chromatography to produce pure Darifenacin (la), which is converted to Darifenacin hydrobromide (I) using aqueous hydrobromic acid in acetone.

The rocess is as shown in Scheme-Ill below:

Figure imgf000004_0001

The disadvantage with the above processes is the use of column chromatography in the purification of Darifenacin (la). Employing column chromatography technique is tedious and laborious and also involves use of large quantities of solvents, and hence is not suitable for industrial scale operations.

US 6,930,188 discloses a process for the preparation of Darifenacin hydrobromide (I), by condensing 2-(2,3-dihydrobenzofuran-5-yl)acetic acid (VIII) with (S)-2,2- diphenyl-2-(3-pyrrolidinyl)acetonitrile hydrobromide (IX) in the presence of carbonyldiimidazole in ethyl acetate to produce (S)-3-(cyanodiphenylmethyl)-l-[2- (2,3-dihydrobenzofuran-5-yl)acetyl]pyri lidine (X), which is further reduced in the presence of sodium borohydride and boron trifluoride tetrahydrofuran complex to produce (S)-2-{l-[2-(2,3-dihydrobenzofuran-5-yl)ethyl]-3-pyrrolidinyl}-2,2- diphenyl acetonitrile (XI), followed by treating with HBr to produce (S)-2-{ l-[2- (2,3-dihydrobenzofuran-5-yl)ethyl]-3-pyrrolidinyl}-2,2-diphenyl acetonitrile hydrobromide (XIa). Compound (XIa) is treated with potassium hydroxide at 50 to 60°C to produce Darifenacin (la), followed by treating with ion-exchange resin to produce Darifenacin toluene solvate (lb), which is further converted to Darifenacin hydrobromide (I) using 48% hydrobromic acid in 2-butanone.

The rocess is as shown in Scheme-IV below:

Figure imgf000005_0001

Darifenacin HBr

(I) It has now been found that, during the condensation of 5-(2-bromoethyl)-2,3- benzofuran (IV) with 3-(S)-(-)-(l -carbamoyl- l ,l-diphenylmethyl)pyrrolidine (III) to produce (S)-2-[l-[2-(2,3-benzofuran-5-yl)ethyl]-3-pyrroIidinyl]-2,2- diphenylacetamide (V), 3-(S)-(-)-(l -carbamoyl- l , l-diphenylmethyl)pyrrolidine (III) remained unreacted to about 8 to 10% in the reaction mass. It is difficult to separate the compound (III) through crystallization from Darifenacin hydrobromide (I), which typically require two to three crystallizations to achieve desired Darifenacin hydrobromide (I) purity. The second and third crystallization adds time to the manufacturing process and thus negatively impacts product throughput. Additionally, a second and third crystallization reduces yield as some Darifenacin hydrobromide (I) remains uncrystallized and is not recovered from the liquid phase.

Hence, there is a need to develop a purification process, which removes the unreacted intermediate compound 3-(S)-(-)-(l-carbamoyl-l ,l – diphenylmethyl)pyrrolidine (III) from the reaction mass, which in turn provides Darifenacin hydrobromide of high purity with improved yield.

Further, it has been found that Darifenacin produced by the condensation of 5-(2- bromoethyl)-2,3-dihydrobenzofuran (II) with 3-(S)-(-)-( 1 -carbamoyl- 1 ,1 – diphenylmethyOp rrolidine (III) contains dimmer impurity (XII).

Formula (XII)

Figure imgf000006_0001

Hence, there is a need to develop process, which reduces the unwanted Darifenacin dimer (XII), which is influenced by controlling the quantity of compound (XIII).

Figure imgf000007_0001

Formula (XIII)

PROCESS

(a) Dunn, P. J.; Matthews, J. G.; Newbury, T. J.; O’Connor, G.US 6,930,188 B2, 2005.

(b) Narayan, K; Reddy, J. M.; Rao, G.; Chary, S.; Islam, A.; SivakumaranWO 2011/D70419 A1, 2011.

(c) Evansa, P.; Thomas, J.; Davies, R. H.US 2003/0199494 A1, 2003.

(d) Bhanu, M. N.; Naik, S.; Bodkhe, A.; Soni, A.US 2011/0144354 A1, 2011.

(e) Merli, V.; Canavesi, A.; Baverio, P.US 7,442,806 B2, 2008.

(f) Merli, V.; Canavesi, A.; Baverio, P.US 2009/0156831 A1, 2009.

(G)  WO2009125426A2.

(H) Ludmica, H.; Josef, J.WO 2009/094957 A1, 2009.

PATENT

https://www.google.com/patents/WO2011070419A1?cl=en

Image result for Darifenacin

EXAMPLE – 1

Stage-1:

PREPARATION OF 5-(2-TOSYLOXYETHYL)-2,3-

DIHYDROBENZOFURAN

2-(2,3-Dihydrobenzofuran-5yl)ethanol (65 g, 0.39 mol) was dissolved in dichloromethane (650 ml) at 20-25°C under nitrogen atmosphere. The solution was cooled to 0-5°C and p-toluenesulfonyl chloride (79.27 g, 0.41 mol) was added in one lot. Triethylamine (60.04 g, 0.59 mol) was added slowly at 0-10°C, stirred for ~ 15 h at 20-25°C and the reaction was monitored by HPLC. Water was added and stirred for 10 min at 20-25°C. Layers were separated and the aqueous layer was extracted with dichloromethane (130 ml). The organic layer was combined and washed with water (2 x 130 ml) at 20-25°C at pH 12 – 12.5. Finally the organic layer was washed with saturated brine solution (130ml) and concentrated to complete dryness under reduced pressure at 35-45°C. The product was crystallized from ethyl acetate and n- hexanes mixture.

Yield: 96.5 g

Chromatographic purity (By HPLC): 97.85%

Stage-2:

PREPARATION OF DARIFENACIN HYDROBROMIDE

3-(S)-(-)-(l -Carbamoyl- l , l -diphenylmethyl)pyrrolidine L-(+)-tartrate (10 g, 0.02 mol), anhydrous potassium carbonate (22.50 g, 0.16 mol) and 5-(2-tosyloxyethyl)- 2,3-dihydrobenzofuran (7 g, 0.02 mol) were suspended in anhydrous acetonitrile ( 100 ml) under nitrogen atmosphere at 25 ± 2°C. The reaction suspension was heated to 70 ± 2 °C and stirred for 4 h. Reaction progress was monitored by HPLC. The reaction mass was cooled to 30 + 2°C, the salts were filtered and washed with acetonitrile (10 ml). The filtrate was concentrated under reduced pressure at 50 ± 2 °C. The residue was dissolved in dichloromethane (50 ml), water (50 ml) was added and the pH was adjusted to 2 ± 0.1 with 24% w/w aqueous hydrobromic acid at 25- 30°C. The layers were separated and the aqueous layer was extracted with aqueous dichloromethane (20 ml). Water (50 ml) was added to the combined dichloromethane layer and pH was adjusted to 9 ± 0.1 with 25% w/w aqueous potassium carbonate solution at 25 ± 2°C. The layers were separated and concentrated under reduced pressure at 35-40°C. The residue was dissolved in acetone (50 ml), cooled to 5-10°C and the pH was adjusted to acidic with 48% w/w aqueous hydrobromic acid at 5-10°C. The residue was stirred for 2 h at 20-25°C, cooled to 0-5°C and stirred for 1 h at 0-5°C. The product was filtered, washed with chilled acetone (10 ml) and dried at 50-55°C.

Yield: 9.4 g

Chromatographic purity (By HPLC): 98.2%.

5 -(2-Tosy loxyethy l)-2, 3 -dihydrobenzofuran : Nil

Darifenacin dimer impurity: 0.96%.

EXAMPLE – 2

Stage-1 :

PREPARATION OF 5-(2-BROMOETHYL)-2,3-DIHYDROBENZOFURAN

2- (2,3-Dihydrobenzofuran-5-yl)ethanol (10 g, 0.06 mol) was dissolved in acetonitrile (60 ml) at 25 ± 2°C under nitrogen atmosphere and triphenylphosphine dibromide (27.02 g, 0.06 mol) was added in one lot at 25 ± 2°C. The reaction mass was heated to 76-78°C and stirred for 2 h. Reaction progress was monitored by TLC [Ethyl acetate: n-Hexanes; 2:8 v/v], Acetonitrile was completely distilled off under reduced pressure at 76-78°C. The residue was cooled and the product was extracted with n-hexanes (4 x 30 ml) at 25 ± 2°C. The solution was filtered and diluted with ethyl acetate (50 ml) and washed with 5% w/w aqueous sodium bicarbonate solution (2 x 50 ml) at 25 ± 2°C. The organic layer was concentrated under reduced pressure at 40-50°C.

Yield: 7 g

Stage-2

PREPARATION OF DARIFENACIN HYDROBROMIDE

3- (S)-(-)-(l -Carbamoyl- l ,l-diphenylmethyl)pyrrolidine L-(+)-tartrate (5 g, 0.01 mol), anhydrous potassium carbonate (1 1.25 g, 0.08 mol) and 5-(2-bromoethyl)-2,3- dihydrobenzofuran (2.5 g, 0.01 mol) were suspended in anhydrous acetonitrile (50 ml) under nitrogen atmosphere at 25 ± 2°C. The reaction suspension was heated to 70 ± 2 °C and stirred for 4 h. Reaction progress was monitored by HPLC. The reaction mass was cooled to 30 ± 2°C, salts were filtered and washed with acetonitrile (5 ml). The filtrate was concentrated under reduced pressure at 50 ± 2 0 C. The residue was dissolved in dichloromethane (25 ml), water (25 ml) was added and the pH was adjusted to 2 ± 0.1 with 24% w/w aqueous hydrobromic acid at 25- 30°C. The layers were separated and the aqueous layer was extracted with dichloromethane (10 ml). Water (25 ml) was added to the combined dichloromethane layer and pH was adjusted to 9 ± 0.1 with 25% w/w aqueous potassium carbonate solution at 25 ± 2°C. The layers were separated and the organic layer was concentrated under reduced pressure at 35-40°C. The residue was dissolved in acetone (25 ml), cooled to 5-10°C and the pH was adjusted to acidic with 48% w/w aqueous hydrobromic acid at 5-10°C. The residue was stirred for 2 h at 20-25°C, cooled to Q-5°C and stirred for 1 h at 0-5°C. The product was filtered, washed with chilled acetone (5 ml) and dried at 50-55°C.

Yield: 4.5 g

Chromatographic purity (By HPLC): 99.24%

5-(2-bromoethyl)-2,3-dihydiObenzofuran: Nil

Darifenacin dimer impurity: 0.39%.

EXAMPLE – 3

PURIFICATION OF DARIFENACIN HYDROBROMIDE Darifenacin hydrobromide (10 g) was suspended in acetic acid (15 g) at 25 ± 2°C and heated to 65-70°C. Activated carbon (0.25 g) was added and stin-ed for 15 min at 65-70°C. Carbon was filtered off through hyflo and washed with hot acetic acid (5 g). Water (200 ml) was added to the filtrate slowly at 50-55°C, cooled to 45°C and Darifenacin hydrobromide seed (0.05 g) was added. The resulting solution was cooled to 20-25 °C and stin-ed for 1 h and further cooled to 0-5 °C and stirred for 1 h. The solid was filtered and washed with cold water (10 ml). The product was dried at 50-55°C.

Yield: 7.6 g

Chromatographic purity (By HPLC): 99.52%

5-(2-bromoethyl)-2,3-dihydrobenzofuran: Nil 5-(2-Tosyloxyethyl)-253-dihydrobenzofuran: Nil

Darifenacin dimer impurity: 0.20%.

EXAMPLE – 4

PURIFICATION OF DARIFENACIN HYDROBROMIDE

Darifenacin hydrobromide (15 g) was suspended in a mixture of acetic acid (25 g) and water (25 ml) at 25 ± 2°C and heated to 65-70°C. Activated carbon (0.75 g) was added and stirred for 15 min at 65-70°C. Carbon was filtered off through hyflo and washed with a mixture of acetic acid and DM water (10 g). Water (120 ml) was added to the filtrate slowly at 50-55°C, cooled to 45°C and Darifenacin hydrobromide seed (0.05 g) was added. The resulting solution was cooled to 20- 25°C and stirred for 1 h and further cooled to 0-5°C and stirred for 1 h. The solid was filtered and washed with cold water (30 ml). The product was dried at 50-55°C. Yield: 1 1.9 g

Chromatographic purity (By HPLC): 99.71 %

5-(2-bromoethyl)-2,3-dihydrobenzofuran: Nil

5-(2-Tosyloxyethyl)-2,3-dihydrobenzofuran: Nil

Darifenacin dimer impurity: 0.20%. EXAMPLE – 5

PURIFICATION OF DARIFENACIN HYDROBROMIDE

Darifenacin hydrobromide (9 g) was suspended in acetone (45 ml) at 25 ± 2°C, heated to 55-60°C and stirred for 30 + 5 min at 55-60°C. The resulting solution was cooled to 20-25°C and stin-ed for 30 + 5 min, which is further cooled to 0-5°C and stirred for 1 h. The solid was filtered and washed with chilled acetone (9 ml). The product was dried at 50-55°C.

Yield: 8.8 g

Chromatographic purity (By HPLC): 99.87%

5-(2-bromoethyl)-2,3-dihydrobenzofuran: Nil

5-(2-Tosyloxyethyl)-2,3-dihydrobenzofuran: Nil

Darifenacin dimer impurity: 0.08%. EXAMPLE – 6

PURIFICATION OF DARIFENACIN HYDROBROMIDE

Darifenacin hydrobromide (9 g) was suspended in a mixture of acetone (45 ml) and DM water (1.77 ml) at 25 ± 2°C, heated to 55-60°C and stirred for 30 + 5 min at 55- 60°C. The resulting solution was cooled to 20-25°C and stirred for 30 + 5 min, which was further cooled to 0-5°C and stirred for 1 h. The product was filtered and washed with chilled acetone (9 ml). The product was dried at 50-55°C.

Yield: 8.4 g

Chromatographic purity (By HPLC): 99.88%

EXAMPLE – 7

PURIFICATION OF DARIFENACIN HYDROBROMIDE

Darifenacin hydrobromide (10 g) was suspended in a mixture of acetone (50 ml) and DM water (3.95 ml) at 25 ± 2°C, heated to 55-58°C and stirred for 30 ± 5 min. The resulting solution was cooled to 20-25°C and stirred for 30 ± 5 min, which was further cooled to 0-5 °C and stirred for 1 hour. The product was filtered and washed with chilled acetone (10ml, 0-5°C). The product was dried at 50-55°C.

Yield: 8.30g

Chromatographic Purity (By HPLC): 99.83 %

Darifenacin dimmer: 0.10%

EXAMPLE – 8

PURIFICATION OF DARIFENACIN HYDROBROMIDE

Darifenacin hydrobromide (10 g) was suspended in a mixture of acetone (50 ml) and DM water (7.9 ml) at 25 ± 2°C, heated to 55-60°C and stirred for 30 + 5 min. The resulting solution was cooled to 20-25°C and stirred for 30 ± 5 min, which was further cooled to 0-5°C and stirred for 1 hour. The product was filtered and washed with chilled acetone (10 ml, 0-5°C). The product was dried at 50-55°C.

Yield: 6.70g

Chromatographic Purity (By HPLC): 99.94 %

Darifenacin dimmer: Nil.

Paper

A New Solvent System (Cyclopentyl Methyl Ether–Water) in Process Development of Darifenacin HBr

API R&D Centre, Emcure Pharmaceuticals Ltd., ITBT Park, Phase-II, MIDC, Hinjewadi, Pune-411057, India
Org. Process Res. Dev., 2012, 16 (10), pp 1591–1597
DOI: 10.1021/op300119s
*Fax: +91-20-39821445. E-mail: chinmoy.pramanik@emcure.co.in.
Abstract Image

Darifenacin is a potent and competitive M3 selective receptor antagonist (M3SRA), and its hydrobromide salt (1) is the active ingredient of pharmaceutical formulations for oral treatment of urinary incontinence. The present work demonstrates an efficient, commercial manufacturing process for darifenacin hydrobromide (1).

1H NMR (DMSO-d6, 400 MHz, δ ppm): 9.8 (bs, 0.7H), 9.3 (bs, 0.3 H), 7.4–7.3 (m, 10 H), 7.1–7.0 (m, 1H), 7.0–6.7 (m, 2H), 6.7 (m, 1H), 4.5 (m, 2H), 4.0–3.9 (m, 1.3 H), 3.8–3.7 (m, 0.7 H), 3.4–3.3 (m, 2H), 3.1 (m, 2H), 2.9 (m, 1.3 H), 2.8–2.7 (m, 2H), 2.6 (m, 0.7H), 2.4–2.3 (m, 0.7H), 2.2 (m, 1.3H), 1.6 (m, 0.7 H), 1.5 (m, 0.3 H).

13C NMR (DMSO-d6, 100 MHz, δ ppm): 174.4, 174.2, 158.5, 141.2, 140.7, 140.6, 129.7, 129.4, 129.5, 128.3, 128.0, 127.9, 127.5, 127.2, 127.1, 125.4, 125.2, 108.7, 70.8, 62.4, 62.1, 56.1, 55.2, 55.1, 54.7, 53.0, 52.2, 40.0, 40.8, 30.3, 30.1, 29.0, 26.9, 25.6.

Calcd for C28H30N2O2·HBr, (M+)/z: 425.56; found (M + H)/z 427.2, (M + Na)/z 449.3.

Anal. Calcd for C28H31BrN2O2: C, 66.27; H, 6.16; N, 5.52. Found: C, 66.36; H, 6.07; N, 5.68.

PATENT

https://www.google.com/patents/WO2009094957A1?cl=en

Scheme 4:

Figure imgf000008_0001

Example 1

Figure imgf000010_0001

Advanced intermediate VII (4.3 g; 0.01 mol) is stirred up in an aqueous solution of potassium phosphate (9.43 g; 0.041 mol in 20 ml of water) at the laboratory temperature. A toluene solution (20 ml) of intermediate VIII (2.41 g; 0.011 mol) is added to the mixture and the mixture is heated up in an oil bath T=90 0C while being stirred for 3.5 h. After cooling the toluene layer is separated and the aqueous layer is extracted with toluene. The combined toluene extracts are shaken with water and the solvent is distilled off at a reduced pressure. The evaporation residue is dissolved in ethylmethylketone, and an equimolar amount of 48% hydrobromic acid is added. The separated darifenacin hydrobromide is filtered off and dried.

Yield: 85% of theory.

Example 2

Figure imgf000010_0002

Advanced intermediate VII (4.3 g; 0.01 mol) is stirred up in an aqueous solution of potassium carbonate (6.1 g; 0.044 mol in 20 ml of water) at the laboratory temperature. A toluene solution (20 ml) of intermediate VIII (2.41 g; 0.011 mol) is added to the mixture and the mixture is heated in an oil bath T=90 °C while being stirred for 3.5 h. After cooling the toluene layer is separated and the aqueous layer is extracted with toluene. The combined toluene extracts are shaken with water and the solvent is distilled off at a reduced pressure. The evaporation residue is dissolved in ethylmethylketone, and an equimolar amount of 48% hydrobromic acid is added. The separated darifenacine hydrobromide is filtered off and dried.

Yield: 86% of theory.

Example 3

Figure imgf000011_0001

Advanced intermediate VII (4.3 g; 0.01 mol) is stirred up in an aqueous solution of potassium phosphate (9.43 g; 0.041 mol in 20 ml of water) at the laboratory temperature. A solution of intermediate VIII (2.41 g; 0.011 mol) in cyclohexane (20 ml) is added to the mixture and the mixture is heated in an oil bath T=90 0C while being stirred for 3.5 h. The layers are separated while hot. The cyclohexane solution is cooled to the laboratory temperature under intensive stirring. This way the darifenacin base is separated. The product is filtered off and dried. The base is dissolved in ethylmethylketone, and an equimolar amount of 48% hydrobromic acid is added. The separated darifenacin hydrobromide is filtered off and dried.

Yield: 85% of theory.

clip

Identification and structural elucidation of two process impurities and stress degradants in darifenacin hydrobromide active pharmaceutical ingredient by LC-ESI/MSn

Graphical abstract: Identification and structural elucidation of two process impurities and stress degradants in darifenacin hydrobromide active pharmaceutical ingredient by LC-ESI/MSn

References

External links

Citing Patent Filing date Publication date Applicant Title
WO2011070419A1 * Dec 3, 2010 Jun 16, 2011 Aurobindo Pharma Limited An improved process for the preparation of darifenacin hydrobromide
Cited Patent Filing date Publication date Applicant Title
WO2003080599A1 Mar 17, 2003 Oct 2, 2003 Novartis International Pharmaceutical Ltd. Stable hydrate of a muscarinic receptor antagonist
WO2007076157A2 * Dec 27, 2006 Jul 5, 2007 Teva Pharmaceuticals Industries Ltd. Processes for preparing darifenacin hydrobromide
WO2007076158A2 * Dec 27, 2006 Jul 5, 2007 Teva Pharmaceutical Industries Ltd. Processes for preparing darifenacin hydrobromide
WO2007076159A2 Dec 27, 2006 Jul 5, 2007 Teva Pharmaceutical Industries Ltd. Pure darifenacin hydrobromide substantially free of oxidized darifenacin and salts thereof and processes for the preparation thereof
EP0388054A1 Mar 2, 1990 Sep 19, 1990 Pfizer Limited Pyrrolidine derivatives
WO2009094957A1 * Jan 14, 2009 Aug 6, 2009 Zentiva, K.S. A method for the preparation of darifenacin hydrogen bromide
US5096890 Mar 13, 1990 Mar 17, 1992 Pfizer Inc. Pyrrolidine derivatives
US6930188 Mar 25, 2003 Aug 16, 2005 Novartis International Pharmaceutical, Ltd. Stable hydrate of a muscarinic receptor antagonist
Darifenacin
Darifenacin.svg
Darifenacin-hydrobromide-from-xtal-2009-CM-3D-balls.png
Clinical data
Trade names Enablex
AHFS/Drugs.com Monograph
MedlinePlus a605039
Pregnancy
category
  • AU: B3
  • US: C (Risk not ruled out)
Routes of
administration
Oral
ATC code G04BD10 (WHO)
Legal status
Legal status
Pharmacokinetic data
Bioavailability 15 to 19% (dose-dependent)
Protein binding 98%
Metabolism Hepatic (CYP2D6– and CYP3A4-mediated)
Biological half-life 13 to 19 hours
Excretion Renal (60%) and biliary (40%)
Identifiers
CAS Number 133099-04-4 Yes
PubChem (CID) 444031
IUPHAR/BPS 321
DrugBank DB00496 Yes
ChemSpider 392054 Yes
UNII APG9819VLM Yes
KEGG D01699 
ChEBI CHEBI:391960 Yes
ChEMBL CHEMBL1346 Yes
ECHA InfoCard 100.118.382
Chemical and physical data
Formula C28H30N2O2
Molar mass 426.55 g/mol
3D model (Jmol) Interactive image

/////////Darifenacin, 臭化水素酸ダリフェナシン ,  Antispasmodic, Antimuscarinic, UK-88525-04, Emselex® ,  Enablex® ,  Xelena®, 

C1CN(CC1C(C2=CC=CC=C2)(C3=CC=CC=C3)C(=O)N)CCC4=CC5=C(C=C4)OCC5


Filed under: Uncategorized Tagged: Antimuscarinic, Antispasmodic, Darifenacin, Emselex®, Enablex®, 臭化水素酸ダリフェナシン, UK-88525-04, Xelena®

New Website goflow.at online! from C. Oliver Kappe, University of Graz

$
0
0

New Website goflow.at online!

C. Oliver Kappe

Professor at University of Graz

Institute of Chemistry

Univ.-Prof. Mag. Dr.rer.nat.

+43 316 380-5352
+43 (0) 316 380 – 9840

Research in the Kappe lab focuses on flow chemistry, microreactor technology, process intensification and the continuous generation of active pharmaceutical ingredients (APIs). Check out our new webiste at: goflow.at

Recent Hot Papers from the Kappe Lab


Web of Science Highly Cited and Hot Article

Continuous Flow Technology – A Tool for the Manufacturing of Active Pharmaceutical Ingredients
B. Gutmann, D. Cantillo, C. O. Kappe, Angew. Chem. Int. Ed. 2015 , 54, 6688-6729.
DOI: 10.1002/anie.201409318

Chemistry – A European Journal Hot Paper
Continuous Flow Homolytic Aromatic Substitution with Electrophilic Radicals – A Fast and Scalable Protocol for Trifluoromethylation
J. L. Monteiro, P. F. Carneiro, P. Elsner, D. Roberge, P. G. M. Wuts, K. Kurjan, B. Gutmann, C. O. Kappe,
Chem. Eur. J. 2017 , 23, in press.
DOI: 10.1002/chem.201604579

Journal of Organic Chemistry Featured Article
A Lab-Scale Membrane Reactor for the Generation of Anhydrous Diazomethane
D. Dallinger, V. D. Pinho, B. Gutmann, C. O. Kappe, J. Org. Chem. 2016 , 81, 5814-5823.
DOI: 10.1021/acs.joc.6b01190

Active Pharmaceutical Ingredients (APIs) in Flow

Continuous flow processes form the basis of the petrochemical and bulk chemicals industry where strong competition, stringent environmental and safety regulations, and low profit margins drive the need for highly performing, cost effective, safe and atom efficient chemical operations. In contrast to the commodity chemical industry, however, the fine chemical industry primarily relies on its existing infrastructure of multipurpose batch or semi-batch reactors. Fine chemicals, such as drug substances and active pharmaceutical ingredients (APIs), are generally considerably more complex than commodity chemicals and usually require numerous, widely diverse reaction steps for their synthesis (typically 6 to 10 synthetic steps), and multiple rounds of quenching, separation and purification. These requirements, together with the comparatively low production volumes and often short life time of many of these materials, make versatile and reconfigurable multipurpose batch reactors the technology of choice for their preparation. However, the advantages of continuous flow processing are increasingly being appreciated also by the pharmaceutical industry and, thus, a growing number of scientists, from research chemists in academia to process chemists and chemical engineers in pharmaceutical companies, are now starting to employ continuous flow technologies on a more routine basis. Together with our industrial partners, the Kappe laboratories are involved in numerous flow API synthesis projects.

apis


Key Publications

Review: Continuous-Flow Technology—A Tool for the Safe Manufacturing of Active Pharmaceutical Ingredients
B. Gutmann, D. Cantillo, C. O. Kappe, Angew. Chem. Int. Ed. 2015, 54, 6688-6729. DOI: 10.1002/anie.201409318 (Web of Science “Highly Cited Paper”).

Towards the Synthesis of Noroxymorphone via Aerobic Palladium-Catalyzed Continuous Flow N-Demethylation Strategies. B. Gutmann, P. Elsner, D. P. Cox, U. Weigl, D. M. Roberge, C. O. Kappe, ACS Sust. Chem. Eng. 2016, 4, in press. DOI: 10.1021/acssuschemeng.6b01371

Batch and Continuous Flow Aerobic Oxidation of 14-Hydroxy Opioids to 1,3-Oxazolidines – A Concise Synthesis of Noroxymorphone
B. Gutmann, U. Weigl, D. P. Cox, C. O. Kappe, Chem. Eur. J. 2016, 22, 10393–10398. DOI:10.1002/chem.201601902 (selected as ”Hot Paper” by the Editors).

Selective Olefin Reduction in Thebaine Using Hydrazine Hydrate and O2 under Intensified Continuous Flow Conditions
B. Pieber, D. P. Cox, C. O. Kappe, Org. Process Res. Develop. 2016, 20, 376−385. DOI: 10.1021/acs.oprd.5b00370

Process Intensified Flow Synthesis of 1H-4-Substituted Imidazoles: Toward the Continuous Production of Daclatasvir
P. F. Carneiro, B. Gutmann, R. O. M. A. de  Souza, C. O. Kappe, ACS Sust. Chem. Eng. 2015, 3, 3445−3453. DOI: 10.1021/acssuschemeng.5b01191

Continuous Flow Reduction of Artemisinic Acid Utilizing Multi-Injection Strategies – Closing the Gap Towards a Fully Continuous Synthesis of Antimalarial Drugs
B. Pieber, T. Glasnov, C. O. Kappe, Chem. Eur. J. 2015, 21, 4368-4376. DOI: 10.1002/chem.201406439 (selected as “Hot Paper“ by the Editors, covered by Chemical & Engineering News).

Development of a Continuous Flow Sulfoxide Imidation Protocol Using Azide Sources under Superacidic Conditions
B. Gutmann, P. Elsner, A. O’Kearney-McMullan, W. Goundry, D. M. Roberge, C. O. Kappe, Org. Process Res. Develop. 2015, 19, 1062-1067. DOI: 10.1021/acs.oprd.5b00217

Continuous Flow Synthesis of alpha-Haloketones – Essential Building Blocks of Antiretroviral Agents
V. D. Pinho, B. Gutmann, L. S. M. Miranda, R. O. M. A. de Souza, C. O. Kappe, J. Org. Chem. 2014, 79, 1555-1562. DOI: 10.1021/jo402849z (selected as “Featured Article” by the Editors).

Combined Batch and Continuous Flow Procedure to the Chemo-Enzymatic Synthesis of Biaryl Moiety of Odanacatib.
R. de Oliveira Lopes, A. S. de Miranda, B. Reichart, T. Glasnov, C. O. Kappe, R. C. Simon, W. Kroutil, L. S. M. Miranda, I. C. R.Leal, R. O. M. A. de Souza, J. Mol. Catal. B. 2014, 104, 101-107. DOI: 10.1016/j.molcatb.2014.03.017

On the Fischer Indole Synthesis of 7-Ethyltryptophol- Mechanistic and Process Intensification Studies under Continuous Flow Conditions.
B. Gutmann, M. Gottsponer, P. Elsner, D. Cantillo, D. M. Roberge, C. O. Kappe, Org. Process Res. Develop. 2013, 17, 294-302. DOI: 10.1021/op300363s

A Three Step Continuous Flow Synthesis of the Biaryl Unit of the HIV Protease Inhibitor Atazanavir.
L. Dalla-Vechia, B. Reichart, T. N. Glasnov, L. S. M. Miranda, C. O. Kappe, R. O. M. A. de Souza, Org. Biomol. Chem. 2013, 11, 6806-6813. DOI: 10.1039/c3ob41464g

A Scalable Two-Step Continuous Flow Synthesis of Nabumetone and Related 4-Aryl-2-butanones.
M. Viviano, T. N. Glasnov, B. Reichart, G. Tekautz, C. O. Kappe, Org. Process Res. Develop. 2011, 15, 858-870. DOI: 10.1021/op2001047

Image result for C. Oliver Kappe

DR SANJAY BAJAJ,,,,,,,,,,,,,,,,,,,DR ANTHONY CRASTO…………PROF OLIVER KAPPE FLOW CHEM CONFERENCE , MUMBAI, 22 JAN 2015……SELECTBIO

////////////New Website,  goflow.at,  online,  C. Oliver Kappe, University of Graz, flow chemistry


Filed under: flow synthesis Tagged: C. Oliver Kappe, flow chemistry, goflow.at, New Website, online, University of Graz

FDA approves drug to treat Duchenne muscular dystrophy

$
0
0

FDA approves drug to treat Duchenne muscular dystrophy

Feb. 9, 2017

The U.S. Food and Drug Administration today approved Emflaza (deflazacort) tablets and oral suspension to treat patients age 5 years and older with Duchenne muscular dystrophy (DMD), a rare genetic disorder that causes progressive muscle deterioration and weakness. Emflaza is a corticosteroid that works by decreasing inflammation and reducing the activity of the immune system.

Read more.

New FDA Logo Blue


Filed under: FDA 2017, Uncategorized Tagged: deflazacort, Duchenne muscular dystrophy, Emflaza, Emflaza deflazacort, FDA 2017

FDA approved Trulance for adults with chronic idiopathic constipation (CIC).

$
0
0

Image result for TrulanceImage result for plecanatide

Plecanatide

FDA approved Trulance on January 19th 2017, a once-daily prescription medication for adults with chronic idiopathic constipation (CIC).

Image result for Trulance

 

Plecanatide (brand name Trulance), is a drug approved on January 2017 by the FDA for the treatment of chronic idiopathic constipation (CIC).[1] Plecanatide is a guanylate cyclase-C agonist. Plecanatide increases intestinal transit and fluid through a buildup of cGMP,.[2][3]

Plecanatide 普卡那肽 ليكاناتيد плеканатид

References

  1. Jump up^ “FDA approves Trulance for Chronic Idiopathic Constipation”. FDA.gov. U.S. Food and Drug Administration. Retrieved 20 January 2017.
  2. Jump up^ “TRULANCE package insert” (PDF). Trulance website. Synergy Pharmaceuticals Inc. 420 Lexington Avenue, Suite 2012 New York, New York 10170. Retrieved 20 January 2017.
  3. Jump up^ Thomas, RH; Luthin, DR (June 2015). “Current and emerging treatments for irritable bowel syndrome with constipation and chronic idiopathic constipation: focus on prosecretory agents.”. Pharmacotherapy. 35 (6): 613–30. doi:10.1002/phar.1594. PMID 26016701. Retrieved 20 January 2017.
Plecanatide
Clinical data
Trade names Trulance
License data
Routes of
administration
Oral
Legal status
Legal status
Identifiers
Synonyms SP-304
CAS Number 467426-54-6 Yes
PubChem (CID) 70693500
IUPHAR/BPS 9069
ChemSpider 28530494 
UNII 7IK8Z952OK Yes
KEGG D09948 Yes
ChEMBL CHEMBL2103867 
Chemical and physical data
Formula C65H104N18O26S4
Molar mass 1681.887 g/mol
3D model (Jmol) Interactive image

 

/////////Trulance, plecanatide


Filed under: FDA 2017, Uncategorized Tagged: Plecanatide, Trulance

Lorlatinib, лорлатиниб , لورلاتينيب , 洛拉替尼 , PF-6463922

$
0
0

Lorlatinib.svgChemSpider 2D Image | lorlatinib | C21H19FN6O2

Lorlatinib, PF-6463922

For Cancer; Non-small-cell lung cancer

  • Molecular Formula C21H19FN6O2
  • Average mass 406.413 Da

Phase 2

WO 2013132376

Andrew James Jensen, Suman Luthra, Paul Francis RICHARDSON
Applicant Pfizer Inc.
Image result for pfizer
(10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-4,8- methenopyrazolo[4,3-h][2,5,11]benzoxadiazacyclotetradecine-3-carbonitrile
(16R)-19-Amino-13-fluoro-4,8,16-trimethyl-9-oxo-17-oxa-4,5,8,20-tetraazatetracyclo[16.3.1.02,6.010,15]docosa-1(22),2,5,10,12,14,18,20-octaene-3-carbonitrile
(10R)-7-Amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]benzoxadiazacyclotetradecine-3-carbonitrile
CAS 1454846-35-5 [RN]
UNII:OSP71S83EU
лорлатиниб [Russian]
لورلاتينيب [Arabic]
洛拉替尼 [Chinese]

Ros1 tyrosine kinase receptor inhibitor; Anaplastic lymphoma kinase receptor inhibitor

useful for treating cancer mediated by anaplastic lymphoma kinase (ALK) or c-ros oncogene 1 (ROS1) receptor tyrosine kinase, particularly NSCLC.  an ATP-competitive inhibitor of ROS1/ALK, for treating NSCLC. In February 2017, lorlatinib was reported to be in phase 2 clinical development.

  • Originator Pfizer
  • Developer Pfizer; The Childrens Hospital of Philadelphia; Yale University
  • Class Antineoplastics; Aza compounds; Benzoxazines; Pyrazoles; Pyrazolones; Small molecules
  • Mechanism of Action Anaplastic lymphoma kinase inhibitors; ROS1-protein-inhibitors
  • Orphan Drug Status Yes – Non-small cell lung cancer

Lorlatinib (PF-6463922) is an experimental anti-neoplastic drug in development by Pfizer. It is a orally-administered small molecule inhibitor of ROS1 and ALK.

In 2015, FDA granted Pfizer orphan drug status for lorlatinib for the treatment of non-small cell lung cancer.[1]

  • 05 Oct 2016 Massachusetts General Hospital plans a phase II trial for Non-small cell lung cancer (Late-stage disease, Metastatic disease) in USA (PO, unspecified formulation) (NCT02927340)
  • 01 Oct 2016 Pfizer completes a phase I trial in pharmacokinetic trial in Healthy volunteers in USA (NCT02804399)
  • 01 Aug 2016 Pfizer initiates a phase I drug-drug interaction trial in Healthy volunteers in Belgium (PO, unspecified formulation) (NCT02838264)

Figure

Structures of ALK inhibitors marketed or currently in the clinic

Synthesis

NEED COLOUR

Clinical studies

Several clinical trials are ongoing. A phase II trial comparing avelumab alone and in combination with lorlatinib or crizotinib for non-small cell lung cancer is expected to be complete in late 2017. A phase II trial comparing lorlatinib with crizotinib is expected to be complete in mid-2018.[2] A phase II trial for treatment of ALK-positive or ROS1-positive non-small cell lung cancer with CNA metastases is not expected to be complete until 2023.[3] Preclinical studies are investigating lorlatinib for treatment of neuroblastoma.

Lorlatinib is an investigational medicine that inhibits the anaplastic lymphoma kinase (ALK) and ROS1 proto-oncogene. Due to tumor complexity and development of resistance to treatment, disease progression is a challenge in patients with ALK-positive metastatic non-small cell lung cancer (NSCLC). A common site for progression in metastatic NSCLC is the brain. Lorlatinib was specifically designed to inhibit tumor mutations that drive resistance to other ALK inhibitors and to penetrate the blood brain barrier.

ABOUT LORLATINIB

ALK in NSCLC ROS1 in NSCLC PRECLINICAL DATA CLINICAL STUDIES Originally discovered as an oncogenic driver in a type of lymphoma, ALK gene alterations were also found to be among key drivers of tumor development in cancers, such as NSCLC.1 In ALK-positive lung cancer, a normally inactive gene called ALK is fused with another gene. This genetic alteration creates the ALK fusion gene and ultimately, the production of an ALK fusion protein, which is responsible for tumor growth.1,2 This genetic alteration is present in 3-5% of NSCLC patients.3,4,5 Another gene that can fuse with other genes is called ROS1. Sometimes a ROS1 fusion protein can contribute to cancer-cell growth and tumor survival. This genetic alteration is present in approximately 1% of NSCLC patients.5 Preclinical data showed lorlatinib is capable of overcoming resistance to existing ALK inhibitors and penetrated the blood brain barrier in ALK-driven tumor models.2 Specifically, in these preclinical models, lorlatinib had activity against all tested clinical resistance mutations in ALK.

A Phase 1/2 clinical trial of lorlatinib in patients with ALK-positive or ROS1-positive advanced NSCLC is currently ongoing. • The primary objective of the Phase 1 portion was to assess safety and tolerability of single-agent lorlatinib at increasing dose levels in patients with ALK-positive or ROS1-positive advanced NSCLC.6 • Data from the Phase 1 study showed that lorlatinib had promising clinical activity in patients with ALK-positive or ROS1- positive advanced NSCLC. Most of these patients had developed CNS metastases and had received ≥1 prior tyrosine kinase inhibitor.7 o The most common treatment-related adverse events (AEs) were hypercholesterolemia (69%) and peripheral edema (37%). Hypercholesterolemia was the most common (11%) grade 3 or higher treatment-related AE and the most frequent reason for dose delay or reduction. No patients discontinued due to treatment-related AEs. At the recommended Phase 2 dose, 4 out of 17 patients (24%) experienced a treatment-related AE of any grade that led to a dose delay or hold.

PATENT

WO2014207606

This invention relates to crystalline forms of the macrocyclic kinase inhibitor, (10R)-7-amino-12-fluoro-2, 10,16-trimethyl-15-OXO-10,15, 16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4, 3-?][2,5,1 1 ]benzoxadiazacyclotetradecine-3-carbonitrile, including crystalline solvates thereof, that may be useful in the treatment of abnormal cell growth, such as cancer, in mammals. The invention also relates to compositions including such crystalline forms, and to methods of using such compositions in the treatment of abnormal cell growth in mammals, especially humans.

Background of the Invention

The compound (10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2/-/-8,4-(metheno)pyrazolo[4,3- ?][2,5,1 1 ]benzoxadiazacyclotetradecine-3-carbonitrile, represented by the formula (I):

(I)

is a potent, macrocyclic inhibitor of both wild type and resistance mutant forms of anaplastic lymphoma kinase (ALK) and c-ros oncogene 1 (ROS1) receptor tyrosine kinase. Preparation of the free base compound of formula (I) as an amorphous solid is disclosed in International Patent Publication No. WO 2013/132376 and in United States Patent Publication No. 2013/0252961 , the contents of which are incorporated herein by reference in their entirety.

Human cancers comprise a diverse array of diseases that collectively are one of the leading causes of death in developed countries throughout the world (American Cancer Society, Cancer Facts and Figures 2005. Atlanta: American Cancer Society; 2005). The progression of cancers is caused by a complex series of multiple genetic and molecular events including gene mutations, chromosomal translocations, and karyotypic abnormalities (Hanahan & Weinberg, The hallmarks of cancer. Cell 2000; 100: 57-70). Although the underlying genetic causes of

cancer are both diverse and complex, each cancer type has been observed to exhibit common traits and acquired capabilities that facilitate its progression. These acquired capabilities include dysregulated cell growth, sustained ability to recruit blood vessels (i.e., angiogenesis), and ability of tumor cells to spread locally as well as metastasize to secondary organ sites (Hanahan & Weinberg 2000). Therefore, the ability to identify novel therapeutic agents that inhibit molecular targets that are altered during cancer progression or target multiple processes that are common to cancer progression in a variety of tumors presents a significant unmet need.

Receptor tyrosine kinases (RTKs) play fundamental roles in cellular processes, including cell proliferation, migration, metabolism, differentiation, and survival. RTK activity is tightly controlled in normal cells. The constitutively enhanced RTK activities from point mutation, amplification, and rearrangement of the corresponding genes have been implicated in the development and progression of many types of cancer. (Gschwind et al., The discovery of receptor tyrosine kinases: targets for cancer therapy. Nat. Rev. Cancer 2004; 4, 361-370; Krause & Van Etten, Tyrosine kinases as targets for cancer therapy. N. Engl. J. Med. 2005; 353: 172-187.)

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase, grouped together with leukocyte tyrosine kinase (LTK) to a subfamily within the insulin receptor (IR) superfamily. ALK was first discovered as a fusion protein with nucleophosmin (NPM) in anaplastic large cell lymphoma (ALCL) cell lines in 1994. (Morris et al., Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin’s lymphoma. Science 1994; 263:1281-1284.) NPM-ALK, which results from a chromosomal translocation, is implicated in the pathogenesis of human anaplastic large cell lymphoma (ALCL) (Pulford et al., Anaplastic lymphoma kinase proteins in growth control and cancer. J. Cell Physiol., 2004; 199: 330-58). The roles of aberrant expression of constitutively active ALK chimeric proteins in the pathogenesis of ALCL have been defined (Wan et. al., Anaplastic lymphoma kinase activity is essential for the proliferation and survival of anaplastic large cell lymphoma cells. Blood, 2006; 107:1617-1623). Other chromosomal rearrangements resulting in ALK fusions have been subsequently detected in ALCL (50-60%), inflammatory myofibroblastic tumors (27%), and non-small-cell lung cancer (NSCLC) (2-7%). (Palmer et al., Anaplastic lymphoma kinase: signaling in development and disease. Biochem. J. 2009; 420:345-361 .)

The EML4-ALK fusion gene, comprising portions of the echinoderm microtubule associated protein-like 4 (EML4) gene and the ALK gene, was first discovered in NSCLC archived clinical specimens and cell lines. (Soda et al., Identification of the transforming EML4-ALK fusion gene in non-small cell lung cancer. Nature 2007; 448:561-566; Rikova et al., Cell 2007; 131 :1 190-1203.) EML4-ALK fusion variants were demonstrated to transform NIH-3T3 fibroblasts and cause lung adenocarcinoma when expressed in transgenic mice, confirming the

potent oncogenic activity of the EML4-ALK fusion kinase. (Soda et al., A mouse model for EML4-ALK-positive lung cancer. Proc. Natl. Acad. Sci. U.S.A. 2008; 105:19893-19897.) Oncogenic mutations of ALK in both familial and sporadic cases of neuroblastoma have also been reported. (Caren et al., High incidence of DNA mutations and gene amplifications of the ALK gene in advanced sporadic neuroblastoma tumors. Biochem. J. 2008; 416:153-159.)

ROS1 is a proto-oncogene receptor tyrosine kinase that belongs to the insulin receptor subfamily, and is involved in cell proliferation and differentiation processes. (Nagarajan et al. Proc Natl Acad Sci 1986; 83:6568-6572). ROS is expressed, in humans, in epithelial cells of a variety of different tissues. Defects in ROS expression and/or activation have been found in glioblastoma, as well as tumors of the central nervous system (Charest et al., Genes Chromos. Can. 2003; 37(1): 58-71). Genetic alterations involving ROS that result in aberrant fusion proteins of ROS kinase have been described, including the FIG-ROS deletion translocation in glioblastoma (Charest et al. (2003); Birchmeier et al. Proc Natl Acad Sci 1987; 84:9270-9274; and NSCLC (Rimkunas et al., Analysis of Receptor Tyrosine Kinase ROS1 -Positive Tumors in Non-Small Cell Lung Cancer: Identification of FIG-ROS1 Fusion, Clin Cancer Res 2012; 18:4449-4457), the SLC34A2-ROS translocation in NSCLC (Rikova et al. Cell 2007;131 :1 190-1203), the CD74-ROS translocation in NSCLC (Rikova et al. (2007)) and cholangiocarcinoma (Gu et al. PLoS ONE 201 1 ; 6(1 ): e15640), and a truncated, active form of ROS known to drive tumor growth in mice (Birchmeier et al. Mol. Cell. Bio. 1986; 6(9):3109-31 15). Additional fusions, including TPM3-ROS1 , SDC4-ROS1 , EZR-ROS1 and LRIG3-ROS1 , have been reported in lung cancer patient tumor samples (Takeuchi et al., RET, ROS1 and ALK fusions in lung cancer, Nature Medicine 2012; 18(3):378-381).

The dual ALK/c-MET inhibitor crizotinib was approved in 201 1 for the treatment of patients with locally advanced or metastatic NSCLC that is ALK-positive as detected by an FDA-approved test. Crizotinib has also shown efficacy in treatment of NSCLC with ROS1 translocations. (Shaw et al. Clinical activity of crizotinib in advanced rson-smali cell lung cancer (NSCLC) harboring ROS1 gene rearrangement. Presented at the Annual Meeting of the American Society of Clinical Oncology, Chicago, June 1-5, 2012.) As observed clinically for other tyrosine kinase inhibitors, mutations in ALK and ROS1 that confer resistance to ALK inhibitors have been described (Choi et ai., EML4-ALK Mutations in Lung Cancer than Confer Resistance to ALK Inhibitors, N Engl J Med 2010; 363:1734-1739; Awad et ai., Acquired Resistance to Crizotinib from a Mutation in CD74-ROS1, Engl J Med 2013; 368:2395-2401 ).

Thus, ALK and ROS1 are attractive molecular targets for cancer therapeutic intervention. There remains a need to identify compounds having novel activity profiles against wild-type and mutant forms of ALK and ROS1 .

The present invention provides crystalline forms of the free base of (10R)-7-amino-12-fluoro-2, 10,16-trimethyl-15-OXO-10,15, 16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3- ?][2, 5,1 1 ]-benzoxadiazacyclotetradecine-3-carbonitrile having improved properties, such as improved crystallinity, dissolution properties, decreased hygroscopicity, improved mechanical properties, improved purity, and/or improved stability, while maintaining chemical and enantiomeric stability.

Comparative Example 1A

Preparation of (10f?)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3- ?l[2,5,1 Hbenzoxadiazacyclo-tetradecine-3-carbonitrile (amorphous)

Example 1A

Step 1 :

Palladium (II) acetate (53 mg, 0.24 mmol) and cataCXium® A (180 mg, 0.5 mmol) were mixed together in toluene (1 .5 mL, de-gassed) and the resulting solution was added via pipette to a stirred solution of compound 7 (0.9 g, 2.4 mmol), compound 15 (1 .0 g, 3.0 mmol) bis-pinacolato diboron (0.9 g, 3.6 mmol) and CsF (1 .9 g, 12.6 mmol) in MeOH/H20 (9:1 , 12 mL, degassed) at 60 °C. The resulting mixture was then stirred at reflux for 3 hrs. A further portion of Palladium (II) acetate (26 mg, 0.12 mmol) and cataCXium® A (90 mg, 0.25 mmol) in toluene (1 .5 mL, de-gassed) was added, and the yellow reaction mixture stirred at 60 °C overnight. After cooling to room temperature, the mixture was diluted with EtOAc (150 mL) and filtered through CELITE®. The filtrate was washed with water (100 mL), then brine (100 mL), dried (Na2S04) and evaporated. The residue was purified by flash chromatography over silica gel, which was eluted with 1 :1 EtOAc/cyclohexane, to give compound 22 as a yellow oil (570 mg, 43% yield). TLC (Rf = 0.40, 1 :1 EtOAc/cyclohexane). 1H NMR (400 MHz, CDCI3) δ 8.03 (m, 1 H), 7.65 (s, 1 H), 7.27 (dd,1 H, J = 9.9, 2.7 Hz), 7.01 (m, 1 H), 6.68 (m, 1 H), 6.40 (m, 1 H), 4.90 (br s, 2 H), 4.20 – 4.30 (m, 2 H), 3.96 (s, 3 H), 3.94 (s, 3 H), 2.55 – 2.85 (m, 3 H), 1 .68 (d, 3 H, J = 6.6 Hz), 1 .24 (s, 9 H). LCMS ES m/z 539 [M+H]+.

Step 2:

To a solution of compound 22 (69% purity, 0.95 g, assumed 1 .05 mmol) in MeOH (20 mL) was added a solution NaOH (1 .0 g, 25 mmol) in water (2 mL). The mixture was stirred at 40 °C for 3.5 hours. The reaction was diluted with water (80 mL), concentrated by 20 mL to remove MeOH on the rotary evaporator, and washed with MTBE (100 mL). The aqueous layer was then acidified carefully with 1 M aq HCI to approx. pH 2 (pH paper). Sodium chloride (15 g) was added to the mixture and the mixture was extracted with EtOAc (100 mL). The organic layer was separated, dried (Na2S04) and evaporated to give compound 23 as a pale yellow solid (480 mg, 87% yield). 1H NMR (400 MHz, CD3OD) δ 8.05 (m, 1 H), 7.45 (s, 1 H), 7.37 (dd,1 H, J = 10.4, 2.8 Hz), 7.10 (dt, 1 H, J = 8.5, 2.4 Hz), 6.50 – 6.60 (m, 2 H), 4.05 – 4.30 (m, 2 H), 3.99 (s, 3 H), 2.60 – 2.80 (m, 3 H), 1 .72 (d, 3 H, J = 6.5 Hz). LCMS ES m/z 525 [M+H]+.

Step 3:

A solution of HCI in dioxane (4 M, 6.0 mL) was added to a solution of compound 23

(480 mg, 0.91 mmol) in MeOH (methanol) (6 mL) and the reaction was stirred at 40 °C for 2.5 hours. The reaction mixture was then concentrated to dryness under reduced pressure. The residue was taken-up in MeOH (50 mL) and acetonitrile (100 mL) was added and the mixture was then again evaporated to dryness, to give compound 24 as an off white solid (400 mg, 87% yield). 1H NMR (400 MHz, CD3OD) δ 8.07 (dd, 1 H, J = 8.9. 5.9 Hz), 7.51 (d, 1 H, J = 1 .7 Hz), 7.42 (dd, 1 H, J = 9.8, 2.6 Hz), 7.23 (d, 1 H, J = 1 .6 Hz), 7.16 (dt, 1 H, J = 8.5, 2.7 Hz), 6.73 (dd, 1 H, J = 1 1 .9, 6.9 Hz), 4.22 (d, 1 H, J = 14.7 Hz), 4.14 (d, 1 H, J = 14.7 Hz), 4.07 (s, 3 H), 2.75 (s, 3 H), 1 .75 (d, 3 H, J = 5.5 Hz). LCMS ES m/z 425 [M+H]+.

Step 4:

A solution of compound 24 (400 mg, assumed 0.91 mmol) as the HCI salt and DIPEA

(diisopropylethylamine) (1 .17 g, 9.1 mmol) in DMF (dimethylformamide) (5.0 mL) and THF (0.5 mL) was added drop-wise to a solution of HATU (2-(1 H-7-azabenzotriazol-1 -yl)-1 ,1 ,3,3-tetramethyl uronium hexafluorophosphate methanaminium) (482 mg, 1 .27 mmol) in DMF (10.0 mL) at 0 °C over 30 minutes. After complete addition, the mixture was stirred at 0 °C for a further 30 mins. Water (70 mL) was added and the mixture was extracted into EtOAc (2 x 60 mL). The combined organics were washed with saturated aqueous NaHC03 (2 x 100 mL), brine (100 mL), dried over Na2S04, and evaporated. The residue was purified by column chromatography over silica gel, which was eluted with 70% EtOAc/cyclohexane giving 205 mg of a pale yellow residue (semi-solid). The solids were dissolved in MTBE (7 mL) and cyclohexane (20 mL) was added slowly with good stirring to precipitate the product. After stirring for 30 minutes, the mixture was filtered, and Example 1A was collected as an

amorphous white solid (1 10 mg, 29% yield). TLC (Rf = 0.40, 70% EtOAc in cyclohexane). 1H NMR (400 MHz, CDCI3) δ 7.83 (d, 1 H, J = 2.0 Hz), 7.30 (dd, 1 H, J = 9.6, 2.4 Hz), 7.21 (dd, 1 H, J = 8.4, 5.6 Hz), 6.99 (dt, 1 H, J = 8.0, 2.8 Hz), 6.86 (d, 1 H, J = 1 .2 Hz), 5.75 – 5.71 (m, 1 H), 4.84 (s, 2 H), 4.45 (d, 1 H, J = 14.4 Hz), 4.35 (d ,1 H, J = 14.4 Hz), 4.07 (s, 3 H), 3.13 (s, 3 H), 1 .79 (d, 3 H, J = 6.4Hz). LCMS ES m/z 407 [M+H]+.

Example 1

Preparation of crystalline hydrate of (10 ?)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo- 10,15,16,17-tetrahvdro-2/-/-8,4-(metheno)pyrazolo[4,3- ?l[2,5,1 Hbenzoxa-diazacyclo-tetradecine-3-carbonitrile (Form 1)

Example 1A Example 1

(amorphous) (Form 1 }

Amorphous (10f?)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3- ?][2,5,11 ]benzoxa-diazacyclo-tetradecine-3-carbonitrile free base, prepared as described in Example 1A (and Example 2 of United States Patent Publication No. 2013/0252961), was dissolved in 1 .0 : 1 .1 (v:v) H20:MeOH at a concentration of 22 mg/mL at 50°C, then allowed to cool to room temperature . This slurry was granulated for approximately 72 hours. The solids were isolated by filtration and vacuum dried overnight at 60°C to produce crystalline hydrate Form 1 of (10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-/?][2,5,1 1 ]benzoxadiazacyclotetradecine-3-carbonitrile.

Example 4

Alternative preparation of crystalline acetic acid solvate of (10 ?)-7-amino-12-fluoro-2, 10,16-trimethyl-15-OXO-10,15, 16,17-tetrahvdro-2H-8,4-(metheno)pyrazolo[4,3- ?U2,5, 1 1 lbenzoxa-diazacyclotetradecine-3-carbonitrile (Form 3)

Step 1 :

To a reaction vessel under N2 were charged compound 9 (9.97 kg, 17.95 mol), compound 21 (3.52 kg, 18.85 mol) and 2-methyltetrahydrofuran (97 L). Triethylamine (7.45 kg, 73.6 mol) was added while keeping the internal temperature below 35°C. The reaction mixture was held for 30 min and n-propylphosphonic anhydride (T3P), 50% solution in ethyl acetate (22.85 kg, 35.9 mol) was charged slowly, maintaining the internal temperature below 25°C. The reaction mixture was held at 20°C for at least 2 h until reaction was deemed complete. Ethyl acetate (35 L) and water (66 L) were added followed by 0.5N Hydrochloric acid solution (80 L). The aqueous layer was removed and the organic layer was washed with brine solution (80 L). The organic layer was concentrated and solvent exchanged with 2-methyl-2-butanol (80 L) give compound 25 (23 wt/wt%) solution in 2-methyl-2-butanol . This solution was carried forward to the next step directly in three batches, assuming 12.00 kg (100% yield) from this step.

Step 2:

2-Methyl-2-butanol (100 L) was combined with potassium acetate (1 .8 kg, 18.34 mol), palladium(ll) acetate (0.10 kg, 0.46 mol) and water (0.10 kg, 5.73 mol). The resulting mixture was purged with nitrogen. Di(1 -adamantyl)n-butylphosphine (0.23 kg, 0.43 mol) was added. An amount of 20% of compound 25 (3.97 kg active or 17.3 L of step 1 solution in 2-methyl-2-butanol) was added, and the resulting reaction mixture was heated at reflux for 2 h. The remaining solution of compound 25 in 2-methyl-2-butanol was subsequently added to the reaction over a period of 5 h. The resulting mixture was heated until the reaction was deemed complete (typically 16 – 20 h). This reaction step was processed in three batches, and the isolation was done in one single batch. Thus, the combined three batches were filtered through CELITE® to remove insoluble materials. The filtrate was concentrated to a low volume (approximately 20 L). Acetonitrile (60 L) was added. The resulting mixture was heated to reflux for 2 – 4 h, then cooled to RT for granulation. The resulting slurry was filtered to give compound 26 as a crude product. The crude product was combined with ethyl acetate (80 L) and Silicycle thiol (5 kg). The resulting mixture was heated for 2 h, cooled to RT and filtered. The filtrate was concentrated to approx. 20 L, and the resulting slurry was granulated and filtered. The filter cake was rinsed with ethyl acetate (4 L) and dried in a vacuum oven to give compound 26 as a pure product (4.74 kg, 43.5% overall last two steps). 1H NMR (CDCI3) δ 8.25 – 8.23 (m, 1 H), 7.28 (1 H, dd, 2.76 and 9.79 Hz), 7.22 (1 H, dd, 5.52 and 8.53 Hz), 7.18 (1 H, d, J = 1 .76 Hz), 7.01 (1 H, dt, J = 2.50 and 8.03 Hz), 5.78 – 5.70 (m, 1 H), 4.76 (1 H, d, J = 14.3 Hz), 4.13 (s, 3H), 3.16 (s, 3H), 1 .78 (d, 3H, J = 6.02 Hz), 1 .45 (s, 18H); 13C NMR (CDCI3) δ 167.0, 162.9, 160.4, 148.7, 146.3, 143.0, 140.7, 139.9, 135.5, 129.9, 129.8, 126.1 , 123.8, 123.5, 1 19.7, 1 13.8, 1 13.5, 1 1 1 .6, 108.1 , 81 .1 , 70.1 , 45.5, 37.0, 29.7, 26.0, 20.7; LCMS (M+1)+ 607.3, 507.1 , 451 .2.

Step 3:

To a reactor under N2 was added compound 26 (4.74 kg, 7.82 mol) and ethyl acetate (54 L). Hydrochloric acid 37% (5.19 L, 63.2 mol) was charged slowly while keeping the internal temperature below 25°C. The reaction mixture was stirred for 24 – 48 h until the reaction was complete. Ethyl acetate (54L) and water (54 L) were added. The reaction mixture was then treated with triethylamine until pH 8 – 9 was reached. The aqueous layer was removed and then the organic layer was washed water (2 x 54 L). The organic layer was concentrated under reduced pressure to approx. 54 L to give compound 27 (unisolated).

Step 4:

Acetic acid (1 .0 kg, 16.6 mol) was added to the organic layer containing compound 27. The reaction mixture was concentrated and then held for at least 3 h with stirring at RT. The resulted slurry was filtered. The filter cake was washed with ethyl acetate (2 L) and dried under vacuum to give 3.20 kg (87.8% yield) of Example 4 acetic acid solvate (Form 3). The spectroscopic data of this material was identical to that of an authentic sample of the crystalline acetic acid Form 3 of (10R)-7-amino-12-fluoro-2, 10, 16-trimethyl-15-oxo-10, 15,16, 17-tetrahydro-2/-/-8,4-(metheno)pyrazolo[4,3- ?][2,5,1 1 ]-benzoxadiazacyclo-tetradecine-3-carbonitrile prepared according to Example 3.

Preparation of Synthetic Intermediates

7 6 5

Step 1 :

A solution of (-)-DIPCI ((-)-B-chlorodiisopinocampheylborane) (57.1 g, 178 mmol) in THF

(tetrahydrofuran) (100 ml) was cooled to -20 to -30 °C. A solution of compound 1 (31 .3 g, 1 19 mmol) in THF (100 ml) was then added dropwise, via addition funnel (30 min addition). The reaction was left to warm up to room temperature (RT). After 2 h, the reaction was cooled to -30 °C and another portion of (-)-DIPCI (38.0 g, 1 19 mmol) was added. After 30 min, the reaction was allowed to warm to RT and after 1 h, the solvents were removed in vacuo and the residue re-dissolved in MTBE (methyl tertiary-butyl ether) (200 ml). A solution of diethanolamine (31 g, 296 mmol) in ethanol/THF (15 ml/30 ml) was added via addition funnel, to the reaction mixture under an ice bath. The formation of a white precipitate was observed. The suspension was heated at reflux for 2 hours then cooled to room temperature, filtered and the mother liquids concentrated in vacuo. The residue was suspended in heptane/EtOAc (7:3, 200 ml) and again

filtered. This procedure was repeated until no more solids could be observed after the liquids were concentrated. The final yellow oil was purified by column chromatography (eluent: cyclohexane/EtOAc 99:1 to 96:4). The resulting colorless oil was further purified by recrystallization from heptanes, to give alcohol compound 2 (25 g, 80% yield, 99% purity and 96% ee) as white crystals. 1H NMR (400 MHz, CDCI3) δ 7.73 (dd, 1 H), 7.32 (dd, 1 H), 6.74 (ddd, 1 H), 4.99 – 5.04 (m, 1 H), 2.01 (d, 1 H), 1 .44 (d, 3 H). LCMS-ES: No ionization, Purity 99%. Chiral GC (column CP-Chirasil-DexnCB): 96% ee; Rt (minor) 17.7 minutes and Rt (major) 19.4 minutes.

Step 2:

A solution of compound 2 (22 g, 83 mmol) in MTBE (350 mL) was cooled under an ice bath and triethylamine (23 mL, 166 mmol) followed by mesyl chloride (9.6 mL, 124 mmol) were added drop-wise. The reaction was then warmed to RT and stirred for 3 h. The reaction mixture was filtered and the solids washed with EtOAc. The mother liquids were concentrated in vacuo to give compound 3 (35 g, 80% yield) as a pale yellow oil. This material was taken into the following step without further purification. 1H NMR (400 MHz, CDCI3) δ 7.78 (dd, 1 H), 7.24 (dd, 1 H), 6.82 (ddd, 1 H), 2.92 (s, 3 H), 1 .64 (d, 3 H). LCMS-ES no ionization.

Step 3:

A suspension of Cs2C03 (65 g, 201 mmol) and compound 4 (13.3 g, 121 mmol) in 2-CH3-THF (2-methyitetrahydrofuran) (600 mL) and acetone (300 mL) was stirred at RT for 30 minutes then heated at 40 °C before drop-wise addition of a solution of compound 3 (34.4 g, 80 mmol) in 2-CH3-THF (300 mL) via addition funnel. The resulting mixture was left stirring at 75 -80 °C for 24 h. The reaction was then filtered through CELITE® with MTBE, the solvents removed in vacuo and the residue purified by column chromatography over silica gel which was eluted with cyclohexane/EtOAc (9:1 to 1 :1) to give compound 5 (14.3 g, 39 % yield, 90% ee) as a white solid. The solids were then re crystallized from heptane/EtOAc to give compound 5 (10.8 g, 37% yield, 95% ee). 1H NMR (400 MHz, CDCI3) 5 7.38 (dd, 1 H), 7.62 (dd, 1 H), 7.10 (dd, 1 H), 6.75 (ddd, 1 H), 6.44 – 6.51 (m, 2 H), 5.34 – 5.39 (m, 1 H), 4.73 (br s, 2 H), 1 .61 (d, 3 H). LCMS-ES m/z 359 [M+H]+. HPLC (Chiralpak IC 4.6 x 250 mm): 95% ee; Rt (minor) 10.4 minutes; Rt (major) 14.7 minutes; eluent: Heptane 80%/IPA 20% with 0.2% DEA, 0.7 mL/min. Step 4:

Compound 5 (20 g, 57 mmol) was dissolved in methanol (300 mL), and sequentially treated with triethylamine (TEA) (15.4 mL, 1 13 mmol) and PdCI2(dppf) (1 ,1 -bis(diphenylphosphino)ferrocene]dichloropalladium(ll) ) (4.1 g, 5.7 mmol). This mixture was heated at 100 °C for 16 hours, under a 100 psi carbon monoxide atmosphere. LCMS indicated consumption of starting material. The reaction mixture was filtered through a pad of CELITE®, and the filtrate evaporated to a brown oil. The crude product was purified by flash

chromatography over silica gel which was eluted with 50% to 75% ethyl acetate in cyclohexane, affording the pure product 6 as a brick-red solid (13.0 g, 79% yield). 1H NMR (400 MHz, CDCI3) δ 1 .65 (d, 3 H), 3.94 (s, 3 H), 4.75 (br s, 2 H), 6.32 (q, 1 H), 6.42 (dd, 1 H), 6.61 (dd, 1 H), 7.00 (ddd, 1 H), 7.28 (dd, 1 H), 7.60 (dd, 1 H), 8.03 (dd, 1 H). LCMS ES m/z 291 for [M+H]+.

Step 5:

Compound 6 (13.0 g, 45 mmol) was dissolved in acetonitrile (195 mL), and cooled to <10 °C in an ice water bath. NBS (N-bromosuccinimide) (7.9 g, 45 mmol) was added drop-wise to the cooled reaction mixture as a solution in acetonitrile (195 mL), monitoring the internal temperature to ensure it did not rise above 10 °C. After addition was complete, the mixture was stirred for 15 minutes. Thin layer chromatography (TLC) (1 :1 cyclohexane/ethyl acetate) showed consumption of starting material. The reaction mixture was evaporated, and the residue redissolved in ethyl acetate (400 mL), and washed with 2M aqueous NaOH (2 x 300 mL), and 10% aqueous sodium thiosulfate solution (300 mL). The organic extracts were dried over MgS04, and evaporated to a red oil (17.6 g). The crude product was purified over silica gel, which was eluted with 10% to 50% ethyl acetate in cyclohexane, which gave compound 7 (12.0 g, 73% yield). 1H NMR (400 MHz, CDCI3) δ 1 .65 (d, 3 H), 3.96 (s, 3 H), 4.74 – 4.81 (br s, 2 H), 6.33 (q, 1 H), 6.75 (d, 1 H), 7.03 (ddd, 1 H), 7.25 (dd, 1 H), 7.66 (d, 1 H), 8.06 (dd, 1 H). LCMS ES m/z 369/371 [M+H]+. A Chiralpak AD-H (4.6 x 100 mm, 5 micron) column was eluted with 10% MeOH (0.1 % DEA) in C02 at 120 bar. A flow rate of 5.0 mL/min gave the minor isomer Rt 0.6 minutes and the major isomer Rt 0.8 minutes (99% ee). Optical rotation: [ ]d20 = -92.4 deg (c=1 .5, MeOH).

Preparation of (/?)-methyl 2-(1 -((N,N-di-Boc-2-amino-5-bromopyridin-3-yl)oxy)ethyl)-4-fluorobenzoic acid (9)

7

Step 1 :

To a solution of compound 7 (2000 g, 5.4 mol) in dry DCM (dichloromethane) (32000 mL) was added DIPEA (N.N-dsisopropyleibylamine) (2100 g, 16.28 mol) and DMAP (4-dimethylaminopyridine) (132 g, 1 .08 mol). Then Boc20 (di-tert-butyl-dicarbonate) (3552 g, 16.28 mol) was added to the mixture in portions. The reaction was stirred at RT for overnight. TLC (petroleum ether/EtOAc =5:1) show the reaction was complete, the mixture was washed with sat. NH4CI (15 L) two times, then dried over Na2S04and concentrated to give a crude product which was purified by column (silica gel, petroleum ether/EtOAc from 20:1 to 10:1) to give compound 8 (2300 g, 75%) as a white solid.

Step 2:

Compound 8 (50 g, 87.81 mmol, 100 mass%) was charged to a round bottom flask (RBF) containing tetrahydrofuran (12.25 mol/L) in Water (5 mL/g, 3060 mmol, 12.25 mol/L) and sodium hydroxide (1 mol/L) in Water (1 .5 equiv., 131 .7 mmol, 1 mol/L). The biphasic mixture was stirred at RT for 14 hours. 1 N HCI was added to adjust pH to < 2. THF was then removed by vacuum distillation. The product precipitated out was collected by filtration. The filter cake was rinsed with water, pulled dried then dried in vacuum oven to constant weight (48 h, 55°C, 25 mbar). 48.3g isolated, 99% yield. 1H NMR (CDCI3, 400MHz) δ 8.24 (1 H, dd, 1 H, J = 5.76 and 3.0 Hz), 8.16 (1 H, d, J = 2.0 Hz), 7.37 (1 H, dd, J = 2.5 and 9.8 Hz), 7.19 (1 H, d, J = 2 Hz), 7.14 – 7.06 (1 H, m), 6.50 (1 H, q, J = 6.3 Hz), 1 .67 (3H, d, J = 8.4 Hz), 1 .48 (18H, s). 13C NMR (CDCI3, 100 MHz), δ 170.1 , 169.2, 167.6, 165.1 , 150.6, 149.2, 148.6, 141 .4, 140.7, 135.2, 135.1 , 124.2, 122.2,122.1 , 1 19.9, 1 15.4, 1 15.1 , 1 13.4, 1 13.2, 100.0, 83.4, 73.3, 27.9, 23.9. LCMS (M+ +1) 557.2, 555.3, 457.1 , 455.1 , 401 , 0, 399.0.

Step 1 :

Ethyl 1 ,3-dimethylpyrazole-5-carboxylate (5.0 g, 30 mmol) was dissolved in 1 ,2-dichloroethane (200 mL), followed by addition of NBS (5.3 g, 30 mmol) and dibenzoyi peroxide (727 mg, 3.0 mmol), in small portions and stirred at 85 °C for 2 hours. The mixture was allowed to cool, diluted to 400 mL with dichloromethane, and washed with water (2 x 200 mL). The organic layer was dried over MgS04, and evaporated to give compound 10 (4.1 g, 42% yield). TLC (EtOAc/Cyclohexane; 1 :10; KMn04): Rf~0.3. 1H NMR (400 MHz, CDCI3) δ 4.47 (s, 2 H), 4.41 (q, 2 H), 4.15 (s, 3 H), 1 .42 (t, 3 H). LCMS ES m/z 324/326/328 [M+H]+.

Step 2:

Compound 10 (3.0 g, 9.2 mmol) was dissolved in methylamine solution (33% solution in ethanol, 70 mL), and stirred at RT for 16 hours. The mixture was evaporated to give compound 11 (1 .8 g, 71 % yield). 1H NMR (400 MHz, CDCI3) δ 4.39 (q, 2 H), 4.14 (s, 3 H), 4.05 (s, 2 H), 2.62 (d, 3 H), 1 .41 (t, 3 H). LCMS ES m/z 276/278 [M+H]+.

Step 3:

Compound 11 (1 .8 g, 6.5 mmol) was dissolved in dichloromethane (20 mL), and the mixture cooled to 0 °C. A solution of di(fe/?-butyl) dicarbonate (1 .75 g, 8 mmol) in dichloromethane (17.5 mL) was added dropwise. The ice bath was removed and the mixture stirred for 18 hours at room temperature. The mixture was diluted to 100 mL with dichloromethane, and washed with water (2 x 50 mL). Organic extracts were dried over magnesium sulfate, and evaporated to give compound 12 (1 .8 g, 72% yield). 1H NMR (400 MHz, CDCI3) δ 4.48 – 4.44 (m, 2 H), 4.41 (q, 2 H), 4.12 (s, 3 H), 2.82 – 2.79 (m, 3 H), 1 .47 (s, 9 H), 1 .41 (t, 3 H). LCMS ES m/z 376/378 [M+H]+ and 276/278 [M-BOC]+.

Step 4:

Compound 12 (4 g, 1 1 mmol) was dissolved in dioxane (43 mL). Sodium amide (1 g, 27 mmol) was added in one portion. The reaction mixture was stirred at 100 °C for 24 h. After this time, the solvent was removed under reduced pressure to give a white solid. The material was suspended in EtOAc (100 mL) and washed with 5% citric acid solution (100 mL). The organic phase was separated and washed with water (100 mL), dried over MgS04, filtered and the solvent removed in vacuo to give compound 13 as a yellow gum (3.1 g, 84% yield). 1H NMR (400 MHz, DMSO-c/6) δ 4.27 (s, 2 H), 3.92 (s, 3 H), 2.70 (s, 3 H), 1 .40 (s, 9 H). LCMS ES m/z 348/350 [M+H]+ and 248/250 [M-BOC]+.

Step 5:

Compound 13 (3 g, 8.6 mmol) was dissolved in DMF (43 mL, 0.2 M). HOBt (1 .2 g, 8.6 mmol) was added, followed by ammonium chloride (0.9 g, 17.2 mmol). EDCI (2.5 g, 13 mmol) was then added, followed by TEA (2.4 mL, 17 mmol). The reaction mixture was stirred at room temperature. After 18h, the solvent was removed under reduced pressure to give a yellow oil

(8.0 g). The residue was dissolved in EtOAc (75ml_). The organic phase was washed with NaHC03 (sat. solution, 70 ml_) and then brine (100 ml_). The combined organic layers were dried over MgS04 and the solvent removed in vacuo to give compound 14 as a dark yellow oil (2.7 g, 91 % yield). This material was used directly in the next step without further purification. 1H NMR (400 MHz, CDCI3) δ 6.74 (br s, 1 H), 5.95 (br s, 1 H), 4.49 (br s, 2 H), 4.16 (s, 3 H), 2.81 (br s, 3 H), 1 .47 (s, 9 H). LCMS ES m/z 347/349 [M+H]+ and 247/249 [M-BOC]+.

Step 6:

Compound 14 (2.7 g, 7.9 mmol) was dissolved in DCM (80 ml_, 0.1 M). TEA (3.3 ml_, 23.8 mmol) was then added and the reaction mixture cooled down to -5 °C. Trifluoroacetic anhydride (2.2 ml_, 15.8 mmol) in DCM (15 ml_) was added dropwise over 30 min. After addition, the reaction mixture was stirred at 0 °C for 1 h. After this time, the solvents were removed under reduced pressure to give a dark yellow oil. This residue was diluted in DCM (100 ml_), washed with 5% citric acid, sat. NaHC03and brine, dried over MgS04, filtered and the solvents removed in vacuo to give a dark yellow oil (2.6 g). The crude product was purified by reverse phase chromatography to give compound 15 as a yellow oil (2.3 g, 87% yield). 1H NMR (400 MHz, CDCI3) δ 4.46 (br s, 2 H), 4.01 (s, 3 H), 2.83 (br s, 3 H), 1 .47 (s, 9 H). LCMS ES m/z 331 /329 [M+H]+ and 229/231 [M-BOC]+ as the base ion.

Preparation o/: 1 -methyl-3-((methylamino)methyl)-1 H-pyrazole-5-carbonitrile (21)

Step 1 :

To /V-benzylmethylamine (2.40 kg, 19.8 mol) and ethyldiisopropylamine (2.61 kg, 20.2 mol) in acetonitrile (6 L) at 16°C was added chloroacetone (1 .96 kg, 21 .2 mol) over 60 mins [exothermic, temp kept <30°C]. The mixture was stirred at 22°C for 18 hours then concentrated to an oily solid. The residue was triturated with MTBE (5 L), and then filtered through a pad of CELITE® (600 g, top) and silica (1 .5 kg, bottom), washing with MTBE (8 L). The filtrate was evaporated to afford compound 16 (3.35 kg, 18.9 mol, 95%) as a brown oil.

Step 2:

Compound 16 (1 .68 kg, 9.45 mol), Boc-anhydride (2.1 kg, 9.6 mol) and 20wt% Pd/C (50% H20, 56 g) in ethanol (5 L) were hydrogenated in an 1 1 -L autoclave at 50 psi [exotherm to 40°C with 20°C jacket]. The atmosphere became saturated with carbon dioxide during the reaction and so needed to be vented and de-gassed twice to ensure sufficient hydrogen uptake and completion of the reaction. The total reaction time was ~1 .5 hours. Two runs (for a total of 18.9 mol) were combined and filtered through a pad of SOLKA-FLOC®, washing with methanol. The filtrate was treated with DMAP (45 g, 0.37 mol) and stirred at room temperature overnight to destroy the excess Boc-anhydride. The mixture was then concentrated to dryness, dissolved in MTBE (6 L) and filtered through a pad of magnesol (1 kg), washing with MTBE (4 L). The filtrate was evaporated to afford compound 17 (3.68 kg, ~95 wt%, 18.7 mol, 99%) as an orange-brown oil.

Step 3:

To compound 17 (3.25 kg, -95 wt%, 16.5 mol) and diethyl oxalate (4.71 kg, 32.2 mol) in methanol (12 L) at 15°C was added 25 wt% sodium methoxide in methanol (6.94 kg, 32.1 mol) over 25 mins [temp kept <25°C]. The mixture was stirred at 20°C for 16 hours then cooled to -37°C and 37% hydrochloric acid (3.1 kg, 31 mol) was added over 5 mins [temp kept <-10°C]. The mixture was cooled to -40°C and methylhydrazine (1 .42 kg, 30.8 mol) was added over 7 mins [temp kept <-17°C]. The mixture was warmed to 5°C over 90 minutes, then re-cooled to 0°C and quenched by addition of 2.4M KHS04 (6.75 L, 16.2 mol) in one portion [exotherm to 27°C]. The mixture was diluted with water (25 L) and MTBE (15 L), and the layers separated. The organic layer was washed with brine (7 L) and the aqueous layers then sequentially re-extracted with MTBE (8 L). The combined organics were evaporated and azeotroped with toluene (2 L) to afford crude compound 18. Chromatography (20 kg silica, 10-40% EtOAc in hexane) afforded compound 18 (3.4 kg, ~95 wt%, 11 .4 mol, 69%) as an orange oil.

Step 4:

Ammonia (3 kg, 167 mol) was bubbled in to cooled methanol (24 L) [temp kept <18°C]. A solution of compound 18 (4.8 kg, ~95 wt%, 16.1 mol) in methanol (1 .5 L) was added over 30 minutes and the mixture stirred at 25°C for 68 hours and then at 30°C for 24 hours. Two runs (from a total of 9.68 kg of ~95 wt% Step 3) were combined and concentrated to ~13 L volume. Water (30 L) was slowly added over 80 minutes, keeping the temperature 30 to 40°C. The resulting slurry was cooled to 20°C, filtered, washed with water (12 L) and pulled dry on the filter overnight. The solids were triturated in MTBE (8 L) and hexane (8 L) at 45°C then re-cooled to 15°C, filtered, washed with hexane (4 L) and dried under vacuum to afford compound 19 (7.95 kg, 29.6 mol, 90%) as an off-white solid.

Step 5:

To compound 19 (7.0 kg, 26.1 mol) in DCM (30 L) at 0°C was added triethylamine (5.85 kg, 57.8 mol). The mixture was further cooled to -6°C then trifluoroacetic anhydride (5.85 kg, 27.8 mol) added over 90 minutes [temp kept 0 to 5°C]. TLC assay showed the reaction was incomplete. Additional triethylamine (4.1 kg, 40.5 mol) and trifluoroacetic acid (4.1 kg, 19.5 mol) were added over 2 hours until TLC showed complete reaction. The reaction mixture was quenched in to water (40 L) [temp to 23°C]. The layers were separated and the aqueous re-extracted with DCM (8 L). The organic layers were sequentially washed with brine (7 L), filtered through a pad of silica (3 kg) and eluted with DCM (10 L). The filtrate was evaporated and chromatographed (9 kg silica, eluent 10-30% EtOAc in hexane). Product fractions were evaporated and azeotroped with IPA to afford compound 20 (6.86 kg, -94 wt%, 25.8 mol, 99%) as an orange oil.

Step 6:

To compound 20 (6.86 kg, -94 wt%, 25.8 mol) in IPA (35 L) at 17°C was added 37% hydrochloric acid (6.4 L, 77.4 mol). The mixture was heated to 35°C overnight then concentrated to a moist solid and residual water azeotroped with additional IPA (8 L). The resulting moist solid was triturated with MTBE (12 L) at 45°C for 30 minutes then cooled to 20°C and filtered, washing with MTBE (5 L). The solids were dried under vacuum at 45°C to afford compound 21 (4.52 kg, 24.2 mol, 94%) as a white solid. 1H-NMR was consistent with desired product; mp 203-205°C; HPLC 99.3%. 1H NMR (CD3OD, 400 MHz) δ 7.12 (1 H, s), 4.28 (2H, s), 4.09 (3H, s), 2.77 (3H, s). 13C NMR (CD3OD, 100 MHz) δ 144.5, 177.8, 1 14.9, 110.9, 45.9, 39.0, 33.2. LCMS (M++1) 151 .1 , 138.0, 120.0.

PATENT

WO2013132376

PATENT

WO 2016089208

PATENT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017021823&redirectedID=true

Preparation of the free base of lorlatinib as an amorphous solid is disclosed in

International Patent Publication No. WO 2013/132376 and in United States Patent No. 8,680,1 1 1 . Solvated forms of lorlatinib free base are disclosed in International Patent Publication No. WO 2014/207606.

Example 1

Lab Scale Preparation of Form 7 of (10 ?)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2/-/-8,4-(metheno)pyrazolo[4,3- ?l[2,5,1 l lbenzoxadiazacyclotetra-decine- -carbonitrile (lorlatinib) Free Base

[AcOH solvate]

Form 7 of lorlatinib free base was prepared by de-solvation of the acetic acid solvate of lorlatinib (Form 3), prepared as described in International Patent Publication No. WO 2014/207606, via an intermediate methanol solvate hydrate form of lorlatinib (Form 2).

The acetic acid solvate of lorlatinib (Form 3) (5 g, 10.72 mmol) was slurried in methanol

(10 mL/g, 1235.9 mmol) at room temperature in an Easymax flask with magnetic stirring to which triethylamine (1 .2 equiv., 12.86 mmol) was added over 10 minutes. The resulting solution was heated to 60°C and water (12.5 mL/g, 3469.3 mmol) was added over 10 minutes, while maintaining a temperature of 60°C. Crystallization was initiated by scratching the inside of the glass vessel to form a rapidly precipitating suspension which was triturated to make the system mobile. The suspension was then cooled to 25°C over 1 hour, then cooled to 5°C and granulated for 4 hours. The white slurry was filtered and washed with 1 mL/g chilled

water/methanol (1 :1) then dried under vacuum at 50°C overnight to provide the methanol solvate hydrate Form 2 of lorlatinib.

Form 7 was then prepared via a re-slurry of the methanol solvate hydrate Form 2 of lorlatinib in heptane. 100 mg of lorlatinib Form 2 was weighed into a 4-dram vial and 3 mL of heptane was added. The mixture was slurried at room temperature on a roller mixer for 2 hours. Form conversion was confirmed by PXRD revealing complete form change to Form 7 of lorlatinib free base.

Paper

http://pubs.acs.org/doi/abs/10.1021/jm500261q

*E-mail: ted.w.johnson@pfizer.com. Phone: (858) 526-4683., *E-mail: paul.f.richardson@pfizer.com. Phone: (858) 526-4290.

Abstract Image

Although crizotinib demonstrates robust efficacy in anaplastic lymphoma kinase (ALK)-positive non-small-cell lung carcinoma patients, progression during treatment eventually develops. Resistant patient samples revealed a variety of point mutations in the kinase domain of ALK, including the L1196M gatekeeper mutation. In addition, some patients progress due to cancer metastasis in the brain. Using structure-based drug design, lipophilic efficiency, and physical-property-based optimization, highly potent macrocyclic ALK inhibitors were prepared with good absorption, distribution, metabolism, and excretion (ADME), low propensity for p-glycoprotein 1-mediated efflux, and good passive permeability. These structurally unusual macrocyclic inhibitors were potent against wild-type ALK and clinically reported ALK kinase domain mutations. Significant synthetic challenges were overcome, utilizing novel transformations to enable the use of these macrocycles in drug discovery paradigms. This work led to the discovery of 8k (PF-06463922), combining broad-spectrum potency, central nervous system ADME, and a high degree of kinase selectivity.

Discovery of (10R)-7-Amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile (PF-06463922), a Macrocyclic Inhibitor of Anaplastic Lymphoma Kinase (ALK) and c-ros Oncogene 1 (ROS1) with Preclinical Brain Exposure and Broad-Spectrum Potency against ALK-Resistant Mutations

La Jolla Laboratories, Pfizer Worldwide Research and Development, 10770 Science Center Drive, San Diego, California 92121, United States
J. Med. Chem., 2014, 57 (11), pp 4720–4744
DOI: 10.1021/jm500261q
(10R)-7-Amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]benzoxadiazacyclotetradecine-3-carbonitrile (8k)
white solid:
TLC Rf = 0.40 (70% EtOAc in cyclohexane);
LC–MS (ESI), m/z 407.1 [M + H]+;
1H NMR (400 MHz, CDCl3) δ 7.83 (d, J = 2.0 Hz, 1 H), 7.30 (dd, J = 9.6, 2.4 Hz, 1 H), 7.21 (dd, J = 8.4, 5.6 Hz, 1 H), 6.99 (dt, J = 8.0, 2.8 Hz, 1 H), 6.86 (d, J = 1.2 Hz, 1 H), 5.75–5.71 (m, 1 H), 4.84 (s, 2 H), 4.45 (d, J = 14.4 Hz, 1 H), 4.35 (d, J = 14.4 Hz, 1 H), 4.07 (s, 3 H), 3.13 (s, 3 H), 1.79 (d, J = 6.4 Hz, 3 H).

References

1H NMR PREDICT

13C NMR PREDICT

Lorlatinib
Lorlatinib.svg
Clinical data
Routes of
administration
PO
Legal status
Legal status
  • experimental
Identifiers
CAS Number 1454846-35-5
ChemSpider 32813339
Chemical and physical data
Formula C22H20FN5O2
Molar mass 405.43 g·mol−1
3D model (Jmol) Interactive image

///////////////////Lorlatinib, PF-6463922,  anti-neoplastic,  Pfizer,  ROS1,  ALK, phase 2, UNII:OSP71S83EU, лорлатиниб لورلاتينيب 洛拉替尼 Orphan Drug, PF 6463922

Fc2ccc3C(=O)N(C)Cc1nn(C)c(C#N)c1c4cc(O[C@H](C)c3c2)c(N)nc4


Filed under: 0rphan drug status, Phase2 drugs, Uncategorized Tagged: ALK, anti-neoplastic, лорлатиниб, Lorlatinib, Orphan Drug, PF-6463922, PFIZER, phase 2, ROS1, UNII:OSP71S83EU, لورلاتينيب, 洛拉替尼

Award for me, 100 Most Impactful Health care Leaders Global listing

$
0
0

str6

 

At award function for my award “100 Most Impactful Health care Leaders Global listing”, conferred on me at Taj lands end, Mumbai, India on 14 Feb 2014 by World Health Wellness congress and awards

 


Filed under: AWARD, Uncategorized Tagged: Anthony crasto, award

FDA approves new psoriasis drug Siliq (brodalumab)

$
0
0

FDA approves new psoriasis drug

The U.S. Food and Drug Administration today approved Siliq (brodalumab) to treat adults with moderate-to-severe plaque psoriasis. Siliq is administered as an injection.

Read more.

For Immediate Release

February 15, 2017

Release

The U.S. Food and Drug Administration today approved Siliq (brodalumab) to treat adults with moderate-to-severe plaque psoriasis. Siliq is administered as an injection.

Siliq is intended for patients who are candidates for systemic therapy (treatment using substances that travel through the bloodstream, after being taken by mouth or injected) or phototherapy (ultraviolet light treatment) and have failed to respond, or have stopped responding to other systemic therapies.

“Moderate-to-severe plaque psoriasis can cause significant skin irritation and discomfort for patients, and today’s approval provides patients with another treatment option for their psoriasis,” said Julie Beitz, M.D., director of the Office of Drug Evaluation III in the FDA’s Center for Drug Evaluation and Research. “Patients and their health care providers should discuss the benefits and risks of Siliq before considering treatment.”

Psoriasis is a skin condition that causes patches of skin redness and flaking. Psoriasis is an autoimmune disorder that occurs more commonly in patients with a family history of the disease, and most often begins in people between the ages of 15 and 35. The most common form of psoriasis is plaque psoriasis, in which patients develop thick, red skin with flaky, silver-white scales.

Siliq’s active ingredient (brodalumab) binds to a protein that causes inflammation, inhibiting the inflammatory response that plays a role in the development of plaque psoriasis.

Siliq’s safety and efficacy were established in three randomized, placebo-controlled clinical trials with a total of 4,373 adult participants with moderate-to-severe plaque psoriasis who were candidates for systemic therapy or phototherapy. More patients treated with Siliq compared to placebo had skin that was clear or almost clear, as assessed by scoring of the extent, nature and severity of psoriatic changes of the skin.

Suicidal ideation and behavior, including completed suicides, have occurred in patients treated with Siliq during clinical trials. Siliq users with a history of suicidality or depression had an increased incidence of suicidal ideation and behavior compared to users without this history. A causal association between treatment with Siliq and increased risk of suicidal ideation and behavior has not been established.

Because of the observed risk of suicidal ideation and behavior, the labeling for Siliq includes a Boxed Warning and the drug is only available through a restricted program under a Risk Evaluation and Mitigation Strategy (REMS) called the Siliq REMS Program. Notable requirements of the Siliq REMS Program include the following:

  • Prescribers must be certified with the program and counsel patients about this risk. Patients with new or worsening symptoms of depression or suicidality should be referred to a mental health professional, as appropriate.
  • Patients must sign a Patient-Prescriber Agreement Form and be made aware of the need to seek medical attention should they experience new or worsening suicidal thoughts or behavior, feelings of depression, anxiety or other mood changes.
  • Pharmacies must be certified with the program and must only dispense to patients who are authorized to receive Siliq.

Siliq is also approved with a Medication Guide to inform patients of the risk of suicidal ideation and behavior, and that because Siliq is a medication that affects the immune system, patients may have a greater risk of getting an infection, or an allergic or autoimmune condition. Patients with Crohn’s disease should not use Siliq. Health care providers should also evaluate patients for tuberculosis (TB) infection prior to initiating treatment with Siliq. Health care providers should not administer Siliq to patients with active TB infection, and should avoid immunizations with live vaccines in patients being treated with Siliq.

The most common adverse reactions reported with the use of Siliq include joint pain (arthralgia), headache, fatigue, diarrhea, throat pain (oropharyngeal pain), nausea, muscle pain (myalgia), injection site reactions, influenza, low white blood cell count (neutropenia) and fungal (tinea) infections.

Siliq is marketed by Bridgewater, New Jersey-based Valeant Pharmaceuticals.


Filed under: FDA 2017 Tagged: Brodalumab, siliq

Deflazacort

$
0
0

Deflazacort structure.svgChemSpider 2D Image | Deflazacort | C25H31NO6

Deflazacort

  • CAS 14484-47-0
  • Molecular Formula C25H31NO6
  • Average mass 441.517 Da
(11b,16b)-21-(Acetyloxy)-11-hydroxy-2′-methyl-5’H-pregna-1,4-dieno[17,16-d]oxazole-3,20-dione
11b,21-Dihydroxy-2′-methyl-5’bH-pregna-1,4-dieno[17,16-d]oxazole-3,20-dione 21-acetate
2-[(4aR,4bS,5S,6aS,6bS,9aR,10aS,10bS)-5-Hydroxy-4a,6a,8-trimethyl-2-oxo-2,4a,4b,5,6,6a,9a,10,10a,10b,11,12-dodecahydro-6bH-naphtho[2′,1′:4,5]indeno[1,2-d][1,3]oxazol-6b-yl]-2-oxoethyl acetate
  • 5’βH-Pregna-1,4-dieno[17,16-d]oxazole-3,20-dione, 11β,21-dihydroxy-2′-methyl-, 21-acetate (8CI)
  • (11β,16β)-21-(Acetyloxy)-11-hydroxy-2′-methyl-5’H-pregna-1,4-dieno[17,16-d]oxazole-3,20-dione
  • 2H-Naphth[2′,1′:4,5]indeno[1,2-d]oxazole, 5’H-pregna-1,4-dieno[17,16-d]oxazole-3,20-dione deriv.
  • Azacort
  • Azacortinol
  • Calcort
  • DL 458IT
  • Deflan
Optical Rotatory Power +62.3 ° Conc: 0.5 g/100mL; Solv: chloroform (67-66-3); Wavlength: 589.3 nm

…………..REF, “Drugs – Synonyms and Properties” data were obtained from Ashgate Publishing Co. (US)Hoechst Marion Roussel (now Aventis Pharma) has developed and launched Deflazacort (Dezacor; Flantadin; Lantadin; Calcort) a systemic corticosteroid developed for the treatment of a variety of inflammatory conditions .

In March 1990, the drug was approved in Spain, and by January 2013, the drug had been launched by FAES Farma . By the end of 1999, the product had been launched in Germany, Italy, Belgium, Switzerland and South Korea

Deflazacort is a corticosteroid first launched in 1985 by Guidotti in Europe for the oral treatment of allergic asthma, rheumatoid arthritis, arthritis, and skin allergy.

In 2017, an oral formulation developed at Marathon Pharmaceuticals was approved by the FDA for the treatment of Duchenne’s muscular dystrophy in patients 5 years of age and older.

Deflazacort (trade name Emflaza or Calcort among others) is a glucocorticoid used as an anti-inflammatory and immunosuppressant.

In 2013, orphan drug designation in the U.S. was assigned to the compound for the treatment of Duchenne’s muscular dystrophy. In 2015, additional orphan drug designation in the U.S. was assigned for the treatment of pediatric juvenile idiopathic arthritis (JIA) excluding systemic JIA.

Also in 2015, deflazacort was granted fast track and rare pediatric disease designations in the U.S. for the treatment of Duchenne’s muscular dystrophy.

Deflazacort is a glucocorticoid used as an anti-inflammatory and immunosuppressant. It was approved in February, 2017 by the FDA for use in treatment of Duchenne muscular dystrophy (trade name Emflaza).
  • Aventis Pharma (Originator), Lepetit (Originator), Guidotti (Licensee), Shire Laboratories (Licensee)

Image result for deflazacort

February 9, 2017 FDA approved

The U.S. Food and Drug Administration today approved Emflaza (deflazacort) tablets and oral suspension to treat patients age 5 years and older with Duchenne muscular dystrophy (DMD), a rare genetic disorder that causes progressive muscle deterioration and weakness. Emflaza is a corticosteroid that works by decreasing inflammation and reducing the activity of the immune system.

Corticosteroids are commonly used to treat DMD across the world. This is the first FDA approval of any corticosteroid to treat DMD and the first approval of deflazacort for any use in the United States.

Image result for Deflazacort

“This is the first treatment approved for a wide range of patients with Duchenne muscular dystrophy,” said Billy Dunn, M.D., director of the Division of Neurology Products in the FDA’s Center for Drug Evaluation and Research. “We hope that this treatment option will benefit many patients with DMD.”

DMD is the most common type of muscular dystrophy. DMD is caused by an absence of dystrophin, a protein that helps keep muscle cells intact. The first symptoms are usually seen between 3 and 5 years of age and worsen over time. The disease often occurs in people without a known family history of the condition and primarily affects boys, but in rare cases it can affect girls. DMD occurs in about one of every 3,600 male infants worldwide.

People with DMD progressively lose the ability to perform activities independently and often require use of a wheelchair by their early teens. As the disease progresses, life-threatening heart and respiratory conditions can occur. Patients typically succumb to the disease in their 20s or 30s; however, disease severity and life expectancy vary.

The effectiveness of deflazacort was shown in a clinical study of 196 male patients who were 5 to 15 years old at the beginning of the trial with documented mutation of the dystrophin gene and onset of weakness before age 5. At week 12, patients taking deflazacort had improvements in a clinical assessment of muscle strength across a number of muscles compared to those taking a placebo. An overall stability in average muscle strength was maintained through the end of study at week 52 in the deflazacort-treated patients. In another trial with 29 male patients that lasted 104 weeks, deflazacort demonstrated a numerical advantage over placebo on an assessment of average muscle strength. In addition, although not statistically controlled for multiple comparisons, patients on deflazacort appeared to lose the ability to walk later than those treated with placebo.

The side effects caused by Emflaza are similar to those experienced with other corticosteroids. The most common side effects include facial puffiness (Cushingoid appearance), weight gain, increased appetite, upper respiratory tract infection, cough, extraordinary daytime urinary frequency (pollakiuria), unwanted hair growth (hirsutism) and excessive fat around the stomach (central obesity).

Other side effects that are less common include problems with endocrine function, increased susceptibility to infection, elevation in blood pressure, risk of gastrointestinal perforation, serious skin rashes, behavioral and mood changes, decrease in the density of the bones and vision problems such as cataracts. Patients receiving immunosuppressive doses of corticosteroids should not be given live or live attenuated vaccines.

The FDA granted this application fast track designation and priority review. The drug also received orphan drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.

The sponsor is receiving a rare pediatric disease priority review voucher under a program intended to encourage development of new drugs and biologics for the prevention and treatment of rare pediatric diseases. A voucher can be redeemed by a sponsor at a later date to receive priority review of a subsequent marketing application for a different product. This is the ninth rare pediatric disease priority review voucher issued by the FDA since the program began.

Emflaza is marketed by Marathon Pharmaceuticals of Northbrook, Illinois.

Medical uses

The manufacturer lists the following uses for deflazacort:[1]

In the United States, deflazacort is only FDA-approved for the treatment of Duchenne muscular dystrophy in people over the age of 5.

Image result for DeflazacortImage result for Deflazacort

Image result for DeflazacortImage result for Deflazacort

Adverse effects

Deflazacort carries the risks common to all corticosteroids, including immune suppression, decreased bone density, and endocrine insufficiency. In clinical trials, the most common side effects (>10% above placebo) were Cushing’s-like appearance, weight gain, and increased appetite.[2]

Pharmacology

Mechanism of action

Deflazacort is an inactive prodrug which is metabolized rapidly to the active drug 21-desacetyldeflazacort.[3]

Relative potency

Deflazacort’s potency is around 70–90% that of prednisone.[4] A 2017 review found its activity of 7.5 mg of deflazacort is approximately equivalent to 25 mg cortisone, 20 mg hydrocortisone, 5 mg of prednisolone or prednisone, 4 mg of methylprednisolone or triamcinolone, or 0.75 mg of betamethasone or dexamethasone. The review noted that the drug has a high therapeutic index, being used at initial oral doses ranging from 6 to 90 mg, and probably requires a 50% higher dose to induce the same demineralizing effect as prednisolone. Thus it has “a smaller impact on calcium metabolism than any other synthetic corticosteroid, and therefore shows a lower risk of growth rate retardation in children and of osteoporosis” in the elderly, and comparatively small effects on carbohydrate metabolism, sodium retention, and hypokalemia.[5]

History

In January 2015, the FDA granted fast track status to Marathon Pharmaceuticals to pursue approval of deflazacort as a potential treatment for Duchenne muscular dystrophy, a rare, “progressive and fatal disease” that affects boys.[6] Although deflazacort was approved by the FDA for use in treatment of Duchenne muscular dystrophy on February 9, 2017,[7][8] Marathon CEO announced on February 13, 2017 that the launch of deflazacort (Emflaza) would be delayed amidst controversy over the steep price Marathon was asking for the drug – $89,000-a-year. In Canada the same drug can be purchased for around $1 per tablet.[9] Marathon has said that Emflaza is estimated to cost $89,000/year which is “roughly 70 times” more than it would cost overseas.[10] Deflazacort is sold in the United Kingdom under the trade name Calcort;[4] in Brazil as Cortax, Decortil, and Deflanil; in India as Moaid, Zenflav, Defolet, DFZ, Decotaz, and DefZot; in Bangladesh as Xalcort; in Panama as Zamen; Spain as Zamene; and in Honduras as Flezacor.[11]

SYNTHESIS

Worlddrugtracker drew this

1 Protection of the keto groups in pregna-1,4-diene derivative  with NH2NHCOOMe using HCOOH, yields the corresponding methyl ester.

2 Cleavage of epoxide  with NH3 in DMAc/DMF gives amino-alcohol,

3 which on esterification with acetic anhydride in the presence of AcOH furnishes acetate.

4 Cyclization of amine using NaOH, Na2CO3 or K2CO3 produces oxazoline derivative ,

5 which is finally deprotected with HCl to afford Deflazacort 

SYNTHESIS FROM CHEMDRUG

The cyclization of 17alpha-azido-3beta,16alpha-acetoxy-5alpha-pregnane-11,20-dione (I) by hydrogenation with H2 over Pt in methanol, followed by a treatment with 10% HCl gives 3beta-hydroxy-5alpha-pregnane-11,20-dione-[17alpha,16alpha-d]-2′-methyloxazoline (II), which is converted into the semicarbazone (III) by treatment with semicarbazide hydrochloride (A) and pyridine in refluxing methanol. The reduction of one ketonic group of (III) with NaBH4 in refluxing ethanol yields the dihydroxy-semicarbazone (IV), which is hydrolyzed with 10% HCl in refluxing methanol to afford the ketodiol (V). The oxidation of (V) with cyclohexanone and aluminum isopropoxide in refluxing toluene gives 11beta-hydroxy-5alpha-pregnane-3,20-dione-[17alpha,16alpha-d]-2′-methyloxazoline (VI). The dehydrogenation of (VI) by treatment with Br2 in dioxane-acetic acid, followed by treatment with Li2CO3 in DMF at 140 C yields the corresponding 1,4-diene derivative (VII). Finally, the reaction of (VII) with I2 by means of azobisisobutyronitrile in CH2Cl2 affords the corresponding 21-iodo compound, which is then acetylated with triethylammonium acetate in refluxing acetone.

The monoacetylation of (V) with acetic anhydride and pyridine at 100 C gives the 3-acetoxy-11-hydroxy compound (IX), which is dehydrated by treatment with methanesulfonyl chloride and then with sodium acetate yielding 3beta-acetoxy-5alpha-pregn-9(11)-ene-20-one-[17alpha,16alpha-d]-2′-methyloxazoline (X). The hydrolysis of (X) with KOH in refluxing methanol affords the corresponding hydroxy compound (XI), which is acetoxylated by treatment with I2 and AZBN as before giving the iodo derivative (XII), and then with triethylammonium acetate also as before, yielding 3beta-hydroxy-21-acetoxy-5alpha-pregn-9(11)-ene-20-one-[17alpha,16alpha-d]-2′-methyloxazoline (XIII). The oxidation of (XIII) with CrO3 in acetone yields the 3,20-diketone (XIV), which by treatment with Br2 and Li2CO3 as before is dehydrogenated affording the 1,4,9(11)-pregnatriene (XV). Finally, the reaction of (XV) with N-bromoacetamide in THF yields 9alpha-bromo-11beta-hydroxy-21-acetoxy-5alpha-pregna-1,4-dieno-3,20-dione-[17alpha,16alpha-d]-2′-methyloxazoline (XVI), which is then debrominated by reaction with chromous acetate and butanethiol in DMSO.

PAPER

Journal of Medicinal Chemistry (1967), 10(5), 799-802

Steroids Possessing Nitrogen Atoms. III. Synthesis of New Highly Active Corticoids. [17α,16α,-d]Oxazolino Steroids

J. Med. Chem., 1967, 10 (5), pp 799–802
DOI: 10.1021/jm00317a009

PATENT

CN 105622713

PATENT CN 106008660

MACHINE TRANSLATED FROM CHINESE may seem funny

Description of the drawings

[0007] Figure 1 is a map of the traditional method of the combination process;

Figure 2 is a two-step method of the present invention.

detailed description

[0008] In order to more easily illustrate the gist and spirit of the present invention, the following examples illustrate:

Example 1

A: Preparation of hydroxylamine

In a 100 ml three-necked flask, 20 g of 16 (17) a-epoxy prednisolone, 30 ml of DMF, 300 ml of chloroform was added and incubated at 30-35 ° C with 8 g of ammonia gas at 1-2 atmospheres Reaction 16 ~ 20 hours, TLC detection reaction end point, after the reaction, the vacuum exhaust ammonia gas, add 3x100ml saturated brine washing 3 times, plus 10ml pure water washing times, then, under reduced pressure to chloroform to dry, add 200ml Ethyl acetate, Ig activated carbon, stirring reflux 60-90 minutes, cooling to 50-55 degrees, hot filter, l-2ml ethyl acetate washing carbon, combined filtrate and lotion, and then below 500C concentrated under pressure 95 % Of ethyl acetate, the system cooled to -5-0 ° C, stirring crystallization 2 ~ 3 hours, filter, 0.5-lml ethyl acetate washing, lotion and filtrate combined sets of approved; filter cake below 70 ° C Drying, get hydroxylamine 18.2g, HPLC content of 99.2%, weight loss of 91%.

[0009] B: Preparation of terracavir

Add 10 g of hydroxylamine, 150 ml of glacial acetic acid and 150 ml of acetic anhydride in a 100 ml three-necked flask. Add 5 g of concentrated sulfuric acid under stirring at room temperature. The reaction was carried out at 30-35 ° C for 12-16 hours. TLC confirmed the end of the reaction. Add 500ml of pure water, and adjust the pH of 7.5.5 with liquid alkali, cool to 10 ~ 15 ° C, stirring crystallization 2-3 hours, filtration, washing to neutral, combined filtrate and lotion, pretreated into Waste water treatment tank, filter cake below 70 V drying, Texaco can be special crude 112.5g, HPLC content of 98.2%, the yield of 112.5% ο the above terracotta crude dissolved in 800ml of alcohol, add 5g activated carbon, Decolorization 1-1.5 hours, hot filter, 10ml alcohol detergent cake, lotion and filtrate combined, atmospheric pressure recovery of about 90% of the alcohol, and then cooled to -5-0 ° C, frozen crystal 2-3 hours, Filtration, filter cake with 4-5ml alcohol washing, 70 ° C below drying, digoxin special product 89.2g, melting point 255.5-256.0 degrees, HPLC content of 99.7%, yield 89.2%. The mother liquor is recycled with solvent and crude.

[0010] Example II

A: Preparation of hydroxylamine

In a 100 ml three-necked flask, 20 g of 16 (17) a-epoxy prednisolone, 120 ml of toluene was added and incubated at 30-35 ° C with 8 g of ammonia and 16 to 20 at atmospheric pressure The reaction was carried out in the presence of 3 x 50 ml of saturated brine and 50 ml of pure water was added. Then, the toluene was dried under reduced pressure to dryness, and 200 ml of ethyl acetate, Ig activated carbon was added, and the mixture was stirred. Reflux 60-90 minutes, cool to 50-55 ° C, hot filter, l2ml ethyl acetate wash carbon, combined filtrate and lotion, and then below 500C under reduced pressure 95% ethyl acetate, the system cooling To 5-0C, stirring crystallization 2 ~ 3 hours, filter, 0.5-lml ethyl acetate washing, lotion and filtrate combined sets of the next batch; filter cake 70 ° C below drying, hydroxylamine 18.0g, HPLC content 99.1%, 90% by weight.

[0011] B: Preparation of terracavir

Add 10 g of hydroxylamine, 500 ml of chloroform and 150 ml of acetic anhydride in a 100 ml three-necked flask, add 5 g of p-toluenesulfonic acid under stirring at room temperature, and incubate at 30-35 ° C for 12-16 hours. TLC confirms the reaction end, After the addition of 500ml of pure water, and with the liquid alkali pH 7.55, down to 10 ~ 15 ° C, stirring 0.5_1 hours, separate the water layer, washed to neutral, combined with water and lotion, pretreated into Waste water treatment tank, organic layer under reduced pressure concentrated chloroform to near dry, adding 200ml hexane, reflux 0.5-1 hours, slowly cooling to -5 ~ O0C, stirring crystallization 2-3 hours, filter, filter cake with 4-5ml Alcohol washing, the filtrate and lotion combined apply to the next batch, the filter cake below 70 ° C drying, Texaco can crude 110.5g, HPLC content of 98.4%, the yield of 110.5%. The above-mentioned diltiazem crude product dissolved in 800ml alcohol, add 5g activated carbon, temperature reflux bleaching 1-1.5 hours, hot filter, 10ml alcohol washing cake, lotion and filtrate combined, atmospheric pressure recovery of about 90% of the alcohol And then cooled to -500C, frozen crystallization for 2-3 hours, filtration, filter cake with 4-5ml alcohol washing, 70 ° C the following drying, digester can special products 88.6g, melting point 255.0-256.0 degrees, HPLC content of 99.5%, the yield of 88.6%. The mother liquor is recycled with solvent and crude.

[0012] Example 3

A: Preparation of hydroxylamine

Add 20 g of 16 (17) a-epoxy prednisolone to 120 ml of ethanol in a 100 ml three-necked flask and incubate at 30-35 ° C with stirring to give Sg ammonia at 16 to 20 hours , TLC test reaction end point, after the reaction, vacuum exhaust ammonia gas, concentrated ethanol to the near dry, cooling, adding 300ml chloroform, stirring dissolved residue, and then add 3x100ml saturated brine washing, plus 10ml pure water washing, washing And then concentrated to reduce the chloroform to dry, add 200ml of ethyl acetate, Ig activated carbon, stirring reflux 60-90 minutes, cooling to 50-55 ° C, hot filter, l2ml ethyl acetate washing carbon, combined filtrate and lotion And then concentrated below 50 ° C to 95% ethyl acetate under reduced pressure. The system was cooled to -5-0 0C, stirred for 2 to 3 hours, filtered, 0.5-l of ethyl acetate, washed and filtrate The filter cake was dried at 70 ° C, 18.6 g of hydroxylamine, 99.5% of HPLC, and 93% by weight.

[0013] B: Preparation of terracavir

In a 100ml three-necked flask, add 10g of hydroxylamine, 500ml toluene, 150ml acetic anhydride, stirring at room temperature by adding 5g concentrated sulfuric acid, insulation at 30-35 degrees stirring reaction 12-16 hours, TLC confirmed the end of the reaction, after the reaction, Add 500ml of pure water, and liquid pH adjustment pH 7.5, cooling to 1 ~ 15 ° C, stirring 0.5-1 hours, the water layer, washed to neutral, combined with water and lotion, pretreated into the wastewater The cells were dried and the organic layer was concentrated to dryness under reduced pressure. 200 ml of hexane was added and refluxed

0.5-1 hours, slowly cool to -5 ~ O0C, stirring crystallization 2-3 hours, filtration, filter cake with 4-5ml hexane, the filtrate and lotion combined apply to the next batch, filter cake below 70 ° C Drying, digoxin crude 112.5g, HPLC content of 97.4%, the yield of 112,5% ο will be the above terracotta crude dissolved in 800ml of alcohol, add 5g activated carbon, heating reflux bleaching 1-1.5 hours, while Hot filter, 10ml alcohol detergent cake, lotion and filtrate combined, atmospheric pressure recovery of about 90% of the alcohol, and then cooled to -500C, frozen crystallization for 2-3 hours, filter, filter cake with 4-5ml alcohol Washing, 70 ° C below the dry, Diges can special products 86.2g, melting point 255.5-256.0 degrees, HPLC content of 99.8%, the yield of 86.2%. The mother liquor is recycled with solvent and crude.

PATENT

https://www.google.com/patents/CN101418032A?cl=en

Example 1

21- bromo -ll (3- hydroxy – pregna–l, 4- diene -3, 20-dione [170, 16o-d] -2′- methyl-oxazoline (4) Preparation:

A dry fitted with a thermometer, a reflux condenser, magnetically stirred flask was added 250mL three compound (2) (19.17 g; Fw: 383.48; 50 mmol), N- bromosuccinimide (9.79 g; Fw: 178.00; 55 mmol), 150 ml of ether; then ammonium acetate (0.39 g; Fw: 77.08; 0.005 mmol) added to the system. System continues to stir at 20 ° C 0.5 h, the reaction is complete. After completion of the reaction was filtered to remove the white precipitate cake was washed with 50 mL of dichloromethane, and the combined organic Xiangde pale yellow clear liquid, the solvent was evaporated under reduced pressure to give a pale yellow solid 21.27 g, yield: 92%, HPLC content of greater than 95%.

Example 2

21- bromo -lip- hydroxy – pregna–l, 4- diene -3, 20-dione [17 “16o-d] -2′- methyl-oxazoline (4) Preparation:

A dry fitted with a thermometer, a reflux condenser, magnetically stirred flask were added sequentially 250mL three compound (2) (19.17 g; Fw: 383.48; 50 mmol), N- bromosuccinimide (9.79 g; Fw : 178.00; 55 mmol), 150 ml of toluene; then ammonium acetate (0.39 g; Fw: 77.08; 0.005 mmol) added to the system. System continues to stir at 110 ° C 5 h, the reaction is complete. After completion of the reaction was cooled to room temperature, the white precipitate was removed by filtration cake was washed with 50 mL of dichloromethane, and the combined organic Xiangde pale yellow clear liquid, concentrated under reduced pressure to remove the solvent to give a pale yellow solid 19.65 g, yield: 85%, HPLC content greater than 95%.

Example 3

21 Jie bromo -11 – hydroxy – pregna-1,4-diene -3, 20-dione [17a, 16o-d] -2′- methyl-oxazoline (4) Preparation:

A dry fitted with a thermometer, a reflux condenser, magnetically stirred flask were added sequentially 250mL three compound (2) (19.17 g; Fw: 383.48; 50 mmol), 1,3- dibromo-5,5-dimethyl- Hein (35.74 g; Fw: 285.94; 125 mmol), 150 ml of ether; then ammonium acetate (0.39 g; Fw: 77.08; 0.005 mmol) added to the system. System Stirring was continued at reflux for 3 h, the reaction was completed. After completion of the reaction a white precipitate was removed by filtration and the cake was washed with 50 mL of diethyl ether, and the combined organic Xiangde pale yellow clear liquid, concentrated under reduced pressure to remove the solvent to give a pale yellow solid 16.18 g, yield: 70%, HPLC content greater than 92%.

Example 4

21- bromo -11 Jie – hydroxy – pregna-1,4-diene -3, 20- dione [17c, 16o-d] -2′- methyl-oxazoline (4) Preparation:

A dry fitted with a thermometer, a reflux condenser, magnetically stirred flask were added sequentially 250mL three compound (2) (19.17 g; Fw: 383.48; 50 mmol), 1,3- dibromo-5,5-dimethyl- Hein (35.74 g; Fw: 285.94; 125 mmol), 150 ml dichloromethane; followed by ammonium acetate (0.039 g; Fw: 77.08; 0.0005 mmol) added to the system. System Stirring was continued at reflux for 24 h, the reaction was completed. After completion of the reaction a white precipitate was removed by filtration and the cake was washed with 50 mL of diethyl ether, and the combined organic Xiangde pale yellow clear liquid, concentrated under reduced pressure to remove the solvent to give a pale yellow solid 16.41 g, yield: 71%, HPLC content of greater than 92. / 0.

Example 5

Deflazacort Preparation:

In a nitrogen-filled dry fitted with a thermometer, magnetic stirring and a reflux condenser 100 mL three-necked flask was charged with Compound (4) (11.56 g; Fw: 462.38; 25 mmol), followed by addition of sodium acetate (8.20g; Fw: 82.03; lOOmmol), 50 mL methanol was added to the system.

Then tetrabutylammonium bromide (O. 81g; Fw: 322.38; 2.5 mmol). Warmed to 50 ° C with stirring

48 h. Until after the completion of the reaction was cooled to room temperature. After completion of the reaction, temperature of the system was cooled to room temperature, the system was supplemented with chloroform 50mL, filtered, and the filter cake was washed with small amount of chloroform and then to confirm that no product was dissolved, and the combined organic phases, the organic phase washed with 10% aqueous sodium carbonate paint 3 times, saturated sodium chloride once. The organic phase was dried over anhydrous sodium sulfate, the inorganic salt was removed to give a pale yellow liquid, was concentrated to dryness, purified ethyl acetate to give the product 9.93g, yield 90%, HPLC content> 990/0.

Example 6

Deflazacort Preparation –

In a nitrogen-filled dry fitted with a thermometer, magnetic stirring and a reflux condenser 100 mL three-necked flask was charged with Compound (4) (11.56 g; Fw: 462.38; 25 mmol), followed by addition of anhydrous potassium acetate (3.68g; Fw: 98.14; 37.5 mmol), 50 mL acetone was added to the system. Followed by tetrabutylammonium iodide (0.10g; Fw: 369.37; 0.25 mmol). Heated to reflux with stirring 2h. Until after the completion of the reaction was cooled to room temperature. After completion of the reaction, temperature of the system was cooled to room temperature, the system was supplemented with chloroform 50mL, filtered, and the filter cake was washed with small amount of chloroform and then to confirm that no product was dissolved, and the combined organic phases, the organic phase was washed 3 times with 10% aqueous sodium carbonate , washed once with saturated sodium chloride. The organic phase was dried over anhydrous sodium sulfate, the inorganic salt was removed to give a pale yellow liquid, was concentrated to dryness, ethyl acetate was purified to give the product 10.93 g, yield 99%, HPLC content> 99%.

Example 7

Deflazacort Preparation:

In a nitrogen-filled dry fitted with a thermometer, magnetic stirring and a reflux condenser 100 mL three-necked flask was charged with Compound (4) (11.56 g; Fw: 462.38; 25 mmol), followed by addition of anhydrous potassium acetate (3.68g; Fw: 98.14; 37.5 mmol), 50 mL acetonitrile was added to the system. Followed by tetrabutylammonium iodide (0.10g; Fw: 369.37; 0.25 mmol). Heated to reflux with stirring 2h. Until after the completion of the reaction was cooled to room temperature. After completion of the reaction, temperature of the system was cooled to room temperature, the system was supplemented with chloroform 50mL, filtered, and the filter cake was washed with small amount of chloroform and then to confirm that no product was dissolved, and the combined organic phases, the organic phase was washed 3 times with 10% aqueous sodium carbonate , washed once with saturated sodium chloride. The organic phase was dried over anhydrous sodium sulfate, the inorganic salt was removed to give a pale yellow liquid, was concentrated to dryness, ethyl acetate was purified to give the product 10.93 g, yield 99%, HPLC content> 99%.

Example 8

Deflazacort Preparation:

In a nitrogen-filled dry fitted with a thermometer, magnetic stirring and a reflux condenser 100 mL three-necked flask was charged with Compound (4) (11.56 g; Fw: 462.38; 25 mmol), followed by addition of anhydrous potassium acetate (2.45g; Fw: 98.14; 25 mmol), the N, N- dimethylformamide, 50 mL added to the system. Followed by tetrabutylammonium iodide (O.IO g; Fw: 369.37; 0.25 mmol). Warmed to 120. C stirring 2h. Until after the completion of the reaction was cooled to room temperature. After completion of the reaction, temperature of the system was cooled to room temperature, the system was supplemented with chloroform 50mL, filtered, and the filter cake was washed with small amount of chloroform and then to confirm that no product was dissolved, and the combined organic phases, the organic phase was washed 3 times with 10% aqueous sodium carbonate , washed once with saturated sodium chloride. The organic phase was dried over anhydrous sodium sulfate, the inorganic salt was removed to give a pale yellow liquid, was concentrated to dryness, ethyl acetate was purified to give the product 10.93 g, yield 99%, HPLC content> 99o / q.

PATENT

https://www.google.com/patents/WO1997021722A1?cl=zh

compound (llβ,16β)-21-(acetyloxy)-11- hydroxy-2 ‘ -methyl-5 ‘H-pregna-1, -dieno[17 , 16-d Joxazole- 3,20-dione, also known, and hereinafter referred to, with the INN (International Nonproprietary Name) deflazacort. Deflazacort is represented by the following formula I

Figure imgf000003_0001

Deflazacort is employed in therapy aince some years as a calcium-sparing corticoid agent. This compound belongs to the more general class of pregneno-oxazolines, for which anti-inflammatory, glucocorticoid and hormone-like pharmacological activities are reported. Examples of compounds of the above class, comprising deflazacort, are disclosed in US 3413286, where deflazacort is referred to as llβ-21-dihydroxy-2 ‘ -methyl-5 ‘ βH-pregna-1,4-dieno.17 , 16- d]oxazole-3,20-dione 21-acetate.

According to the process disclosed by US 3413286, deflazacort is obtained from 5-pregnane-3β-ol-ll , 20- dione-2 ‘-methyloxazoline by 2 , -dibromination with Br2– dioxane, heating the product in the presence of LiBr- iC03 for obtaining the 1,4-diene, and converting this latter into the 21-iodo and then into the desired 21- acetyloxy compound. By hydrolysis of deflazacort, the llβ-21-dihydroxy-2 ‘ -methyl-5 ‘βH-pregna-1, -dieno[ 17 , 16- d-]oxazoline-3, 20-dione of formula II is obtained:

Figure imgf000004_0001

The compound of formula II is preferably obtained according to a fermentation process disclosed in

EP-B-322630; in said patent, the compound of formula II is referred to as llβ-21-dihydroxy-2 ‘-methyl-5 ‘ βH- pregna-1,4-dieno[17,16-d-]oxazoline-3,20-dione.

The present invention provides a new advantageous single-step process for obtaining deflazacort, by acetylation of the compound of formula II.

CLIP

Image result for Deflazacort NMR

tructure of deflazacort and its forced degradation product (A), chromatogram plot of standard deflazacort (B), contour plot of deflazacort (C). Deflazacort was found to be a stable drug under stress condition such as thermal, neutral and oxidative condition. However, the forceddegradation study on deflazacort showed that the drug degraded under alkaline, acid and photolytic conditions.

Mass fragmentation pathway for degradant product of deflazacort.

PATENT

CN 103059096

Figure CN103059096AD00051

Example 1: Protective reaction To the reaction flask was added 20 g of 1,4-diene-11? -hydroxy-16,17-epoxy_3,20-dione pregnone (Formula I) 20% of the aqueous solution of glacial acetic acid 300g, stirring 5 minutes, temperature 10 ° C ~ 15 ° C, adding ethyl carbazate 14g, temperature control 30 ° C reaction 6 hours; TLC detection reaction is complete, cooling to 0 ° C ~ 5 ° C for 2 hours, until dry, washed to neutral; 60 ° C vacuum dry to dry creatures 20. 5g; on P, oxazoline ring reaction The above protective products into the reaction bottle, add 41ml Of the DMAC dissolved, temperature 25 ~ 30 ° C, access to ammonia, to keep the reaction bottle micro-positive pressure, the reaction of 32 hours, atmospheric pressure exhaust ammonia and then decompression pumping ammonia for 30 minutes; 5 ° C, temperature 5 ~ 0 ° C by adding 5ml glacial acetic acid, then add 21ml acetic anhydride, heated to 35 ° C reaction 4 hours, the sample to confirm the reaction completely; slowly add 5% sodium hydroxide solution 610ml and heated to 60 ~ 70 ° C reaction 2 hours; point plate to confirm the end of the reaction, cooling to 50 ° C, half an hour by adding refined concentrated hydrochloric acid 40ml, insulation 50 ~ 55 ° C reaction 10 hours; to the end of the reaction temperature to room temperature, chloroform Extraction, drying and filtration, concentration of at least a small amount of solvent, ethyl acetate entrained twice, leaving a small amount of solvent, frozen crystallization filter high purity [17a, 16a-d] terfu Kete intermediate. Example 2: Protective reaction 20 g of 1,4-diene-l1-la-hydroxy-16,17-epoxy_3,20_dione progestin (Formula I) was added to the reaction flask and 15% Formic acid solution 300g, stirring for 5 minutes, temperature 10 ~ 15 ° C, adding methyl carbazate 12g, temperature control 30 ° C reaction 5 hours to test the end of the reaction, cooling to O ~ 5 ° C stirring 2 hours crystallization, Suction to dry, washed to neutral; 60 ° C vacuum drying to dry protection of 20g; on P, oxazoline ring reaction The protection of the reaction into the reaction flask, add 30ml of DMF dissolved, temperature control 25 ~ 30 ° C, access to ammonia, keep the reaction bottle in the micro-positive pressure, reaction 30 hours, atmospheric pressure exhaust ammonia and then decompression pumping ammonia for 30 minutes, ice water cooled to 5 ° C, temperature 5 ~ 10 ° C add 5ml of glacial acetic acid, then add 20ml acetic anhydride, heated to 30 ° C reaction for 5 hours to confirm the reaction is complete; slowly add 20% sodium carbonate aqueous solution 500ml and heated to 60 ~ 70 ° C reaction 4 hours, the point plate to confirm the reaction The temperature of 55 ~ 60 ° C for 10 hours; to be the end of the reaction temperature to room temperature, chloroform extraction, drying and filtration, concentration of a small amount of solvent, acetic acid isopropyl The ester was entrained twice, leaving a small amount of solvent, frozen and crystallized to obtain high purity [17a, 16a-d] oxazoline residues. [0024] Example 3: Protective reaction 20 g of I, 4-diene-16,17-epoxy-3,11,20-triketone pregnone (Formula I) was added to the reaction flask and 20% Formic acid solution 300g, stirring for 5 minutes, temperature 10 ~ 15 ° C, adding hydrazine carbamate 15g, temperature control 30 ° C reaction 5 hours to test the end of the reaction, cooling to O ~ 5 ° C stirring 2 hours crystallization, To the dry, washed to neutral; 60 ° C vacuum drying to dry protection of 22g; on P, oxazoline ring reaction of the protection of the reaction into the bottle, add 30ml of DMAC dissolved temperature control 35 ~ 40 ° C, access to ammonia, keep the reaction bottle in the micro-positive pressure, reaction 40 hours, atmospheric pressure exhaust ammonia and then decompression pumping ammonia for 30 minutes, ice water cooling to 5 ° C, temperature 5 ~ 10 ° C add 5ml of glacial acetic acid, then add 20ml acetic anhydride, heated to 40 ° C reaction 5 hours to confirm the reaction is complete; slowly add 20% potassium carbonate aqueous solution 500ml and heated to 60 ~ 70 ° C reaction 7 hours, the point plate to confirm the reaction The temperature of the reaction to the end of the temperature to room temperature, chloroform extraction, drying filter, concentrated to a small amount of solvent, acetic acid isopropyl The ester was entrained twice, leaving a small amount of solvent, frozen and crystallized to obtain high purity [17a, 16a-d] oxazoline residues.

PATENT

CN 102936274

Figure CN102936274BD00041

xample 1

[0028] A 30 g 16, 17 α- epoxy – pregn -20- substituting methyl hydrazine -3-acetyl-1,4-diene, 11- dione (a) and 150 mL of chloroform and 15 mLDMF mixed, pressure reactor, stirring ammonia gas to the reactor pressure to 0.15 MPa (during ventilation control the reaction temperature at 10-15 ° C), 30 ° C heat reaction, TLC track the progress of the reaction. Completion of the reaction, the material was transferred to a glass reaction flask, the temperature of the material to be reduced to below 10 ° C, add acetic acid adjusted to pH 5 to 6, the solvent was removed under reduced pressure; reaction flask was added 30 mL of acetic acid, 30 g of acetic anhydride, The reaction temperature was controlled at 30 ° C, the reaction 6 hours, the reaction mixture was poured into cold 500 mL10% sodium hydroxide solution, stirred for 1 hour, filtration to give product 30.6 g, 102% mass yield, product by HPLC , a purity of 95.2%.

[0029] Example 2

[0030] A 30 g 16, 17 α- epoxy – pregn -20- substituting methyl hydrazine -3-acetyl-1,4-diene, 11- dione (a) and 150 mL of chloroform and 30 mL of pyridine were mixed, added pressure reactor, stirring ammonia gas to the reactor pressure to 0. 15 MPa (during ventilation control the reaction temperature at 10~15 ° C), 15 ° C heat reaction, TLC track the progress of the reaction. Completion of the reaction, the material was transferred to a glass reaction flask, the temperature of the material to be reduced to below 10 ° C, add acetic acid adjusted to pH 5 to 6, the solvent was removed under reduced pressure; reaction flask was added 30 mL of acetic acid, 30 g of acetic anhydride, The reaction temperature was controlled at 30 ° C, the reaction 6 hours, the reaction mixture was poured into cold 500 mL10% sodium hydroxide solution, stirred for 1 hour, filtration to give product 28.6 g, yield 95% by mass, product by HPLC , a purity of 94.8%.

[0031] Example 3

[0032] A 30 g 16, 17 α- epoxy – pregn -20- substituting methyl hydrazine -3-acetyl-1,4-diene, 11- dione (a) and 150 mL of chloroform and 30 mLDMF mixed, pressure reactor, stirring ammonia gas to the reactor pressure to 0.15 MPa (during ventilation control the reaction temperature at 10~15 ° C), 40 ° C heat reaction, TLC track the progress of the reaction.Completion of the reaction, the material was transferred to a glass reaction flask, the temperature of the material to be reduced to below 10 ° C, add acetic acid adjusted to pH 5 to 6, the solvent was removed under reduced pressure; reaction flask was added 30 mL of acetic acid, 30 g of acetic anhydride, The reaction temperature was controlled at 30 ° C, the reaction for 6 hours. The reaction mixture was poured into cold 500 mL10% sodium hydroxide solution, stirred for 1 hour, filtration to give the product 31.2 g, yield 104% quality products by HPLC , a purity of 95.4%.

[0033] Example 4

[0034] A 30 g 16, 17 α- epoxy – pregn -20- substituting methyl hydrazine -3-acetyl-1,4-diene, 11- dione (a) and 150 mL of chloroform and 30 mLDMF mixed, pressure reactor, stirring ammonia gas to the reactor pressure to 0.5 MPa (during ventilation control the reaction temperature at 10~15 ° C), 40 ° C heat reaction, TLC track the progress of the reaction. Completion of the reaction, the material was transferred to a glass reaction flask, the temperature of the material to be reduced to below 10 ° C, add acetic acid adjusted to pH 5 to 6, the solvent was removed under reduced pressure; reaction flask was added 30 mL of acetic acid, 30 g of acetic anhydride, The reaction temperature was controlled at 30 ° C, the reaction 6 hours, the reaction mixture was poured into cold 500 mL10% sodium hydroxide solution, stirred for 1 hour, filtration to give the product 31. I g, 102% mass yield, product by by HPLC, the purity was 95.2%.

[0035] Example 5

[0036] A 30 g 16, 17 α- epoxy – pregn -20- substituting methyl hydrazine -3-acetyl-1,4-diene, 11- dione (a) and 150 mL of chloroform and 30 mLDMF mixed, pressure reactor, stirring ammonia gas to the reactor pressure to 0.15 MPa (during ventilation control the reaction temperature at 10~15 ° C), 40 ° C heat reaction, TLC track the progress of the reaction. Completion of the reaction, the material was transferred to a glass reaction flask, the temperature of the material to be reduced to below 10 ° C, add acetic acid adjusted to pH 5 to 6, the solvent was removed under reduced pressure; reaction flask was added 60 mL of acetic acid, 15 g of acetic anhydride, The reaction temperature was controlled at 30 ° C, the reaction 6 hours, the reaction mixture was poured into cold 500 mL10% sodium hydroxide solution, stirred for 1 hour, filtration to give the product 29. 5 g, yield 98% by mass, the product of by HPLC, purity of 95%.

[0037] Example 6

[0038] A 30 g 16, 17 α- epoxy – pregn -20- substituting methyl hydrazine -3-acetyl-1,4-diene, 11- dione (a) and 150 mL of chloroform and 30 mLDMF mixed, pressure reactor, stirring ammonia gas to the reactor pressure to 0.15 MPa (during ventilation control the reaction temperature at 10~15 ° C), 40 ° C heat reaction, TLC track the progress of the reaction. The reaction was complete, the material was transferred to a glass reaction flask until the material temperature drops below 10 ° C, plus acetic acid to adjust the pH to 5 to 6, the solvent was removed under reduced pressure; the reaction flask was added 30 mL of acetic acid, 30 g of maleic dianhydride, the reaction temperature was controlled at 30 ° C, the reaction 6 hours, the reaction mixture was poured into cold 500 mL10% sodium hydroxide solution, stirred for 1 hour, filtration to give the product 30 g, 100% mass yield, product by HPLC purity of 95.2%.

[0039] Example 7

[0040] A 30 g 16, 17 α- epoxy – pregn -20- substituting methyl hydrazine -3-acetyl-1,4-diene, 11- dione (a) and 150 mL of chloroform and 30 mLDMF mixed, pressure reactor, stirring ammonia gas to the reactor pressure to 0.15 MPa (during ventilation control the reaction temperature at 10~15 ° C), 40 ° C heat reaction, TLC track the progress of the reaction. Completion of the reaction, the material was transferred to a glass reaction flask, the temperature of the material to be reduced to below 10 ° C, add acetic acid adjusted to pH 5 to 6, the solvent was removed under reduced pressure; reaction flask was added 30 mL of acetic acid, 30 g of propionic anhydride, The reaction temperature was controlled at 30 ° C, the reaction for 6 hours. The reaction mixture was poured into cold 500 mL10% sodium hydroxide solution, stirred for 1 hour, filtration to give the product 27.6 g, 92% yield of quality products by HPLC , a purity of 93.5%.

[0041] Example 8

[0042] A 30 g 16, 17 α- epoxy – pregn -20- substituting methyl hydrazine -3-acetyl-1,4-diene, 11- dione (a) and 150 mL of chloroform and 30 mLDMF mixed, pressure reactor, stirring ammonia gas to the reactor pressure to 0.15 MPa (during ventilation control the reaction temperature at 10~15 ° C), 40 ° C heat reaction, TLC track the progress of the reaction. Completion of the reaction, the material was transferred to a glass reaction flask, the temperature of the material to be reduced to below 10 ° C, add acetic acid adjusted to pH 5 to 6, the solvent was removed under reduced pressure; reaction flask was added 30 mL of acetic acid, 30 g of acetic anhydride, The reaction temperature is controlled at 50 ° C, the reaction for 6 hours. The reaction mixture was poured into cold 500 mL10% sodium hydroxide solution, stirred for 1 hour, filtration to give the product 29.8 g, 99% yield of quality products by HPLC , a purity of 94.8%.

References

  1. Jump up^ “Refla: deflazacort” (PDF).
  2. Jump up^http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/208684s000,208685s000lbl.pdf
  3. Jump up^ Möllmann, H; Hochhaus, G; Rohatagi, S; Barth, J; Derendorf, H (1995). “Pharmacokinetic/pharmacodynamic evaluation of deflazacort in comparison to methylprednisolone and prednisolone”. Pharmaceutical Research. 12 (7): 1096–100. PMID 7494809.
  4. ^ Jump up to:a b “Calcort”. electronic Medicines Compendium. June 11, 2008. Retrieved on October 28, 2008.
  5. Jump up^ Luca Parente (2017). “Deflazacort: therapeutic index, relative potency and equivalent doses versus other corticosteroids”. BMC Pharmacol Toxicol. doi:10.1186/s40360-016-0111-8.
  6. Jump up^ Ellen Jean Hirst (January 19, 2015), Duchenne muscular dystrophy drug could get OK for U.S. sales in 2016, The Chicago Tribune, retrieved February 13, 2017,has been shown to prolong lives … a progressive and fatal disease that has no drug treatment available in the US
  7. Jump up^ “FDA approves drug to treat Duchenne muscular dystrophy”. http://www.fda.gov. 2017-02-09. Retrieved 2017-02-10.
  8. Jump up^ “Marathon Pharmaceuticals to Charge $89,000 for Muscular Dystrophy Drug”. http://www.wsj.com. 2017-02-10. Retrieved 2017-02-10.
  9. Jump up^ Clifton Sy Mukherjee (February 10, 2017). “Brainstorm Health Daily”. Retrieved February 13, 2017.
  10. Jump up^ Joseph Walker and Susan Pulliam (February 13, 2017), Marathon Pharmaceuticals to Charge $89,000 for Muscular Dystrophy Drug After 70-Fold Increase, The Wall Street Journal, retrieved February 13, 2017,FDA-approved deflazacort treats rare type of disease affecting boys
  11. Jump up^ “Substâncias: DEFLAZACORT” (in Portuguese). Centralx. 2008. Retrieved on October 28, 2008.
Deflazacort
Deflazacort structure.svg
Clinical data
Trade names Emflaza, Calcort, others
AHFS/Drugs.com International Drug Names
Routes of
administration
By mouth
ATC code
Legal status
Legal status
Pharmacokinetic data
Protein binding 40%
Metabolism By plasma esterases, to active metabolite
Biological half-life 1.1–1.9 hours (metabolite)
Excretion Renal (70%) and fecal (30%)
Identifiers
CAS Number
PubChem CID
DrugBank
ChemSpider
UNII
KEGG
ChEMBL
ECHA InfoCard 100.034.969
Chemical and physical data
Formula C25H31NO6
Molar mass 441.517 g/mol
3D model (Jmol)
CN102746358A * Apr 22, 2011 Oct 24, 2012 天津金耀集团有限公司 Novel technology for synthesis of pregnane 21-bit bromide
CN102746358B * Apr 22, 2011 Feb 10, 2016 天津金耀集团有限公司 一种合成孕甾21位溴化物的工艺
CN102936274A * Nov 12, 2012 Feb 20, 2013 浙江仙居君业药业有限公司 Preparation method for [17alpha, 16alpha-d] methyl oxazoline
CN102936274B * Nov 12, 2012 Apr 1, 2015 江西君业生物制药有限公司 Preparation method for [17alpha, 16alpha-d] methyl oxazoline

///////FDA 2017, Emflaza, Calcort, Deflazacort, orphan drug designation, FAST TRACK

[H][C@@]12C[C@@]3([H])[C@]4([H])CCC5=CC(=O)C=C[C@]5(C)[C@@]4([H])[C@@]([H])(O)C[C@]3(C)[C@@]1(N=C(C)O2)C(=O)COC(C)=O

Filed under: 0rphan drug status, FAST TRACK FDA, FDA 2017, Uncategorized Tagged: Calcort, deflazacort, Emflaza, FAST TRACK, FDA 2017, Orphan Drug Designation

1-Amino-3-ethyl-5-nitratemethyladamantane hydrochloride (MN-05)

$
0
0

str1

1-Amino-3-ethyl-5-nitratemethyladamantane hydrochloride (MN-05)

Cas 1835197-63-1
C13 H22 N2 O3, 254.33
Tricyclo[3.3.1.13,7]decane-1-methanol, 3-amino-5-ethyl-, 1-nitrate
Amantadine and its derivatives have a variety of biological activity, in the field of medicine has a wide range of applications. Rimantine (1-aminoethyl adamantane, Rimantadine) is currently widely used in clinical prevention and treatment of influenza drugs. Amantadine is widely used in the treatment of influenza and Parkinson & apos; s Disease (PD) (Schwab et al., J. Am. Med. Asos. 1669, 208: 1168). Memantine is the only NMDA receptor antagonist approved by the US FDA for the treatment of moderate to severe Alzheimer’s disease (AD). NMDA receptors are a subtype of the important excitatory amino acid ionotropic glutamate receptors in the central nervous system and are important receptors in the learning and memory process. NMDA receptor pathway is opened after the non-selective allow some cations, such as Ca 2+ , K + and Na + into the cells, these ions, especially calcium ions can cause a series of biochemical reactions, and ultimately lead to neurotoxicity , Causing neuronal apoptosis. Memantine is a noncompetitive antagonist of the NMDA receptor open channel, which binds to binding sites within the ion channel and blocks the intramolecular flow and acts as a neuroprotective effect. The combination of memantine to NMDA receptors is reversible and has a moderate dissociation rate that ensures both pharmacological effects and ensures that it does not accumulate in the channel and affects normal physiological functions (Lipton et al. Journal of neurochemistry. 2006, 97: 1611-1626). At the same time, the antagonism of memantine to NMDA receptor has a strong voltage dependence, only in the neuronal depolarization can be combined with the receptor, which can block the pathological conditions of neurons continue to depolarize the NMDA receptor Activation, without blocking NMDA receptor activation under normal physiological conditions (Wenk et al., CNS drug reviews. 2003, 9 (3): 275-308; McKeage., Drugs & aging.2010,27 (2): 177-179 ). This protection mechanism is also important for the treatment of other central nervous system diseases such as stroke, PD, ALS and so on, and therefore it has a good prospect for the treatment of these diseases.
Nitric oxide (NO) also has a variety of biological activities in the body, it plays the role of signal molecules. Nitric oxide molecules can penetrate the cell wall into the smooth muscle cells, so that relaxation, expansion of blood vessels, lower blood pressure. But also into the platelet cells, reduce its activity, thereby inhibiting its agglutination and adhesion to the vascular endothelium to prevent thrombosis, prevent atherosclerosis. NO is a free radical gas, with an unpaired electron, the body is very unstable, very easy to react with free radicals, which can reduce the number of free radicals. The accumulation of free radicals can cause nucleic acid cleavage, enzyme passivation, polysaccharide depolymerization, and lipid peroxidation eventually leads to neuronal death (Yan et al. Free Radic. Biol. Med. 2013, 62: 90-101). NO has a strong ability to react with various free radicals, which can effectively reduce the number of free radicals, but its synthesis in vivo requires the participation of nitric oxide synthase (NOS). Under normal circumstances, NOS activity is relatively low, the need for nitro-like molecules or saponins substances activated. The introduction of NO-releasing groups on small molecule drugs can increase NO content in the body and have significant efficacy, such as nitroglycerin.
Because of the complex pathogenesis of AD, at present, the clinical treatment of AD is limited, only four acetylcholinesterase inhibitors and an NMDA receptor inhibitor. These drugs for the role of a single target molecules, can only alleviate some aspects of clinical symptoms of AD can not fundamentally cure the disease, blocking the process of neurodegeneration.
PATENT
Figure 2 depicts the synthesis of compound NM-004.
Example 3, Synthesis of compound NM-004a
A 50 mL round bottom flask was cooled in an ice-water bath and 20 mL of concentrated sulfuric acid, 2 mL of n-hexane and 970 mg (4 mmol) of compound NM-003a were added to the round bottom flask. To maintain the ice bath, slowly add formic acid (1.8mL). After dripping, continue the ice bath reaction for 3 hours. The reaction solution was poured into 100 mL of ice water to precipitate a solid. The mixture was allowed to stand and filtered to give a pale yellow solid. After drying in solid, it is dissolved in ethyl acetate and the aqueous solution of sodium hydroxide is basified to pH to about 9-10. The aqueous layer is separated. The organic layer was extracted with an aqueous solution of sodium hydroxide (30 mL x 3) and the aqueous solution was combined with a dilute hydrochloric acid solution to acidify the aqueous layer to a pH of about 3. Filtered and dried to give the pure compound NM-004a 640 mg (77%). ESI-MS: m / z 207 ([MH] ). 1 H-NMR (DMSO-d6, ppm): 0.76 (t, 3H, J = 7.5Hz), 1.11 (q, 2H, J = 7.5Hz) (M, 2H), 1.47 (s, 2H), 1.51-1.64 (m, 2H), 1.66-1.81 (m, 4H), 2.01 (m, 2H), 11.99 (s, 1H).
Example 4, Synthesis of compound NM-004b
To a 50 mL round bottom flask, 624 mg (3 mmol) of compound NM-004a was added and the mixture was cooled in an ice bath. Add 0.55mL concentrated nitric acid, stir well. To the mixture was added 3.5 mL of concentrated sulfuric acid, and the reaction was carried out in an ice bath for 1 hour. After which 2.5 mL of acetonitrile (4.8 mmol) was added dropwise and the reaction was continued for 1 hour in an ice bath. The reaction solution was poured into 20 mL of ice water and vigorously stirred for 30 minutes and allowed to stand overnight. The precipitate was removed by filtration, and the solid was washed with the appropriate amount of water and dried to obtain the compound NM-004b (580 g, 73%) without further purification. ESI-MS: m / z 266 ([M + H] + ). 1 H-NMR (DMSO-d6, ppm): 0.74 (t, 3H, J = 7.5Hz), 1.15 (q, 2H, J = 7.5 Hz, 1.26-1.35 (m, 2H), 1.36-1.47 (m, 2H), 1.52-1.70 (m, 4H), 1.72-1.86 (m, 5H), 1.88-1.98 (m, 2H) M, 1H), 7.43 (s, 1H).
Example 5, Synthesis of compound NM-004c
The compound NM-004b 878 mg (3.3 mmol) was dissolved in 10 mL of dry tetrahydrofuran and cooled in an ice-water bath. 0.5 mL of triethylamine and 0.5 mL of ethyl chloroformate were added to the mixture, followed by an ice bath for 30 minutes. The ice bath was removed and reacted at room temperature for 4 hours. The filtrate was filtered and the filtrate was washed with an appropriate amount of tetrahydrofuran. To the filtrate by adding sodium borohydride 1.5g, dropping funnel slowly dropping 1mL water, 1 hour drop finished. After dripping at room temperature, the reaction was continued for 1 hour. TLC monitoring, the reaction is completed, the reaction system to add water 30mL, dry spin dry tetrahydrofuran. The aqueous layer was extracted with ethyl acetate (20 mL x 4), combined with ethyl acetate, 25 mL of 0.5 N hydrochloric acid, saturated aqueous sodium chloride solution and water, and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give crude oil. Silica gel column (petroleum ether: ethyl acetate = 1: 1), and finally a white solid, NM-004c, 348 mg (42%) was obtained. ESI-MS: m / z 252.2 ([M + H] + ). 1 H-NMR (DMSO-d6, ppm): 0.76 (t, 3H, J = 7.5Hz), 1.03-1.20 (m, 4H) 1.28 (m, 4H), 1.58 (m, 4H), 1.75 (m, 5H), 2.09 (s, 1H), 3.02 (d, 2H, J = 5.5 Hz), 4.38 (t, 1H, J = ), 7.33 (s, 1H).
Example 6, Synthesis of compound NM-004d
To a 250 mL round bottom flask was added 1.26 g (5 mmol) of compound NM-004c, 3 g of solid sodium hydroxide, 20 mL of diethylene glycol and refluxed at 170 ° C for 15 hours. After cooling to room temperature, the reaction solution was poured into 40 g of crushed ice and the mixture was stirred. The mixture was extracted with ethyl acetate (20 mL x 4). The combined ethyl acetate layer, 30 mL of water and 30 mL of saturated sodium chloride solution were washed and dried over anhydrous sodium sulfate. The solvent was evaporated to dryness to give a crude product as a pale yellow oil. The crude product was dissolved in 50 mL of dry ethyl acetate and the resulting dry HCl was passed under stirring to precipitate a large amount of white solid. The solid was washed with an appropriate amount of dry ethyl acetate and dried to give 850 mg (69.4%) of white solid NM-004d. ESI-MS: m / z 210.3 ([M + H] + ). 1 H-NMR (DMSO-d6, ppm): 0.74 (t, 3H, J = 7.6 Hz), 1.15 (q, 2H, J = 7.6 Hz, 1.26-1.35 (m, 2H), 1.36-1.47 (m, 2H), 1.53-1.68 (m, 4H), 1.74-1.85 (m, 3H), 1.88-1.96 (m, 2H) M, 1H), 7.43 (s, 3H).
Example 7, Synthesis of compound NM-004e
Take the compound NM-004d 2.45 g, (10 mmol) in 20 mL of water, basify the sodium hydroxide solution to pH 10, and extract with ethyl acetate (30 mL x 4). The combined ethyl acetate was washed with 30 mL of water and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give 1.57 g (7.5 mmol) of free amine as a colorless oil. (15.6 mmol) of triethamine, 2.55 g (11.7 mmol) of Boc anhydride and 10 mg of DMAP were added to 50 mL of distilled steam in tetrahydrofuran, and the reaction was monitored by TLC at room temperature for 5 hours. After completion of the reaction, the reaction solution was quenched by adding 30 mL of saturated ammonium chloride solution to the reaction solution. The solvent was evaporated under reduced pressure and extracted with ethyl acetate (50 mL x 4). The combined ethyl acetate was washed with 30 mL of 0.1 N hydrochloric acid and 30 mL of saturated aqueous sodium chloride solution and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to give the crude product as a colorless oil. The crude product was separated on a silica gel column (petroleum ether: ethyl acetate = 1: 1) and 1.58 g (68%) of the white solid NM-004e was obtained. ESI-MS: m / z 310.3 ([M + H] + ). 1 H-NMR (DMSO-d6, ppm): 0.75 (t, 3H, J = 7.5Hz), 1.03-1.19 (m, 4H) 1.24 (m, 4H), 1.36 (s, 9H), 1.44-1.58 (m, 4H), 1.52-1.73 (m, 2H), 2.08 (s, 1H), 3.02 (d, 2H, J = 5.5 Hz) , 4.38 (t, 1H, J = 5.5 Hz), 6.36 (s, 1H).
Example 8, Synthesis of compound NM-004f
The compound NM-004e (620 mg, 2 mmol) was dissolved in 10 mL of dry water, dichloromethane and cooled in an ice-water bath. Add acetic anhydride and fuming nitric acid mixture (acetic anhydride and fuming nitric acid volume ratio equal to 3: 2) 2mL. Ice bath reaction 10-15 minutes. The reaction solution was poured into 10 mL of 1N sodium bicarbonate solution, and the dichloromethane was separated. The aqueous layer was extracted with dichloromethane (10 mL x 3), combined with dichloromethane and washed with 10 mL of water. Dried over anhydrous sodium sulfate, filtered and the dichloromethane was distilled off under reduced pressure to give the crude product as a colorless oil. Silica gel column separation (petroleum ether: dichloromethane = 10: 1) was obtained as a colorless oil NM-004 f 505 mg (73.4%). ESI-MS: m / z 377.2 ([M + Na] + ). 1 H-NMR (DMSO-d6, ppm): 0.76 (t, 3H, J = 7.5Hz), 1.08-1.23 (m, 4H) 1.26-1.49 (m, 14H), 1.56-1.82 (m, 5H), 2.12 (m, 1H), 4.23 (s, 2H), 6.50 (s, 1H).
Example 9, Synthesis of compound NM-004
To the compound NM-004f710 mg (2 mmol) was added 5 mL of a saturated solution of hydrogen chloride and reacted at room temperature. At the end of the reaction, a white solid precipitates. Filtration, anhydrous ether washing white solid, you can get NM-004 pure. After drying, NM-004380 mg (65.5%) was obtained. ESI-MS: m / z 255.1 ([M + H] + ). 1 H-NMR (DMSO-d6, ppm): 0.78 (t, 3H, J = 7.5Hz), 1.15-1.28 (m, 4H) (M, 2H), 1.40-1.55 (m, 4H), 1.57-1.67 (m, 2H), 1.71 (s, 2H), 2.23 (m, 1H), 4.30 (s, 2H) S, 3H).

Med. Chem. Commun., 2017, 8,135-147

DOI: 10.1039/C6MD00509H, Research Article
Zheng Liu, Si Yang, Xiaoyong Jin, Gaoxiao Zhang, Baojian Guo, Haiyun Chen, Pei Yu, Yewei Sun, Zaijun Zhang, Yuqiang Wang
A series of memantine nitrate derivatives, as dual functional compounds with neuroprotective and vasodilatory activity for neurodegenerative diseases, was designed and synthesized

Synthesis and biological evaluation of memantine nitrates as a potential treatment for neurodegenerative diseases

Zheng Liu,a   Si Yang,a   Xiaoyong Jin,a   Gaoxiao Zhang,a  Baojian Guo,a   Haiyun Chen,a   Pei Yu,a   Yewei Sun,*a  Zaijun Zhang*a and   Yuqiang Wanga  
*Corresponding authors
aInstitute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
E-mail: yxy0723@163.com, zaijunzhang@163.com
Fax: +86 20 8522 4766
Tel: +86 20 8522 5030
Med. Chem. Commun., 2017,8, 135-147

DOI: 10.1039/C6MD00509H

A series of memantine nitrate derivatives, as dual functional compounds with neuroprotective and vasodilatory activity for neurodegenerative diseases, was designed and synthesized. These compounds combined the memantine skeleton and a nitrate moiety, and thus inhibited the N-methyl-D-aspartic acid receptor and released NO in the central nervous system. The biological evaluation results revealed that the new memantine nitrates were effective in protecting neurons against glutamate-induced injury in vitro. Moreover, memantine nitrates dilated aortic rings against phenylephrine-induced contraction. The structure–activity relationships of neuroprotection and vasodilation were both analyzed. In further studies, compound MN-05 significantly protected cortical neurons by inhibiting Ca2+ influx, reducing free radical production and maintaining the mitochondrial membrane potential. Further research on MN-05 is warranted.

1-Amino-3-ethyl-5-nitratemethyladamantane hydrochloride (MN-05).

Compound MN- 05 was synthesized using a similar method to that as described for synthesis of compound MN-01 from compound 16. White solid, 65.5% yield. ESI-MS: m/z 255.1 [M + H]+ .

1H NMR (300 MHz, DMSO-d6) δ 0.75-0.80 (t, J = 7.5 Hz, 3H, CH3), 1.16-1.24 (q, J = 7.5 Hz, 2H, CH2), 1.24-1.25 (m, 2H), 1.30-1.39 (m, 2H), 1.43 (s, 2H), 1.45-1.57 (dd, J = 12 Hz, 6 Hz, 2H), 1.57-1.63 (dd, J = 12 Hz, 6 Hz, 2H), 1.71 (s, 2H), 2.23 (m, 1H, CH), 4.30 (s, 2H, CH2O), 8.21 (s, 3H, NH2HCl).

13C NMR (75 MHz, DMSO-d6) δ 7.4, 28.6, 34.5, 35.0, 36.8, 40.3, 41.6, 43.9, 52.3, 80.9. Anal. Calcd for C13H23N2O3Cl·0.3 H2O: C, 52.72%; H, 8.03%; N, 9.46%. Found: C, 52.72%; H, 7.92%; N, 9.51%

//////// memantine nitrates ,  neurodegenerative diseases, MN 05
[O-][N+](=O)OCC12CC3(CC(N)(C1)CC(C2)C3)CC

“ALL FOR DRUGS” CATERS TO EDUCATION GLOBALLY, No commercial exploits are done or advertisements added by me. This article is a compilation for educational purposes only.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent


Filed under: Uncategorized Tagged: MN 05, MNO5

AZD 8931, Sapitinib,

$
0
0

AZD8931 (Sapitinib)Figure imgf000027_0003

AZD 8931, Sapitinib, SAPATINIB

PHASE 2, at AstraZeneca for the treatment of non-small cell lung cancer.

CAS 848942-61-0,

MF C23H25ClFN5O3, MW 473.9,

pan-EGFR/pan-erbB inhibitor

4-[[4-[(3-Chloro-2-fluorophenyl)amino]-7-methoxy-6-quinazolinyl]oxy]-N-methyl-1-piperidineacetamide

4-(3-Chloro-2-fluoroanilino)-7-methoxy-6-[[1-(N-methylcarbamoylmethyl)piperidin-4-yl] oxy]quinazoline

4-(3-Chloro-2-fluoroanilino)-7-methoxy-6-[[1-(N-methylcarbamoylmethyl)piperidin-4-yl]oxy]quinazoline

2-[4-[4-(3-Chloro-2-fluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-1-piperidyl]-N-methyl-acetamide

AZD8931 is an oral, equipotent inhibitor of ErbB1, ErbB2 and ErbB3 receptor signaling.

WO 2005028469

Inventors Robert Hugh Bradbury, Laurent Francois Andre Hennequin, Bernard Christophe Barlaam
Applicant Astrazeneca Ab, Astrazeneca Uk Limited

Image resultDeregulation of the HER receptor family, comprising four related receptor tyrosine kinases (EGFR, HER2, HER3, and HER4), promotes proliferation, invasion, and tumor cell survival.Such deregulation has been observed in many human cancers, including lung, head and neck, and breast. Numerous small molecules have been investigated for inhibition of tyrosine kinases with the aminoquinazoline motif coming to the forefront as a privileged scaffold. Three of the clinically available treatments, gefitinib (1),lapatinib (2), and erlotinib (3),as well as the candidate drug dacomitinib (4), contain this arrangement

Figure

Figure 1. Structure of gefitinib (1), lapatinib (2), erlotinib (3), dacomitinib (4), and AZD8931 (5).

SYNTHESIS

PATENT

https://www.google.com/patents/WO2005028469A1?cl=en

PAPER

The first radiosynthesis of [11C]AZD8931 as a new potential PET agent for imaging of EGFR, HER2 and HER3 signaling
Bioorganic & Medicinal Chemistry Letters (2014), 24, (18), 4455-4459.

Image for unlabelled figure

Synthesis of the reference standard AZD8931 (11a) and its precursor ...

Synthesis of the reference standard AZD8931 (11a)

Reagents and conditions: (a) SnCl2·H2O, concd HCl; (b) formamide, 168–170 °C; (c) l-methionine, methanesulfonic acid, 120 °C; (d) Ac2O, pyridine, DMAP, 100 °C; (e) POCl3, DEA, 100 °C; (f) 3-chloro-2-fluoroaniline, i-PrOH, refluxing; (g) conc. NH3, MeOH; (h) (1) Boc2O, CH2Cl2, dioxane; (2) methanesulfonyl chloride, Et3N, CH2Cl2; (i) Compound 8, CsF, DMA, 85 °C; (j) TFA; (k) Compound 11a: 2-chloro-N-methylacetamide, KI, K2CO3, CH3CN, refluxing; compound

PAPER

Discovery of AZD8931, an Equipotent, Reversible Inhibitor of Signaling by EGFR, HER2, and HER3 Receptors
ACS Medicinal Chemistry Letters (2013), 4, (8), 742-746.

Discovery of AZD8931, an Equipotent, Reversible Inhibitor of Signaling by EGFR, HER2, and HER3 Receptors

Centre de Recherches, AstraZeneca, Z.I. La Pompelle, B.P. 1050, Chemin de Vrilly, 51689 Reims, Cedex 2, France
Oncology iMed, AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, United Kingdom
Abstract Image

Deregulation of HER family signaling promotes proliferation and tumor cell survival and has been described in many human cancers. Simultaneous, equipotent inhibition of EGFR-, HER2-, and HER3-mediated signaling may be of clinical utility in cancer settings where the selective EGFR or HER2 therapeutic agents are ineffective or only modestly active. We describe the discovery of AZD8931 (2), an equipotent, reversible inhibitor of EGFR-, HER2-, and HER3-mediated signaling and the structure–activity relationships within this series. Docking studies based on a model of the HER2 kinase domain helped rationalize the increased HER2 activity seen with the methyl acetamide side chain present in AZD8931. AZD8931 exhibited good pharmacokinetics in preclinical species and showed superior activity in the LoVo tumor growth efficacy model compared to close analogues. AZD8931 is currently being evaluated in human clinical trials for the treatment of cancer.

4-(3-Chloro-2-fluoroanilino)-7-methoxy-6-{[1-(N-methylcarbamoylmethyl)piperidin-4-yl]oxy}quinazoline
(2). 2 as a white solid (60%).1H NMR (CDCl3):
δ 1.98 (m, 2H), 2.08 (m, 2H), 2.46 (m, 2H), 2.85 (m, 2H), 2.87 (d, 3H), 3.07 (s, 2H), 4.02 (s, 3H), 4.49 (m, 1H),
7.16 (m, 4H), 7.31 (m, 2H), 8.49 (m, 1H), 8.71 (s, 1H). MS-ESI m/z MH+ 474 [MH]+. Anal.
(C23H25ClFN5O3
.0.21 H2O) C, H, N. Found C, 57.88; H, 5.45; N, 14.67; Requires C, 57.83; H, 5.36; N, 14.66%.

PATENT

WO 2010122340

Compound (I) is disclosed in International Patent Application Publication number WO2005/028469 as Example 1 therein and is of the structure:

Figure imgf000002_0001

Compound (I)

Compound (I) is an erbB receptor tyrosine kinase inhibitor, in particular compound (I) is a potent inhibitor of EGFR and erbB2 receptor tyrosine kinases. Compound (I) also inhibits erbB3 mediated signalling through the inhibition of phosphorylation of erbB3 following ligand stimulated EGFR/erbB3 and/or erbB2/erbB3 heterodimerisation. Compound (I) is expected to be useful in the treatment of hyperproliferative disorders such as cancer.

WO 03/082831 discloses the preparation of various 4-(3-chloro-2- fluoroanilino)quinazo lines. However, compound (I) is not disclosed therein.

WO2005/028469 discloses as Example 1 therein the preparation of compound (I) as follows: 2-Chloro-N-methylacetamide (32 mg, 0.3 mmol) was added to a mixture of

4-(3-chloro-2-fluoroanilino)-7-methoxy-6-[(piperidin-4-yl)oxy]quinazoline (120 mg, 0.3 mmol), potassium iodide (16 mg, 0.1 mmol), and potassium carbonate (50 mg, 0.36 mmol) in acetonitrile (5 ml). The mixture was heated at reflux for one hour. After evaporation of the solvents under vacuum, the residue was taken up in dichloromethane. The organic solution was washed with water and brine, dried over magnesium sulfate. After evaporation of the solvents under vacuum, the residue was purified by chromatography on silica gel (eluant: 1% to 2% 7 N methanolic ammonia in dichloromethane) to give compound (I).

Scheme 1 :

Figure imgf000008_0001

Example 1 : Preparation of 4-(3-Chloro-2-fluoroanilino)-7-methoxy-6-{[l-(N- methylcarbamoylmethyl)piperidin-4-yl]oxy } quinazoline (Compound (I)).

Compound (I) was prepared according to the scheme shown below:

Figure imgf000019_0001

Compound (III) Compound (IV)

Compound (V)

Figure imgf000019_0002

Compound (I)Compound (II)

Step 1. Preparation of tert-butyl 4-(5-cyano-2-methoxyphenoxy)piperidine-l- carboxylate (Intermediate 2). 3-hydroxy-4-methoxybenzonitrile (Compound (X), 6.00 g, 39.62 mmole), tert-butyl (4-methanesulfonyloxy)piperidine-l-carboxylate (16.6 g, 59.44 mmoles) (Chemical & Pharmaceutical Bulletin 2001, 49(7), 822-829); and potassium carbonate (6.71 g, 47.55 mmoles) were suspended in isopropanol (78.98 g) and the mixture was heated at reflux with stirring. Additional tert-butyl (4-methanesulfonyloxy)piperidine-l- carboxylate (2.08 g, 7.43 mmoles) was added to push the reaction to completion. The mixture was then cooled and quenched by the addition of water (100.47 g). Seeding with intermediate 2 followed by cooling to 00C resulted in a crystalline product, which was isolated by filtration. The filter cake was washed with a mixture of water (8.86 g) and isopropanol (6.97 g), followed by water (23.64 g) and then dried to give Intermediate 2 (10.75 g, 80% yield); 1H NMR (400 MHz, DMSO-d6) δ ppm 1.39 (s, 9 H) 1.48 (m, 2 H) 1.88 (m, 2 H) 3.13 (m, 2 H) 3.67 (m, 2 H) 3.83 (s, 3 H) 4.56 (tt, J=8.1, 3.8 Hz, 1 H) 7.13 (d, J=8.4 Hz, 1 H) 7.42 (dd, J=8.4, 1.9 Hz, 1 H) 7.51 (d, J=1.9 Hz, 1 H); Mass Spectrum: m/z (M + H)+ 333.1. Step 2. Preparation of 4-methoxy-3-(piperidin-4-yloxy)benzonitrile (Compound

(VI)). Intermediate 2 (39.31 g, 118.26 mmoles) was suspended in ethanol (155.53 g) and heated to 40 0C. To this slurry was slowly added HCl (46.61 g, 573.04 mmoles). The mixture was heated to 60 0C and held for 3 hours. The reaction mixture was cooled to 200C and seed was charged initiating crystallisation. The resulting solid was isolated by filtration at 00C, washed twice with ethanol (62.21 g) and then dried to give compound (VI) as the hydrochloride salt (29.84 g, 77% yield); 1H NMR (400 MHz, DMSO-d6) δ ppm 1.84 (m, 2 H) 2.09 (m, 2 H) 3.02 (ddd, J=12.7, 8.9, 3.4 Hz, 2 H) 3.20 (m, 2 H) 3.84 (s, 3 H) 4.63 (tt, J=7.7, 3.6 Hz, 1 H) 7.15 (d, J=8.5 Hz, 1 H) 7.45 (dd, J=8.5, 1.9 Hz, 1 H) 7.56 (d, J=1.9 Hz, 1 H) 9.16 (br. s, 2 H); Mass Spectrum: m/z (M + H)+ 233.2. Step 3. Preparation of 2-[4-(5-cyano-2-methoxyphenoxy)piperidin-l-yl]-JV- methylacetamide (Compound (V)). Compound (VI) (28.36 g, 95.82 mmoles), 2-chloro-N- methylacetamide (12.37 g, 114.98 mmoles) and potassium carbonate (33.11 g, 239.55 mmoles) were suspended in acetonitrile (161.36 g). The reaction mixture was heated at reflux for 3 hours. The reaction mixture was cooled to 200C and water (386.26 g) was charged. The reaction was heated to 75°C and the volume reduced by distillation. Upon cooling crystallisation occurred. The resulting solid was isolated by filtration, washed twice with water (77.25 g and 128.75 g) and then dried to give compound (V) (27.95 g, 94% yield); 1H NMR (400 MHz, DMSO-J6) δ ppm 1.68 (m, 2 H) 1.91 (m, 2 H) 2.29 (m, 2 H) 2.61 (d, J=4.7 Hz, 3 H) 2.67 (m, 2 H) 2.88 (s, 2 H) 3.83 (s, 3 H) 4.41 (tt, J=8.3, 4.0 Hz, 1 H) 7.11 (d, J=8.4 Hz, 1 H) 7.40 (dd, J=8.4, 1.9 Hz, 1 H) 7.47 (d, J=I.9 Hz, 1 H) 7.68 (q, J=4.7 Hz, 1 H); Mass Spectrum: m/z (M + H)+ 304.2.

Step 4. Preparation of 2-[4-(5-cyano-2-methoxy-4-nitrophenoxy)piperidin-l-yl]-N- methylacetamide (Compound (IV)). Compound (V) (8.78 g, 26.11 mmoles) was suspended in acetic acid (22.82 g, 364.87 mmoles) and the resulting reaction mixture cooled to 5°C. To this was added sulfuric acid (23.64 g, 234.95 mmoles) maintaining the reaction temperature below 300C. To the resulting solution was added nitric acid (2.40 g, 26.63 mmoles). The reaction mixture was then heated to 35°C and held for 3 hours. Additional nitric acid (117 mg, 1.31 mmoles) and sulphuric acid (1.31 g 13.1 mmoles) were charged and the reaction mixture was heated at 35°C for 30 minutes. The solution was cooled to 200C and quenched with aqueous ammonia (92.45 g 1.36 moles), resulting in an increase in temperature to 500C. To the resulting slurry was added, propionitrile (61.58 g 1.12 moles) and water (19 g). The reaction mixture was heated to 80 0C resulting in a clear solution, which upon settling gave two layers. The bottom layer was removed. The reaction mixture was cooled to 20 0C resulting in a thick slurry. The solid was isolated by filtration, washed with propionitrile (6.16 g 112.0 mmoles) and dried to afford compound (IV) (7.44 g, 82% yield); 1H NMR (400 MHz, DMSO-de) δ ppm 1.72 (m, 2 H) 1.97 (m, 2 H) 2.35 (m, 2 H) 2.61 (d, J=4.7 Hz, 3 H) 2.66 (m, 2 H) 2.90 (s, 2 H) 3.96 (s, 3 H) 4.73 (tt, J=8.4, 4.0 Hz, 1 H) 7.71 (q, J=4.7 Hz, 1 H) 7.82 (s, 1 H) 7.86 (s, 1 H). Mass Spectrum: m/z (M + H)+ 349.2

Step 5. Preparation of 2-[4-(4-amino-5-cyano-2-methoxyphenoxy)piperidin-l-yl]-N- methylacetamide (Compound (III)). Compound (IV) (7.42 g, 19.38 mmoles) was suspended in water (44.52 g) and methanol (5.35 g). To this was added sodium dithionite (11.91 g, 58.15 mmoles) and the resulting reaction mixture was heated to 600C. To the reaction mixture was added hydrochloric acid (46.98 g, 463.89 mmoles)), resulting in a solution, which was held at 60 0C for 3 hours. The reaction mixture was then allowed to cool to 20 0C. Aqueous sodium hydroxide (15.51 g 182.2 mmoles) was charged followed by 2-methyltetrahydrofuran (58.0 g). The reaction mixture was heated to 60 0C, which upon settling gave two layers and the lower aqueous layer was discarded. The volume of the reaction mixture was reduced by vacuum distillation and methyl tert-butyl ether (18.54 g) was added to give a slurry which was cooled to 10 0C. and then the solid was collected by filtration. The solid was washed with 2- methyltetrahydrofuran (5.8 g) and dried to give compound (III) (5.4 g, 78% yield); 1H NMR (400 MHz, DMSO-de) δ ppm 1.62 (m, 2 H) 1.82 (m, 2 H) 2.20 (m, 2 H) 2.60 (d, J=4.7 Hz, 3 H) 2.65 (m, 2 H) 2.86 (s, 2 H) 3.72 (s, 3 H) 4.00 (tt, J=8.3, 4.0 Hz, 1 H) 5.66 (br. s, 2 H) 6.39 (s, 1 H) 6.94 (s, 1 H) 7.65 (q, J=4.7 Hz, 1 H). Mass Spectrum: m/z (M + H)+ 319.2.

Step 6. Preparation of 2-[4-(5-cyano-4-{[(dimethylamino)methylene]amino}-2- methoxyphenoxy)piperidin-l-yl]-Λ/-methylacetamide (Compound (H)). Compound (III) (18.21 g, 52.05 mmoles) was suspended in 2-methyltetrahydrofuran (99.62 g). To this was added acetic acid (162.79 mg), and N,N-dimethylformamide dimethyl acetal (DMA) (8.63 g, 70.27 mmoles) and the resulting reaction mixture was heated at 76 0C for 16 hrs. Additional N,N-dimethylformamide dimethyl acetal (639.41 mg, 5.20 mmoles) was added to the reaction mixture to ensure the reaction completed. The reaction mixture was cooled to 300C during which time crystallisation occurred. The resulting solid was isolated by filtration, washed with 2-methyltetrahydrofuran (14.23 g) and dried to afford compound (II) (19.53 g, 97% yield); 1H NMR (400 MHz, DMSO-J6) δ ppm 1.65 (m, 2 H) 1.86 (m, 2 H) 2.24 (m, 2 H) 2.60 (d, J=4.7 Hz, 3 H) 2.66 (m, 2 H) 2.87 (s, 2 H) 2.95 (s, 3 H) 3.04 (s, 3 H) 3.81 (s, 3 H) 4.19 (tt, J=8.2, 3.8 Hz, 1 H) 6.72 (s, 1 H) 7.15 (s, 1 H) 7.67 (q, J=4.7 Hz, 1 H) 7.90 (s, 1 H); Mass Spectrum: m/z (M + H)+ 374.2.

Step 7. Preparation of compound (I). 2-[4-(5-cyano-4-

{ [(dimethylamino)methylene] amino } -2-methoxyphenoxy)piperidin- 1 -yl] -JV-methylacetamide (compound (II), 7.00 g, 17.71 mmoles), was suspended in methoxybenzene (35.8 g). Acetic acid (16.6 g) was charged and to the resulting solution was added 3-chloro-2-fluoroaniline (2.71 g, 18.07 mmoles). The reaction mixture was heated at 90 0C for 20 hours then cooled to 200C. Water (37.04 g) was charged to the reaction mixture, and the organic layer discarded. To the resulting aqueous mixture was charged isopropanol (39.00 g), followed by aqueous ammonia (20.79 g, 25%). The reaction mixture was heated to 30 0C and seeded with compound (I), which induced crystallisation. The reaction was then cooled to 00C and the product isolated by filtration. The filter cake was washed twice with a mixture of water (7.28 g) and isopropanol (4.68 g), then dried to afford the compound (I) (5.65 g, 55% yield); 1H NMR (400 MHz, DMSO-J6) δ ppm 1.79 (m, 2 H) 2.04 (m, 2 H) 2.38 (m, 2 H) 2.62 (d, J=4.5 Hz, 3 H) 2.74 (m, 2 H) 2.94 (s, 2 H) 3.93 (s, 3 H) 4.56 (tt, J=8.1, 3.8 Hz, 1 H) 7.21 (s, 1 H) 7.28 (m, 1 H) 7.50 (m, 2 H) 7.73 (q, J=4.5 Hz, 1 H) 7.81 (s, 1 H) 8.36 (s, 1 H) 9.56 (br.s, 1 H); Mass Spectrum: m/z (M + H)+ 474.2, 476.2.

Example 2: Preparation of 4-(3-Chloro-2-fluoroanilino)-7-methoxy-6-{[l-(N- methylcarbamoylmethyl)piperidin-4-yl]oxy } quinazoline (Compound (I)). Compound (I) was prepared according to the scheme shown below:

Figure imgf000023_0001

Compound (III) Compound (IV)

Compound (V)

Figure imgf000023_0002

Compound (Xl)

Figure imgf000023_0003

Compound (I)

Steps 1, 2, 3 and 4 as set forth in Example 1.

Step 5, alternate 1. Preparation of compound (III). 2-[4-(5-Cyano-2-methoxy-4- nitrophenoxy)piperidin-l-yl]-N-methylacetamide (compound (IV), 15.00 g, 42.50 mmoles) was suspended in water (90.00 g) and methanol (59.38 g). To this was added sodium dithionite (30.47 g, 148.75 mmoles) and water (90.00 g), the resulting reaction mixture was heated to 30 0C and held for 2 hrs. To the reaction mixture was added hydrochloric acid (27.98 g, 276.25 mmoles)), resulting in a solution, which was held at 600C for 2 hours. Aqueous sodium hydroxide (30.60 g 382.49 mmoles) was added followed by a line wash of water (30.00 g). The reaction mixture was cooled to 25°C to give a slurry which was collected by filtration. The solid was washed with water (30.00 g) and dried to give compound (III) (13.50 g, 82% yield); 1H NMR (400 MHz, DMSO-d6) δ ppm 1.62 (m, 2 H) 1.82 (m, 2 H) 2.20 (m, 2 H) 2.60 (d, J=4.7 Hz, 3 H) 2.65 (m, 2 H) 2.86 (s, 2 H) 3.72 (s, 3 H) 4.00 (tt, J=8.3, 4.0 Hz, 1 H) 5.66 (br. s, 2 H) 6.39 (s, 1 H) 6.94 (s, 1 H) 7.65 (q, J=4.7 Hz, 1 H). Mass Spectrum: m/z (M+H)+ 319.2.

Step 5, alternate 2. Preparation of compound (III). Compound (IV) (8.00 g, 22.67 mmoles) and 1% platinum + 2 % vanadium catalyst on carbon (1.23 g, 0.023 mmoles) were suspended in Acetonitrile (94.00 g). The reaction mixture was hydrogenated at a pressure of 3 Bar G and at a temperature of 35°C for 3 hrs. Once complete, the reaction mixture was filtered to remove the catalyst which is washed with acetonitrile (31.33 g). The volume of the reaction mixture was reduced by vacuum distillation to give a slurry which was cooled to 00C and then the solid was collected by filtration. The solid was washed with acetonitrile (12.53 g) and dried to give compound (III) (5.88 g, 78% yield); 1H NMR (400 MHz, DMSO-d6) δ ppm 1.62 (m, 2 H) 1.82 (m, 2 H) 2.20 (m, 2 H) 2.60 (d, J=4.7 Hz, 3 H) 2.65 (m, 2 H) 2.86 (s, 2 H) 3.72 (s, 3 H) 4.00 (tt, J=8.3, 4.0 Hz, 1 H) 5.66 (br. s, 2 H) 6.39 (s, 1 H) 6.94 (s, 1 H) 7.65 (q, J=4.7 Hz, 1 H). Mass Spectrum: m/z (M+H)+ 319.2.

Step 6. Preparation of N, ΛT-bis(3-chloro-2-fluorophenyl)imidoformamide (compound (XI)). 3-chloro-2-fluroaniline (51.21 g, 341.22 mmoles) was suspended in cyclohexane (87.07 g). To this ethyl orthoformate (22.28 g, 150.32 mmoles) and acetic acid (0.94 g, 15.03 mmoles) were added. The resulting reaction mixture was heated, with stirring, to 48°C for 12 hours. Following this the reaction mixture was cooled to 200C over 12 hours and the solid product was isolated by filtration. The filter cake was washed with cylcohexane (26.12 g) and dried in vacuo at 40 0C to give compound (XI) as a white crystalline product (33.95 g, 93% yield); IH NMR Spectrum (400 MHz, DMSO-d6) δ ppm 7.14 (t, 2 H) 7.22 (m, 2 H) 8.14 (s, 1 H), 9,98 (s, 1 H); Mass Spectrum (by GC-MS EI): m/z (M+) 300.0.

Step 7, alternate 1 : Preparation of compound (I). 2-[4-(4-Amino-5-cyano-2- methoxyphenoxy)piperidin-l-yl]-N-methylacetamide (compound (III)) (10 g, 29.84 mmol) and TV, ΛT-bis(3-chloro-2-fluorophenyl)imidoformamide (compound (XI)) (11.46 g, 37.3 mmol) were suspended in 2-methyltetrahydrofuran (30.4 ml) and heated to 800C. To this yellow suspension was added acetic acid (7.6 ml, 127.33 mmol) and the resulting solution was heated to 92°C for 6 hours. 2-methyltetrahydrofuran (66.5 ml) and water (28.5 ml) were added and mixture was cooled to 550C before adding 50%w/w sodium hydroxide (7 ml, 131.29 mmol) resulting in a temperature rise to 63°C. The temperature was raised further to 69°C and after settling the aqueous phase was discarded. The organic phase was washed with water (3 x 20 ml) and each aqueous phase was discarded after settling. 2- methyltetrahydrofuran (100 ml, 997 mmol) was added and the volume reduced by distillation. Seed was added to induce crystallisation and the resulting mixture was cooled to 15°C. The crystalline form was initially obtained following a spontaneous crystallisation from the experiment as described. The resulting solid was isolated by filtration, washed twice with 2- methyltetrahydrofuran (19 ml) and dried under vacuum at 400C to yield compound (I) as a white solid (12.14 g, 95%). 1H NMR (400 MHz, DMSO-J6) δ ppm 1.12 (d, J= 6Hz, 1.3H), 1.26 -1.36 (m, 0.4H), 1.75-1.97 (m, 3.3H), 2.02-2.15 (m, 2H), 2.35-2.44 (m, 2H), 2.64 (d, J= 4.7Hz, 3H), 2.72-2.80 (m, 2H), 2.95 (s, 2H), 3.52-3.59 (m, 0.4H), 3.72-3.87 (m, 0.86H), 3.95 (s, 3H), 4.53-4.63 (m, IH), 7.22 (s, IH), 7.29 (dt J= IHz J= 8Hz, IH), 7.51 (dt J= 7.4Hz, J= 18Hz, 2H), 7.71-7.77 (m, IH), 7.82 (s, IH), 8.37 (s, IH), 9.57 (s, IH). Mass Spectrum: m/z (M+H)+ 474.0. The NMR data above includes signals for the 2-methyltetrahydrofuran solvent which is present in a 0.43 molar equivalence. The signals pertaining to the solvent are at δ ppm shifts of 1.12, 1.26-1.36, 3.52-3.59 and 3.72-3.87. The cluster at 1.75-1.93 contains signals for the solvent and the parent compound. The XRPD for this compound is shown in Figure 2.

Step 7, alternate 2. Preparation of compound (I). Compound (III) (15 g, 44.76 mmol) and compound (XI) (17.19 g, 55.95 mmol) were suspended in 2-methyltetrahydrofuran (45.6 ml) and heated to 83°C. To this yellow suspension was added acetic acid (11.4 ml, 190.99 mmol) and the resulting solution was heated to 92°C for 3 Vi hours. 2-methyltetrahydrofuran (105 ml) and water (50 ml) were added and mixture was cooled to 49°C before adding 50%w/w sodium hydroxide (10.74 ml, 201.4 mmol), resulting in a temperature rise to 62°C. The temperature was maintained at 62°C and after settling the aqueous phase was discarded. The organic phase was washed with water (3 x 30 ml) and each aqueous phase was discarded after settling. The mixture was cooled to 15°C and seed was added to induce crystallisation. The crystalline form was initially obtained following a spontaneous crystallisation from the experiment as described. The resulting solid was isolated by filtration, washed twice with 2- methyltetrahydrofuran (21 ml) and dried under vacuum at 400C to yield compound (I) as a white solid (20.12 g, 95%). 1H NMR (400 MHz, DMSO-J6) δ ppm 1.75-1.86 (m, 2H), 2.02- 2.15 (m, 2H), 2.35-2.44 (m, 2H), 2.64 (d, J= 4.7Hz, 3H), 2.72-2.80 (m, 2H), 2.95 (s, 2H), 3.95 (s, 3H), 4.53-4.63 (m, IH), 7.22 (s, IH), 7.29 (dt J= IHz J= 8Hz, IH), 7.51 (dt J= 7.4Hz, J= 18Hz, 2H), 7.71-7.77 (m, IH), 7.82 (s, IH), 8.37 (s, IH), 9.57 (s, IH). Mass Spectrum: m/z (M+H)+ 474.0. The XRPD for this compound is shown in Figure 3.

Step 7, alternate 3. Preparation of compound (I). Compound (III) (15.1 g, 45.06 mmol) and compound (XI) (17.31 g, 56.32 mmol) were suspended in 2-methyltetrahydrofuran (46 ml) and heated to 800C. To this yellow suspension was added acetic acid (12 ml, 458 mmol) and the resulting solution was heated to 92° C for 7 hours. 2-methyltetrahydrofuran (100 ml) and water (43 ml) were added and mixture was cooled to 59°C before adding 50%w/w sodium hydroxide (11 ml, 207 mmol), resulting in a temperature rise to 71.5°C. The temperature was adjusted to 69°C and the aqueous phase was discarded after settling. The organic phase was washed with water (2 x 43 ml) and each aqueous phase was discarded after settling. 2-methyltetrahydrofuran (72 ml) was removed by distillation at atmospheric pressure and was replaced by addition of isopropyl alcohol (72 ml). A further 72 ml of solvent was removed by distillation at atmospheric pressure and replaced by isopropyl alcohol (72 ml). Seed was added to induce crystallisation and the resulting mixture was cooled to 15°C. The solid was isolated by filtration, washed twice with isopropylalcohol (32 ml) and dried under vacuum at 400C to yield compound (I) as a white solid (20.86 g, 87%). 1H NMR (400 MHz, DMSO-J6) δ ppm 1.04 (d, J= 6Hz, 6H),1.75-1.88 (m, 2H), 2.02-2.15 (m, 2H), 2.35-2.44 (m, 2H), 2.64 (d, J= 4.7Hz, 3H), 2.72-2.80 (m, 2H), 2.95 (s, 2H), 3.73-3.84 (m, IH), 3.95 (s, 3H), 4.34 (d, J = 4.2Hz, IH), 4.53-4.63 (m, IH), 7.22 (s, IH), 7.29 (dt J= IHz J= 8Hz, IH), 7.51 (dt J= 7Hz, J= 18Hz, 2H), 7.71-7.77 (m, IH), 7.82 (s, IH), 8.37 (s, IH), 9.57 (s, IH). Mass Spectrum: m/z (M+H)+ 474.0. The NMR data include signals for 1 mole equivalent isopropanol present. The XRPD for this compound is shown in Figure 4.

Example 3. Preparation of 4-(3-Chloro-2-fluoroanilino)-7-methoxy-6-{[l-(N- methylcarbamoylmethyl)piperidin-4-yl]oxy } quinazoline di- [(2E)-but-2-enedioate] (compound (I) difumarate salt). Compound (I) difumarate salt was prepared according to the scheme shown below:

Figure imgf000027_0001

Compound (III) Compound (IV) Compound (V)

Figure imgf000027_0002

Compound (Xl)

Figure imgf000027_0003

Difumarate Compound (I)

Steps 1, 2, 3, 4, 5 and 6 were performed as set forth in Example 2. Step 7. Preparation of compound (I) difumarate salt. Compound (III) (17.90 mmoles) and N, ΛT-bis(3-chloro-2-fluorophenyl)imidoformamide (compound (XI)) (7.04 g, 23.27 mmoles) were suspended in tert-butyl alcohol (88.95 g). To this suspension fumaric acid (10.39 g, 89.52 mmoles) was added and the mixture was heated to 800C, with stirring, for 2.5 hrs. Water (11.40 g, 632.80 mmoles) was charged and the reaction continued for a further 21.5 hrs. The reaction was cooled to 200C over 12 hours, during which time crystallisation occurred. The resulting solid was isolated by filtration and was washed with a mixture of water (1.00) and tert-butyl alcohol (7.80 g) followed by a wash with a mixture of water (0.50 g) and tert-butyl alcohol (7.30 g). The solid was dried in vacuo at 40 0C to give compound (I) difumarate salt (8.17 g, 61.40%) as a mustard yellow powder; 1H NMR (400 MHz, DMSO- dβ) δ ppm 1.83 (m, 2 H, broad) 2.07 (m, 2 H, broad) 2.64 (d, J=5.0 Hz, 3 H) 2.80 (m, 2 H, broad) 3.03 (s, 2 H) 3.94 (s, 3 H) 4.58 (m, 1 H) 6.63 (s, 4 H) 7.22 (s, 1 H) 7.29 (td, J=8.5, 1.0 Hz, 1 H) 7.51 (m, 2 H) 7.82 (m, 2 H) 8.37 (s, 1 H); Mass Spectrum: m/z (M+H)+ 474.0. Example 4. Preparation of 4-(3-Chloro-2-fluoroanilino)-7-methoxy-6-{[l-(N- methylcarbamoylmethyl)piperidin-4-yl]oxy}quinazoline (compound (I)).

Compound (I) was prepared according to the scheme shown below:

Figure imgf000029_0001

Compound (III) Compound (IV)

Compound (V)

Figure imgf000029_0002

Compound (XII)

Figure imgf000029_0003

Compound (I)

Steps 1, 2, 3, 4 and 5 were performed as set forth in Example 2.

Step 6. Preparation of N’-(3-chloro-2-fluoro-phenyl)-N,N-dimethyl-formamidine (compound (XII)). 3-chloro-2-fluroaniline (5.30 g, 35.29 mmoles) was dissolved in 2- methyltetrahydrofuran (52.94 g). To this N,N-dimethylformamide dimethyl acetal (6.07 g, 49.41 mmoles) and acetic acid (0.11 g, 1.76 mmoles) were added. The resulting reaction mixture was heated, with stirring, to 76 0C for 3 hours. Following this the solvent was removed in vacuo at 400C to give compound (XII) as a yellow oil (6.60 g, 93% yield); IH NMR Spectrum (400 MHz, DMSO-d6) δ ppm 2.74 (s, 0.29H), 2.89 (s, 0.31H), 2.94 (s, 2.75H), 3.03 (s, 2.66H), 3.34 (br s, 0.70H), 5.48 (s, 0.06H) 6.91-7.10 (m, 3H), 7.79 (s, 1 H), 7.96 (s, 1 H). The NMR data above includes signals for N,N-dimethylformamide dimethyl acetal which is present in a 0.06 molar equivalence. The signals pertaining to N5N- dimethylformamide dimethyl acetal are at δ ppm shifts of 3.75, and 6.90-6.95. The signal at δ ppm 3.35 is due to residual water. Mass Spectrum (by LCMS EI): m/z (M+H)+ 201.2. Step 7: Preparation of compound (I). 2-[4-(4-Amino-5-cyano-2- methoxyphenoxy)piperidin-l-yl]-N-methylacetamide (compound (III)) (0.50 g, 1.45 mmol) and N’-(3-chloro-2-fluoro-phenyl)-N,N-dimethyl-formamidine (compound (XII)) (0.32 g, 1.52 mmol) were suspended in methoxybenzene (3.1 ml). To this yellow suspension was added acetic acid (1.52 ml, 25.51 mmol) and the resulting solution was heated to 90 0C for 14 hours. The reaction mixture was cooled to 20 0C and water (2.58 mL) was added. The organic layer was removed and the aqueous layer washed with methoxybenzene (1.4 mL). Ethanol (2.45 mL) and ammonia (1.94 ml, 25.55 mmoles) were added to the aqueous layer. The solution was heated to 900C resulting in the loss of some ethanol by evaporation. The solution was cooled to 40 0C. Seed was added to induce crystallisation and the resulting mixture was cooled to 20 0C. The solid was isolated by filtration to yield compound (I) as a white solid (0.61 g, 73% yield). IH NMR (400 MHz, DMSO-d6) δ ppm 1.75-1.87 (m, 2H), 2.02-2.15 (m, 2H), 2.35-2.44 (m, 2H), 2.64 (d, J= 4.8Hz, 3H), 2.72-2.80 (m, 2H), 2.95 (s, 2H), 3.35 (s, 5.4H), 3.75 (s, 1.3H), 3.95 (s, 3H), 4.58 (hept., J=4.0Hz, IH), 6.90-6.95 (m, 1.3H), 7.23 (s, 1.8H), 7.26-7.34 (m, IH), 7.45-7.58 (m 2H), 7.72-7.78 (m, IH), 7.83 (s, IH), 8.38 (s, IH), 9.58 (s, IH). The NMR data above includes signals for the methoxybenzene solvent which is present in a 0.40 molar equivalence. The signals pertaining to the solvent are at δ ppm shifts of 3.75, and 6.90-6.95. The cluster at 7.26-7.34 contains signals for the solvent and the parent compound. The signal at δ ppm 3.35 is due to residual water. Mass Spectrum: m/z (M + H)+ 474.0, 476.0. Example 5. Preparation of compound (I) difumarate Form A – 2-[4-({4-[(3-Chloro-2- fluorophenyl)amino]-7-methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide di- [(2E)-but-2-enedioate] Form A. A solution of fumaric acid (2.7 g, 23.22 mmol) in methanol (95 ml) was added to a mixture of 2-[4-({4-[(3-Chloro-2-fluorophenyl)amino]-7- methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide (compound (I)) (5.62 g at 89% w/w, 10.55 mmol) in isopropanol (100 ml) maintaining the temperature >65°C. The mixture was heated at reflux for one hour before clarification. The reaction mixture was cooled to 300C over 90 minutes and held for 30 minutes to establish crystallisation. The reaction was cooled to 00C over 2 hours and held for 1 hour before isolation by filtration. The filter cake was washed twice with cold isopropanol (2 x 10 ml) and dried in vacuo at 500C to give the title compound as a white solid (5.84 g, 78%); 1H NMR Spectrum: (DMSO) 1.85 (m, IH), 2.08 (m, IH), 2.50 (m, IH), 2.66 (d, 3H), 2.83 (m, IH), 3.05 (s, 2H), 3.96 (s, 3H), 4.58 (m, IH), 6.64 (s, 4H), 7.23 (s, IH), 7.28 (m, IH), 7.46 (ddd, IH), 7.55 (m, IH), 7.70 (broad q, IH), 7.85 (s, IH), 8.38 (s, IH).

Example 6. Preparation of compound (I) difumarate Form A: 2-[4-({4-[(3-Chloro-2- fluorophenyl)amino]-7-methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide di- [(2E)-but-2-enedioate] Form A. A solution of fumaric acid (1.4 kg, 12.1 mol) in methanol (26.6 kg) was added to a mixture of 2-[4-({4-[(3-chloro-2-fluorophenyl)amino]-7- methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide (2.93 kg, 84.8% w/w, 5.24 mol) in isopropanol (39 kg) maintaining the temperature >65°C. A line wash of methanol (3.6 kg) was charged. The mixture was heated at reflux for one hour before clarification, followed by a line wash of methanol (7 kg). The reaction mixture was distilled at atmospheric pressure to remove 47 kg of distillates. Isopropanol (15.8 kg was added and the reaction mixture distilled to remove 15.6 kg of distillates. Crystallisation occurred during the distillation. Isopropanol (21 kg) was added and the reaction cooled to 00C over 8 hours and held for 1 hour before isolation by filtration. The filter cake was washed with cold 50:50 isopropanol:MeOH (4 kg) followed by cold isopropanol (4 kg) and dried in vacuo at 500C to give the title compound as a white solid (3.64 kg, 98%); 1H NMR Spectrum: (DMSO) 1.85 (m, IH), 2.08 (m, IH), 2.50 (m, IH), 2.66 (d, 3H), 2.83 (m, IH), 3.05 (s, 2H), 3.96 (s, 3H), 4.58 (m, IH), 6.64 (s, 4H), 7.23 (s, IH), 7.28 (m, IH), 7.46 (ddd, IH), 7.55 (m, IH), 7.70 (broad q, IH), 7.85 (s, IH), 8.38 (s, IH).

Example 7. Preparation of compound (I) difumarate Form A: 2-[4-({4-[(3-Chloro-2- fluorophenyl)amino]-7-methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide di- [(2E)-but-2-enedioate] Form A. 2-[4-({4-[(3-Chloro-2-fluorophenyl)amino]-7- methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide (compound (I)) (60.19 g at 88% w/w, 111.8 mmol) was dissolved in ethyl acetate (1550 ml). The solution was clarified by filtration and the filter washed with ethyl acetate (53 ml). The solution was cooled to 400C. A clarified solution of fumaric acid (26.60 g, 257.0 mmol) in isopropanol (408 ml) was then added over 1 hour. The filter used to clarify the fumaric acid solution was then washed with isopropanol (37 ml). After holding for 1 hour at 400C the reaction was cooled to 200C over 1 hour. The reaction mixture was held for 13.5 hours before isolating the product by filtration. The filter cake was washed twice with ethyl acetate (82 ml) : isopropanol (24 ml) and then dried in vacuo at 400C to give the title compound as a white solid (72.32 g, 90%); 1H NMR Spectrum: (DMSO) 1.85 (m, IH), 2.08 (m, IH), 2.50 (m, IH), 2.66 (d, 3H), 2.83 (m, IH), 3.05 (s, 2H), 3.96 (s, 3H), 4.58 (m, IH), 6.64 (s, 4H), 7.23 (s, IH), 7.28 (m, IH), 7.46 (ddd, IH), 7.55 (m, IH), 7.70 (broad q, IH), 7.85 (s, IH), 8.38 (s, IH). Example 8. Preparation of compound (I) difumarate Form A: 2-[4-({4-[(3-Chloro-2- fluorophenyl)amino]-7-methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide di- [(2E)-but-2-enedioate] Form A. 2-[4-({4-[(3-Chloro-2-fluorophenyl)amino]-7- methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide (compound (I)) (2.75 g at assumed 100% w/w, 5.80 mmol) was dissolved in ethyl acetate (94 ml) and isopropanol (14 ml). The solution was distilled such that 25.2 ml of distillates were collected. The solution was cooled to 400C. A clarified solution of fumaric acid (1.38 g, 11.90 mmol) in isopropanol (21 ml) was then added over 1 hour. Compound (I) difumarate Form A seed was added (3.7 mg, 5.3 μmol). The filter used to clarify the fumaric acid solution was then washed with isopropanol (2 ml). After holding for 1 hour at 400C the reaction was cooled to 200C over 2 hours. The reaction mixture was held for 15 hours before isolating the product by filtration. The filter cake was washed twice with ethyl acetate (4.3 ml): isopropanol (1.2 ml) and then dried in vacuo at 400C to give the title compound as a white solid (72.32 g, 90%); 1H NMR Spectrum: (DMSO) 1.85 (m, IH), 2.08 (m, IH), 2.50 (m, IH), 2.66 (d, 3H), 2.83 (m, IH), 3.05 (s, 2H), 3.96 (s, 3H), 4.58 (m, IH), 6.64 (s, 4H), 7.23 (s, IH), 7.28 (m, IH), 7.46 (ddd, IH), 7.55 (m, IH), 7.70 (broad q, IH), 7.85 (s, IH), 8.38 (s, IH).

Example 9. Preparation of compound (I) difumarate Form A: 2-[4-({4-[(3-Chloro-2- fluorophenyl)amino]-7-methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide di- [(2E)-but-2-enedioate] Form A. 2-[4-({4-[(3-Chloro-2-fluorophenyl)amino]-7- methoxyquinazolin-6-yl}oxy)piperidin-l-yl]-N-methylacetamide (compound (I)) (1 g, 1.86 mmoles) and fumaric acid (0.44 g, 3.81 mmoles) were suspended in water (4.4 g) and heated to 85°C. The reaction mixture was cooled to 600C at l°C/minute and compound (I) Form A seed was added when the temperature was 77°C. The resulting solid was isolated by filtration, washed twice with acetone (0.7O g per wash) and dried in a vacuum oven at 400C to afford the title compound (0.89 g, 68% yield), IH NMR (400 MHz, DMSO-d6) d ppm 1.84 (m, 2 H) 2.08 (m, 2 H) 2.55 (m, 2 H) 2.63 (d, J=4.7 Hz, 3 H) 2.86 (m, 2 H) 3.12 (s, 2 H) 3.93 (s, 3 H) 4.59 (tt, J=7.8, 3.7 Hz, 1 H) 6.62 (s, 4 H) 7.21 (s, 1 H) 7.27 (td, J=8.1, 1.3 Hz, 1 H) 7.49 (m, 2 H) 7.86 (m, 2 H) 8.36 (s, 1 H) 9.63 (br. s., 1 H). Compound (I) difumarate Form A is a free flowing powder.

PAPER

http://pubs.acs.org/doi/abs/10.1021/acs.oprd.6b00412

The Development of a Dimroth Rearrangement Route to AZD8931

The Department of Pharmaceutical Sciences, AstraZeneca, Silk Road Business Park, Macclesfield, Cheshire SK10 2NA, United Kingdom
The Department of Pharmaceutical Technology and Development, AstraZeneca, Silk Road Business Park, Macclesfield, Cheshire SK10 2NA, United Kingdom
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00412

Abstract Image

Recently, the aminoquinazoline motif has been highly prevalent in anticancer pharmaceutical compounds. Synthetic methods are required to make this structure on a multikilo scale and in high purity. The initial route to aminoquinazoline AZD8931 suffered from the formation of late-stage impurities. To avoid these impurities, a new high-yielding Dimroth rearrangement approach to the aminoquinazoline core of AZD8931 was developed. Assessment of route options on a gram scale demonstrated that the Dimroth rearrangement is a viable approach. The processes were then evolved for large-scale production with learning from a kilo campaign and two plant-scale manufactures. Identification of key process impurities offers an insight into the mechanisms of the Dimroth rearrangement as well as the hydrogenation of a key intermediate. The final processes were operated on a 30 kg scale delivering the target AZD8931 in 41% overall yield.

2-[4-[4-(3-chloro-2-fluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-1-piperidyl]-N-methyl-acetamide IPA solvate (5) as a white solid (38.1 kg, 84.2% yield); 1H NMR (400 MHz, DMSO-d6) δ ppm 1.81 (m, 2 H), 2.06 (m, 2 H), 2.39 (m, 2 H), 2.63 (d, J = 4.7 Hz, 3 H), 2.75 (m, 2 H), 2.95 (s, 2 H), 3.94 (s, 3 H), 4.57 (Dt, J = 8.1, 4.2 Hz, 1 H), 7.22 (s, 1 H), 7.29 (t, J = 8.0 Hz, 1 H), 7.51 (m, 2 H), 7.74 (br d, J = 4.6 Hz, 1 H), 7.83 (s, 1 H), 8.37 (s, 1 H), 9.58 (br.s, 1 H); m/Z ES+ 474.2 [MH]+; HRMS found [MH]+ = 474.1706, C23H25ClFN5O3 requires [MH]+ = 474.1630; Assay (QNMR) 97.5 wt %/wt.

1H NMR PREDICT

13C NMR PREDICT

CHEMICAL & PHARMACEUTICAL BULLETIN, vol. 49, no. 7, 2001, pages 822 – 829
Citing Patent Filing date Publication date Applicant Title
WO2013051883A3 * Oct 5, 2012 Jun 6, 2013 Hanmi Science Co., Ltd. Method for preparing 1-(4-(4-(3,4-dichloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yloxy)piperidin-1-yl)-prop-2-en-1-one hydrochloride and intermediates used therein
US8859767 Oct 5, 2012 Oct 14, 2014 Hanmi Science Co., Ltd Method for preparing 1-(4-(4-(3,4-dichloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yloxy)piperidin-1-yl)-prop-2-en-1-one hydrochloride and intermediates used therein

////////////////AZD 8931, Sapitinib, pan-EGFR, pan-erbB inhibitor, SAPATINIB, PHASE 2, 848942-61-0

CNC(=O)CN1CCC(CC1)OC2=C(C=C3C(=C2)C(=NC=N3)NC4=C(C(=CC=C4)Cl)F)OC

“ALL FOR DRUGS” CATERS TO EDUCATION GLOBALLY, No commercial exploits are done or advertisements added by me. This article is a compilation for educational purposes only.

P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent


Filed under: Phase2 drugs Tagged: 848942-61-0, AZD 8931, pan-EGFR, pan-erbB inhibitor, phase 2, SAPATINIB, Sapitinib

Zydus receives approval from USFDA to initiate Phase II clinical studies of Saroglitazar Magnesium in patients with Primary Biliary Cholangitis (PBC)

$
0
0

Zydus receives approval from USFDA to initiate Phase II clinical studies of Saroglitazar Magnesium in patients with Primary Biliary Cholangitis (PBC) Read more: https://goo.gl/eugRnZ #ZydusAnnouncement

Zydus receives approval from USFDA to initiate Phase II clinical studies of Saroglitazar Magnesium in patients with Primary Biliary Cholangitis (PBC)

Ahmedabad, India, February 23, 2017

Image result for INDIAN FLAG ANIMATED

Zydus Cadila, a research-driven, global healthcare provider, today announced that the USFDA has approved the group’s plans to initiate a Phase 2 clinical trial of Saroglitazar Magnesium (Mg) in patients with Primary Biliary Cholangitis (PBC) of the liver. This randomized, double-blind Phase 2 trial will evaluate Saroglitazar Magnesium 2mg and 4 mg Vs. Placebo.

Speaking on the development, Mr. Pankaj R. Patel, Chairman and Managing Director, Zydus Cadila said, “We are very thankful to the USFDA for their timely and useful feedback on the clinical trial designs of Saroglitazar Mg in patients with Primary Biliary Cholangitis (PBC). This development underlines our commitment to bridging unmet healthcare needs with innovative therapies.”

Primary Biliary Cholangitis (PBC) is a liver disease, caused due to progressive destruction of the bile ducts in the liver which leads to reduction of bile flow – a condition referred to as cholestasis. PBC is often discovered incidentally due to abnormal results on routine liver blood tests. Progression of PBC leads to symptoms of cirrhosis like yellowing of the skin, swelling of legs and feet (edema), ascites, internal bleeding (varices) and thinning of the bones (osteoporosis). The buildup of toxic bile in the liver leads to liver inflammation and fibrosis which can progress to cirrhosis. People with cirrhosis are at increased risk of hepatocellular carcinoma or liver cancer, which is a leading cause of liver transplants or death.

With an increasing number of people being affected by PBC which can lead to progressive cholestasis and even turn fatal, there is a pressing need to develop therapies which help to achieve an adequate reduction in alkaline phosphotase (ALP) or bilirubin and bring in better tolerance and efficacy.

About Lipaglyn™ Lipaglyn™ is a prescription drug authorized for sale in India only. Lipaglyn™ was launched in India during Sept 2013 for the treatment of Hypertriglyceridemia and Diabetic Dyslipidemia in Patients with Type 2 Diabetes not controlled by statins. Saroglitazar Mg is an investigational new drug with the USFDA, and is currently under clinical investigation for three significant unmet medical needs in the United States – Primary Biliary Cholangitis (PBC), Non-alcoholic Steatohepatitis (NASH) and Severe Hypertriglyceridemia (TG>500).

About Zydus Zydus Cadila is an innovative, global healthcare provider that discovers, develops, manufactures and markets a broad range of healthcare therapies, including small molecule drugs, biologic therapeutics and vaccines. The group employs over 19,500 people worldwide, including 1200 scientists engaged in R & D, and is dedicated to creating healthier communities globally. For more information, please visit http://www.zyduscadila.com

http://zyduscadila.com/wp-content/uploads/2017/02/USFDA-approval-for-clinical-trial-of-Saro-Mg.pdf

Image result for Saroglitazar Magnesium

Image result for Saroglitazar Magnesium

Saroglitazar magnesium
CAS: 1639792-20-3

Molecular Formula, 2C25-H28-N-O4-S.Mg,

Molecular Weight, 901.4354

Magnesium, bis((alphaS)-alpha-(ethoxy-kappaO)-4-(2-(2-methyl-5-(4-(methylthio)phenyl)-1H-pyrrol-1-yl)ethoxy)benzenepropanoato-kappaO)-, (T-4)-

(2S)-2-Ethoxy-3-(4-(2-(2-methyl-5-(4-(methylsulfanyl)phenyl)-1H-pyrrol-1-yl(ethoxy)phenyl)propanoic acid, magnesium salt (2:1)

Image result for RANJIT DESAI ZYDUS

DR RANJIT DESAI

ZYDUS

Image result for INDIAN FLAG ANIMATED

//////////Zydus,  USFDA, Phase II,  clinical studies, Saroglitazar Magnesium,  Primary Biliary Cholangitis,  (PBC)

[Mg+2].CCO[C@@H](Cc1ccc(OCCn2c(C)ccc2c3ccc(SC)cc3)cc1)C(=O)[O-].CCO[C@@H](Cc4ccc(OCCn5c(C)ccc5c6ccc(SC)cc6)cc4)C(=O)[O-]


Filed under: Phase2 drugs Tagged: (PBC), clinical studies, Phase II, Primary Biliary Cholangitis, Saroglitazar magnesium, usfda, zydus

Zydus Cadila to launch India’s 1st Tetravalent Inactivated Influenza vaccine – VaxiFlu – 4

$
0
0

Zydus Cadila to launch India’s 1st Tetravalent Inactivated Influenza vaccine – VaxiFlu – 4

Ahmedabad, February 24, 2017

Zydus Cadila, a research-driven, global healthcare provider has received approvals from the Drug Controller General of India (DCGI), Central Drugs Standard Control Organization (CDSCO) and the Central Drug Laboratory (CDL) to market the Tetravalent Inactivated Influenza vaccine for seasonal flu, VaxiFlu – 4. With this, Zydus Cadila will become the first Indian pharma company and second in the world to launch a Tetravalent Inactivated Influenza vaccine. The vaccine provides protection from the four influenza viruses- H1N1, H3N2, Type B (Brisbane) and Type B (Phuket).

Image result for VaxiFlu - 4

VaxiFlu – 4 will be marketed by Zydus Vaxxicare – a division of the group focussing on preventives. The Tetravalent Inactivated Influenza vaccine has been developed at the Vaccine Technology Centre (VTC) in Ahmedabad which has proven capabilities in researching, developing, and manufacturing of safe and efficacious vaccines. The group was also the first to indigenously develop, manufacture and launch India’s first vaccine against H1N1 – Vaxiflu-S.

VTC further plans to develop a wide spectrum of vaccines against bacterial, viral and protozoal infections and has a robust pipeline of vaccines like Pentavalent (DTP-Hib-HepB), Conjugated Typhoid Vaccine, HPV, MMRV, Malaria and Hepatitis B vaccines. The group also markets the anti-rabies vaccine and the typhoid vaccine.

Speaking on the development Mr. Pankaj R. Patel, Chairman and Managing Director, Zydus Cadila said, “Disease prevention is the key to public health in both the developing and the developed world and vaccines have the potential to improve the quality of life in both spectrums. In countries such as India, there is a pressing need for low cost, high quality vaccines that can address healthcare challenges. With the launch of vaccines like VaxiFlu – 4 we are serving the cause of public health and meeting the twin challenge of affordability and accessibility.”

Influenza, or the “flu” as it is commonly called, is an infection of the respiratory tract. It is a dreaded disease and the morbidity and mortality rates associated with influenza are especially high during pandemics. Annually it is estimated that it attacks 5-10% of adults and 20-30% of children globally and causes significant levels of illness, hospitalization and death. In India, the 2009 swine flu pandemic infected more than 10 million people and resulted in more than 18000 deaths worldwide.

The last major outbreak in India occurred in 2015 with more than 33000 registered cases of influenza and over 2000 deaths. There are different strains of influenza viruses that infect human beings, the predominant ones being influenza A and influenza B. The common subtypes of influenza A found in general circulation amongst people are H1N1 (which was responsible for the devastating swine flu pandemic) and H3N2.

The subtypes of influenza B commonly found in circulation are influenza B (Brisbane – Victoria lineage) and influenza B (Phuket – Yamagata lineage). Vaccination against influenza is the most effective way to protect oneself against the dangers of influenza. Majority of the influenza vaccines available in India are inactivated trivalent influenza vaccines.

These vaccines provide protection against 2 strains of influenza A and 1 strain of influenza B. Protection against only 1 subtype of influenza B often leads to a vaccine mismatch i.e. the antigen of influenza B present in the trivalent vaccine may not match the influenza B subtype circulating during the season, leading to suboptimal protection. A quadrivalent vaccine, by virtue of having a comprehensive coverage against 2 strains of both influenza A and influenza B, provides a broader protection and significantly reduces the risk of vaccine mismatch. Vaxiflu – 4 is the first quadrivalent influenza vaccine in india.

About Zydus Zydus Cadila is an innovative, global pharmaceutical company that discovers, develops, manufactures and markets a broad range of healthcare therapies, including small molecule drugs, biologic therapeutics and vaccines. The group employs over 19,500 people worldwide, including 1200 scientists engaged in R & D, and is dedicated to creating healthier communities globally. For more information, please visit http://www.zyduscadila.com

Zydus’ vaccine research programme The Vaccine Technology Centre (VTC) is the vaccine research centre of the Zydus Group. The group has two state-of-the-art R & D Centers, one located in Catania, Italy and the other in Ahmedabad, in the western part of India. The goup has been developing vaccines for the basic vaccine programmes such as Diphtheria, Pertussis, Tetanus, Haemophilus Influenzae type B, Hepatitis B, Measles, Mumps, Rubella, Varicella, Influenza and Typhoid fever. In addition, it is developing new vaccines such as Human Papilloma Virus, Leishmaniasis, Malaria, Haemorrhagic Congo Fever, Ebola and Japanese Encephalitis.

Ref

Zydus Cadila to launch India’s 1st Tetravalent Inactivated Influenza vaccine – VaxiFlu – 4 Read more: https://goo.gl/xuSTfK #ZydusAnnouncement

Image result for Trivalent Inactivated Influenza vaccine

Image result for Trivalent Inactivated Influenza vaccine

///////////Zydus Cadila, Tetravalent Inactivated,  Influenza vaccine, VaxiFlu – 4


Filed under: Uncategorized, vaccine Tagged: Influenza vaccine, Tetravalent Inactivated, VaxiFlu - 4, zydus cadila

Tradipitant, традипитант , تراديبيتانت , 曲地匹坦 ,

$
0
0

LY686017.svgTradipitant.png

Tradipitant

VLY-686,  LY686017

традипитант
تراديبيتانت [Arabic]
曲地匹坦 [Chinese]
  • Molecular Formula C28H16ClF6N5O
  • Average mass 587.903 Da
622370-35-8  CAS
Methanone, [2-[1-[[3,5-bis(trifluoromethyl)phenyl]methyl]-5-(4-pyridinyl)-1H-1,2,3-triazol-4-yl]-3-pyridinyl](2-chlorophenyl)-
(2-(1-(3,5-bis(trifluoromethyl)benzyl)-5-(pyridin-4-yl)-1H-1,2,3-triazol-4-yl)pyridin-3-yl)(2-chlorophenyl)methanone
[2-[1-[[3,5-bis(trifluoromethyl)phenyl]methyl]-5-(4-pyridinyl)-1H-1,2,3-triazol-4-yl]-3-pyridinyl](2-chlorophenyl)methanone

PHASE 2, Gastroparesis; Pruritus

pyridine-containing NK-1 receptor antagonist ie tradipitant, useful for treating anxiety, pruritus and alcoholism.

Vanda Pharmaceuticals, under license from Eli Lilly, was developing tradipitant, a NK1 antagonist, for treating anxiety disorder, pruritus and alcohol dependence. The company was also investigating the drug for treating gastroparesis. In February 2017, tradipitant was reported to be in phase 2 clinical development for treating anxiety and pruritus.

  • Originator Eli Lilly
  • Developer Eli Lilly; National Institute on Alcohol Abuse and Alcoholism; Vanda Pharmaceuticals
  • Class Antipruritics; Anxiolytics; Chlorobenzenes; Pyridines; Small molecules; Triazoles
  • Mechanism of Action Neurokinin 1 receptor antagonists; Substance P inhibitors

Highest Development Phases

  • Phase II Gastroparesis; Pruritus
  • Discontinued Alcoholism; Social phobia
  • The drug had been in phase II clinical trials at Lilly and the National Institute on Alcohol Abuse and Alcoholism for the treatment of alcoholism; however, no recent development has been reported for this research.
  • A phase II clinical trial for the treatment of social phobia has been completed by Lilly.

PATENT WO 2003091226

Albert Kudzovi Amegadzie, Kevin Matthew Gardinier, Erik James Hembre, Jian Eric Hong, Louis Nickolaus Jungheim, Brian Stephen Muehl, David Michael Remick, Michael Alan Robertson, Kenneth Allen Savin, Less «
Applicant Eli Lilly And Company

Image result for Eli Lilly And Company

Image result for tradipitant

SYNTHESIS

Condensation of 2-chloropyridine with thiophenol  in the presence of K2CO3 in DMF at 110ºC yields sulfide intermediate,

which is then oxidized by means of NaOCl in AcOH to give 2-(benzenesulfonyl)pyridine.

This is treated with (iPr)2NH and n-BuLi in THF at -60 to -70°C and subsequently couples with 2-chlorobenzaldehyde  in THF at -60 to -70°C to furnish (2-(phenylsulfonyl)pyridin-3-yl)-(2-chlorophenyl)methanone.

Ketone  couples with the enolate of 4-acetylpyridine (formed by treating 4-acetylpyridine (VII) with t-BuOK in DMSO) in the presence of LiOH in DMSO and subsequently is treated with PhCOOH in iPrOAc to give rise to pyridine benzoate derivative.

This finally couples with 1-azidomethyl-3,5-bistrifluoromethylbenzene  (obtained by treating 3,5-bis(trifluoromethyl)benzylchloride with NaN3 ini DMSO) in the presence of K2CO3 in t-BuOH to afford the title compound Tradipitant.

Tradipitant (VLY-686 or LY686017) is an experimental drug that is a neurokinin 1 antagonist. It works by blocking substance P, a small signaling molecule. Originally, this compound was owned by Eli Lilly and named LY686017. VLY-686 was purchased by Vanda Pharmaceuticals from Eli Lilly and Company in 2012.[1] Vanda Pharmaceuticals is a U.S. pharmaceutical company that as of November 2015 only has 3 drugs in their product pipeline: tasimelteon, VLY-686, and iloperidone.[2]

Tachykinins are a family of peptides that are widely distributed in both the central and peripheral nervous systems. These peptides exert a number of biological effects through actions at tachykinin receptors. To date, three such receptors have been characterized, including the NK-1 , NK-2, and NK-3 subtypes of tachykinin receptor.

The role of the NK-1 receptor subtype in numerous disorders of the central nervous system and the periphery has been thoroughly demonstrated in the art. For instance, NK-1 receptors are believed to play a role in depression, anxiety, and central regulation of various autonomic, as well as cardiovascular and respiratory functions. NK- 1 receptors in the spinal cord are believed to play a role in pain transmission, especially the pain associated with migraine and arthritis. In the periphery, NK-1 receptor activation has been implicated in numerous disorders, including various inflammatory disorders, asthma, and disorders of the gastrointestinal and genitourinary tract.

There is an increasingly wide recognition that selective NK-1 receptor antagonists would prove useful in the treatment of many diseases of the central nervous system and the periphery. While many of these disorders are being treated by new medicines, there are still many shortcomings associated with existing treatments. For example, the newest class of anti-depressants, selective serotonin reuptake inhibitors (SSRIs), are increasingly prescribed for the treatment of depression; however, SSRIs have numerous side effects, including nausea, insomnia, anxiety, and sexual dysfunction. This could significantly affect patient compliance rate. As another example, current treatments for chemotherapy- induced nausea and emesis, such as the 5-HT3receptor antagonists, are ineffective in managing delayed emesis. The development of NK-1 receptor antagonists will therefore greatly enhance the ability to treat such disorders more effectively. Thus, the present invention provides a class of potent, non-peptide NK-1 receptor antagonists, compositions comprising these compounds, and methods of using the compounds.

Indications

Pruritus

It is being investigated by Vanda Pharmaceuticals for chronic pruritus (itchiness) in atopic dermatitis. In March 2015, Vanda announced positive results from a Phase II proof of concept study.[3] A proof of concept study is done in early stage clinical trials after there have been promising preclinical results. It provides preliminary evidence that the drug is active in humans and has some efficacy.[4]

Alcoholism

VLY-686 reduced alcohol craving in recently detoxified alcoholic patients as measured by the Alcohol Urge Questionnaire.[5] In a placebo controlled clinical trial of recently detoxified alcoholic patients, VLY-686 significantly reduced alcohol craving as measured by the Alcohol Urge Questionnaire. It also reduced the cortisol increase seen after a stress test compared to placebo. The dose given was 50 mg per day.

Social anxiety disorder

In a 12-week randomized trial of LY68017 in 189 patients with social anxiety disorder, 50 mg of LY68017 did not provide any statistically significant improvement over placebo.[6]

PATENT

WO03091226,

https://www.google.com/patents/WO2003091226A1?cl=en

PATENT

WO2008079600, 

The compound {2-[l-(3,5-bis-trifluoromethyl-benzyl)-5-pyridin-4-yl-lH-[l,2,3]triazol-4-yl]- pyridin-3-yl}-(2-chlorophenyl)-methanone, depicted below as the compound of Formula I, was first described in PCT published application WO2003/091226.

Figure imgf000003_0001

(I)

Because the compound of Formula I is an antagonist of the NK-I subtype of tachykinin receptor, it is useful for the treatment of disorders associated with an excess of tachykinins. Such disorders include depression, including major depressive disorder; anxiety, including generalized anxiety disorder, panic disorder, obsessive compulsive disorder, and social phobia or social anxiety disorder; schizophrenia and other psychotic disorders, including bipolar disorder; neurodegenerative disorders such as dementia, including senile dementia of the Alzheimer’s type or Alzheimer’s disease; disorders of bladder function such as bladder detrusor hyper-reflexia and incontinence, including urge incontinence; emesis, including chemotherapy-induced nausea and acute or delayed emesis; pain or nociception; disorders associated with blood pressure, such as hypertension; disorders of blood flow caused by vasodilation and vasospastic diseases, such as angina, migraine, and Reynaud’s disease; hot flushes; acute and chronic obstructive airway diseases such as adult respiratory distress syndrome, bronchopneumonia, bronchospasm, chronic bronchitis, drivercough, and asthma; inflammatory diseases such as inflammatory bowel disease; gastrointestinal disorders or diseases associated with the neuronal control of viscera such as ulcerative colitis, Crohn’s disease, functional dyspepsia, and irritable bowel syndrome (including constipation-predominant, diarrhea- -?-

predominant, and mixed irritable bowel syndrome); and cutaneous diseases such as contact dermatitis, atopic dermatitis, urticaria, and other eczematoid dermatitis.

In PCT published application, WO2005/042515, novel crystalline forms of the compound of Formula I, identified as Form IV and Form V, are identified. Also described in WO2005/042515 is a process for preparation of the compound of Formula I, comprising reacting (2-chlorophenyl)-[2-(2- hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone or a phosphate salt thereof with l-azidomethyl-3,5- bistrifluoromethylbenzene in the presence of a suitable base and a solvent. Use of this procedure results in several shortcomings for synthesis on a commercial scale. For example, use of the solvent DMSO, with (2- chlorophenyl)-[2-(2-hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone phosphate, requires a complex work-up that has a propensity to emulsify. This process also requires extraction with CH2CI2, the use of which is discouraged due to its potential as an occupational carcinogen, as well as the use of MgSC>4 and acid-washed carbon, which can generate large volumes of waste on a commercial scale. Conducting the reaction with (2-chlorophenyl)-[2-(2-hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone in isopropyl alcohol, as also described in WO2005/042515, is also undesirable due to the need to incorporate a free base step. Furthermore, variable levels of residual l-azidomethyl-3,5-bistrifluoromethylbenzene, a known mutagen, are obtained from use of the procedures described in WO2005/042515.

An improved process for preparing the compound of Formula I would control the level of 1- azidomethyl-3,5-bistrifluoromethylbenzene impurity, and improve the yield. We have discovered that use of the novel salt, (2-chlorophenyl)-[2-(2-hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone benzoate, as well as use of tert-butanol as the reaction solvent, improves reaction times and final yield, and decreases impurities in the final product. In addition, a novel process for the preparation of (2-chlorophenyl)- [2-(2- hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone benzoate, in which a pre-formed enolate of 4-acetyl pyridine is added to (2-phenylsulfonyl-pyridine-3-yl)-(2-chlorophenyl)methanone, results in an overall improved yield and improved purity, and is useful on a commercial scale.

EXAMPLES

Example 1 {2-[l-(3,5-bistrifluoromethylbenzyl)-5-pyridin-4-yl-lH-[l,2,3]triazol-4-yl]-pyridin-3-yl}-(2-chlorophenyl)- methanone (Form IV)

Figure imgf000005_0001

Suspend (2-chlorophenyl)-[2-(2-hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl] methanone benzoate (204.7 g; 1.04 equiv; 445 mmoles) in t-butanol (614 mL) and treat the slurry with potassium carbonate (124.2 g; 898.6 mmoles). Heat to 7O0C with mechanical stirring for 1 hour. Add l-azidomethyl-3,5- bistrifluoromethylbenzene (115.6 g; 1.00 equiv; 429.4 mmoles) in a single portion, then heat the mixture to reflux. A circulating bath is used to maintain a condenser temperature of 3O0C. After 18 hours at reflux, HPLC reveals that the reaction is complete (<2% l-azidomethyl-3,5-bistrifluoromethylbenzene remaining). The mixture is cooled to 7O0C, isopropanol (818 mL) is added, then the mixture is stirred at 7O0C for 1 hour. The mixture is filtered, and the waste filter cake is rinsed with isopropanol (409 mL). The combined filtrate and washes are transferred to a reactor, and the mechanically stirred contents are heated to 7O0C. To the dark purple solution, water (1.84 L) is added slowly over 35 minutes. The solution is cooled to 6O0C, then stirred for 1 hour, during which time a thin precipitate forms. The mixture is slowly cooled to RT, then the solid is filtered, washed with 1 : 1 isopropanol/water (614 mL), subsequently washed with isopropanol (410 mL), then dried in vacuo at 450C to produce 200.3 g of crude {2-[l-(3,5- bistrifluoromethylbenzyl)-5-pyridin-4-yl-lH-[l,2,3]triazol-4-yl]-pyridin-3-yl}-(2-chlorophenyl)-methanone as a white solid. Crude {2-[l-(3,5-bistrifluoromethylbenzyl)-5-pyridin-4-yl-lH-[l,2,3]triazol-4-yl]-pyridin- 3-yl}-(2-chlorophenyl)-methanone (200.3 g) and isopropyl acetate (600 mL) are charged to a 5L 3-neck jacketed flask, then the contents heated to 750C. After dissolution is achieved, the vessel contents are cooled to 550C, then the solution polish filtered through a 5 micron filter, and the filter rinsed with a volume of isopropyl acetate (200 mL). After the polish filtration operation is complete, the filtrates are combined, and the vessel contents are adjusted to 5O0C. After stirring for at least 15 minutes at 5O0C, 0.21 grams of {2-[l-(3,5-bistrifluoromethylbenzyl)-5-pyridin-4-yl-lH-[l,2,3]triazol-4-yl]-pyridin-3-yl}-(2- chlorophenyl)-methanone Form IV seed (d90 = 40 microns) is added, and the mixture stirred at 5O0C for at least 2 h. Heptanes (1.90 L) are then added over at least 2 h. After the heptanes addition is completed, the slurry is stirred for an hour at 5O0C, cooled to 230C at a rate less then 2O0C per hour, then aged at 230C for an hour prior to isolation. The mixture is then filtered in portions through the bottom outlet valve in the reactor into a 600 mL filter. The resulting wetcake is washed portionwise with a solution containing heptanes (420 mL) and isopropyl acetate (180 mL), which is passed directly through the 5L crystallization vessel. The wetcake is blown dry for 5 minutes with nitrogen, then transferred to a 500 mL plastic bottle. The product is dried at 5O0C for 4 h. to produce 190.3g of pure {2-[l-(3,5- bistrifluoromethylbenzyl)-5-pyridin-4-yl-lH-[l,2,3]triazol-4-yl]-pyridin-3-yl}-(2-chlorophenyl)- methanone, Form IV in 75.0% yield with 100% purity, as determined by HPLC analysis. Particle size is reduced via pin or jet mill. 1H NMR (400 MHz, CDCl3): 5.46 (s, 2H); 7.19 (m, 5H); 7.36 (dd, IH, J = 4.9, 7.8); 7.45 (s, 2H); 7.59 (m, IH); 7.83 (s, IH); 7.93 (dd, IH, J = 1.5, 7.8); 8.56 (dd, IH, J= 1.5, 4.9); 8.70 (d, 2H, J= 5.9).

Preparation 1-A (2-chlorophenyl)-[2-(2-hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone benzoate Charge powdered KOfBu (221.1 g, 1.93 moles, 1.40 eq.) to Reactor A, then charge DMSO (2 L) at

250C over 10 min. The KOfBu/DMSO solution is stirred for 30 min at 230C, then a solution of 4-acetyl pyridine (92 mL, 2.07 moles, 1.50 eq) in DMSO (250 mL) is prepared in reactor B. The contents of reactor B are added to Reactor A over 10 minutes, then the Reactor A enolate solution is stirred at 230C for Ih. In a separate 12-L flask (Reactor C), solid LiOH (84.26 g, 3.45 moles, 2.0 eq) is poured into a mixture of (2- phenylsulfonyl-pyridin-3-yl)-(2-chlorophenyl)methanone (500.0 g, 1.34 moles, 1.0 eq) and DMSO (2L), with stirring, at 230C. The enolate solution in reactor A is then added to Reactor C over a period of at least 15 minutes, and the red suspension warmed to 4O0C. The reaction is stirred for 3h, after which time HPLC analysis reveals less than 2% (2-phenylsulfonyl-pyridin-3-yl)-(2-chlorophenyl)methanone. Toluene (2.5 L) is charged, and the reactor temperature cooled to 3O0C. The mixture is quenched by addition of glacial acetic acid (316 mL, 5.52 moles, 4.0 eq), followed by 10 % NaCl (2.5 L). The biphasic mixture is transferred to a 22-L bottom-outlet Morton flask, and the aqueous layer is removed. The aqueous layer is then extracted with toluene (750 mL). The combined organic layers are washed with 10 % NaCl (750 mL), then concentrated to 4 volumes and transferred to a 12-L Morton flask and rinsed with isopropyl acetate (4 vol, 2 L). The opaque amber solution is warmed to 75 degrees to 750C over 40 min. Benzoic acid (171. Ig, 1.34 moles, 1.0 eq) is dissolved in hot isopropyl acetate (1.5 L), and charged to the crude free base solution over at least 30 min. The crude solution containing benzoate salt is stirred for 0.5 h at 750C then cooled to 23 0C. When solids are first observed, the cooling is stopped and the mixture is aged for an hour at the temperature at which crystals are first observed. Alternatively, if seed crystal is available, the mixture may be seeded with (2-chlorophenyl)-[2-(2-hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone benzoate (2.25g) at 750C, followed by stirring for 0.5 h at 750C, then cooling to 230C over at least 1.5 h. The mixture is then cooled to <5 0C, then filtered through paper on a 24cm single-plate filter. The filtercake is then rinsed with cold z-PrOAc (750 mL) to produce granular crystals of bright orange-red color. The wet solid is dried at 550C to produce 527.3 g (83% yield) with 99.9% purity. (2-chlorophenyl)-[2-(2-hydroxy-2- pyridin-4-yl-vinyl)pyridin-3-yl]methanone benzoate. Anal. Calcd. for C26Hi9N2ClO4: C, 68.05; H, 4.17; N, 7.13. Found: C, 67.89; H, 4.15; N 6.05. HRMS: calcd for C19H13ClN2O2, 336.0666; found 336.0673.

The synthesis of(2-chlorophenyl)-[2-(2-hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone benzoate proceeds optimally when the potassium enolate of 4-acetyl pyridine is pre-formed using KOfBu in DMSO. Pre-formation of the enolate allows the SNAR (nucleophilic aromatic substitution) reaction to be performed between room temperature and 4O0C, which minimizes the amount of degradation. Under these conditions, the SNAR is highly regioselective, resulting in a ratio of approximately 95:5 preferential C – acylation. In all cases, less polar solvents such as THF or toluene, or co-solvents of these solvents mixed with DMSO, results in a substantial increase of acylation at the oxygen in the SNAR, and leads to a lower yield of product. This is a substantial improvement over the procedures described in WO2005/042515 for synthesis of the free base or the phosphate salt, in which the SNAR is performed at 60-700C, resulting in a substantial increase in chemical impurity. Using the conditions described in WO2005/042515, when scaled to 2kg, results in maximum yields of 55%, with sub-optimal potency. In comparison, the improved conditions described herein can be run reproducibly from 0.4 to 2kg scale to give yields of 77-83%, with >99% purity. In addition, the reaction can be held overnight at 4O0C with minimal degradation, whereas holding the reaction for 1 h past completion at 60-70°C results in substantial aromatized impurity. The reaction may also be performed using sodium tert-amylate as the base, in combination with an aprotic solvent, such as DMSO or DMF.

The title compound exists as a mixture of tautomers and geometric isomers. It is understood that each of these forms is encompassed within the scope of the invention.

Figure imgf000008_0001

Preparation 1-B

(2-chlorophenyl)-[2-(2-hydroxy-2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone toluate The procedure described in Preparation 1-A is followed, with the following exception. Solid toluic acid (1.0 eq) is added to the crude free base solution at 550C, then the solution cooled to 45 0C. The solution is stirred for one hour at 45 0C, then slowly cooled to 23 0C. When solids are first observed, the cooling is stopped and the mixture is aged for an hour at the temperature at which crystals are first observed. Alternatively, if seed crystal is available, the mixture may be seeded, aged for 3 h at 450C , then cooled to O0C over 4 h. The isolation slurry is filtered, and the wetcake washed with MeOH (3 volumes). The wetcake is dried at 5O0C to provide 14.0 g (76.4%) of (2-chlorophenyl)-[2-(2-hydroxy-2-pyridin-4-yl- vinyl)pyridin-3-yl]methanone toluate as a light red powder.

As with the benzoate salt, the toluate salt can also exist as a mixture of tautomers and geometric isomers, each of which is encompassed within the scope of the invention. (2-chlorophenyl)-[2-(2-hydroxy- 2-pyridin-4-yl-vinyl)pyridin-3-yl]methanone toluate . 13C NMR (125 MHz,DMS0-d6) δ 194.5, 167.8, 167.4, 155.5, 150.7 (2C), 147.4, 144.0, 143.4, 142.7, 138.6, 133.0, 130.8, 130.7, 130.5, 129.8(2C), 129.5(2C), 128.5, 128.0, 127.9, 119.9 (2C), 118.6, 92.6, 21.5.

Preparation 1-C

(2-phenylsulfonyl-pyridin-3-yl)-(2-chlorophenyl)methanone

A solution of 1.3 eq of diisopropylamine (based on 2-benzenesulfonyl pyridine) in 5 volumes of THF in a mechanically stirred 3 -necked flask is cooled to -70 to -75 0C. To this solution is added 1.05 eq of w-butyllithium (1.6M in hexanes) at such a rate as to maintain the temperature below -6O0C. The light yellow solution is stirred at -60 to -70 0C for 30 minutes. Once the temperature has cooled back down to – 60 to -650C, 1.0 eq of 2-benzene-sulfonyl pyridine, as a solution in 3 volumes of THF, is added at the fastest rate that will maintain the reaction temperature under -6O0C. A yellow suspension forms during the addition that becomes yellow-orange upon longer stirring. This mixture is stirred for 3 hours at -60 to – 750C, and then 1.06 eq of 2-chlorobenzaldehyde, as a solution in 1 volume of THF, is added dropwise at a sufficient rate to keep the temperature under -55 0C. The suspension gradually turns orange-red, thins out, and then becomes a clear red solution. The reaction mixture is allowed to stir at -60 to -7O0C for 1 hour, 3N aqueous HCl (7 volumes) is added over 20-30 minutes, and the temperature is allowed to exotherm to 0-100C. The color largely disappears, leaving a biphasic yellow solution. The solution is warmed to at least 1O0C, the layers are separated, and the aqueous layer is back-extracted with 10 volumes of ethyl acetate. The combined organic layers are washed with 10 volumes of saturated sodium bicarbonate solution and concentrated to about 2 volumes. Ethyl acetate (10 volumes) is added, and the solution is once again concentrated to 2 volumes. The thick solution is allowed to stand overnight and is taken to the next step with no purification of the crude alcohol intermediate. The crude alcohol intermediate is transferred to a 3 -necked flask with enough ethyl acetate to make the total solution about 10 volumes. The yellow solution is treated with 3.2 volumes of 10% aqueous (w/w) potassium bromide, followed by 0.07 eq of 2,2,6,6-Tetramethylpiperidine-N-oxide (TEMPO). The orange mixture is cooled to 0-50C and treated with a solution of 1.25 eq of sodium bicarbonate in 12% w/w sodium hypochlorite (9 volumes) and 5 volumes of water over 30-60 minutes while allowing the temperature to exotherm to a maximum of 2O0C. The mixture turns dark brown during the addition, but becomes yellow, and a thick precipitate forms. The biphasic light yellow mixture is allowed to stir at ambient temperature for 1-3 hours, at which time the reaction is generally completed. The biphasic mixture is cooled to 0-50C and stirred for 3 hours at that temperature. The solid is filtered off, washed with 4 volumes of cold ethyl acetate, followed by 4 volumes of water, and dried in vacuo at 450C to constant weight. Typical yield is 80-83% with a purity of greater than 98%. 1H NMR (600 MHz, CDCl3-^) δ ppm 7.38 (td, ./=7.52, 1.28 Hz, 1 H) 7.47 (dd, ./=7.80, 1.30 Hz, 1 H) 7.51 (td, ./=7.79, 1.60 Hz, 1 H) 7.51 (t, ./=7.89 Hz, 2 H) 7.50 – 7.54 (m, J=7.75, 4.63 Hz, 1 H) 7.60 (t, J=7.43 Hz, 1 H) 7.73 (dd, J=7.75, 1.60 Hz, 1 H) 7.81 (dd, J=7.79, 1.56 Hz, 1 H) 8.00 (dd, ./=8.44, 1.10 Hz, 2 H) 8.76 (dd, ./=4.63, 1.61 Hz, 1 H).

Preparation 1-D 1 -azidomethyl-3,5-bistrifluoromethyl-benzene

Sodium azide (74.3 g, 1.14 mol) is suspended in water (125 mL), then DMSO (625 mL) is added. After stirring for 30 minutes, a solution consisting of 3,5-Bis(trifluoromethyl)benzyl chloride (255.3 g, 0.97 moles) and DMSO (500 mL) is added over 30 minutes. (The 3,5-Bis(trifluoromethyl)benzyl chloride is heated to 350C to liquefy prior to dispensing (MP = 30-320C)). The benzyl chloride feed vessel is rinsed with DMSO (50 mL) into the sodium azide solution, the mixture is heated to 4O0C, and then maintained for an hour at 4O0C, then cooled to 230C.

In Process Analysis: A drop of the reaction mixture is dissolved in d6-DMSO and the relative intensities of the methylene signals are integrated (NMR verified as a 0.35% limit test for 3,5- Bis(trifluoromethyl)benzyl Chloride). Work-up: After the mixture reaches 230C , it is diluted with heptanes (1500 mL), then water (1000 mL) is added, and the mixture exotherms to 350C against a jacket setpoint of 230C. The aqueous layer is removed (-2200 mL), then the organic layer (approximately 1700 mL) is washed with water (2 X 750 mL). The combined aqueous layers (-3700 mL) are analyzed and discarded.

The solvent is then partially removed via vacuum distillation with a jacket set point of 850C, pot temperature of 60-650C and distillate head temperature of 50-550C to produce 485g (94.5% yield) of 51 Wt% solution title compound as a clear liquid. Heptanes can be either further removed by vacuum distillation or wiped film evaporation technology. 1H NMR (400 MHz, CDCl3): 4.58 (s, 2H); 7.81 (s, 2H); 7.90 (s, IH).

Preparation 1-E 2-benzene-sulfonyl pyridine Charge 2-chloropyridine (75 mL, 790 mmol), thiophenol (90 mL, 852 mmol), and DMF (450 mL) to a 2L flask. Add K2CO3 (134.6 g, 962 mmol), then heat to HO0C and stir for 18 hours. Filter the mixture, then rinse the waste cake with DMF (195 mL). The combined crude sulfide solution and rinses are transferred to a 5-L flask, and the waste filtercake is discarded. Glacial acetic acid (57 mL, 995 mmol) is added to the filtrate, then the solution is heated to 4O0C, and 13 wt % NaOCl solution (850 mL, 1.7 mol) is added over 2 hours. After the reaction is complete, water (150 mL) is added, then the pH of the mixture adjusted to 9 with 20 % (w/v) NaOH solution (250 mL). The resulting slurry is cooled to <5 0C, stirred for 1.5 h, then filtered, and the cake washed with water (3 x 200 mL). The product wetcake is dried in a 550C vacuum oven to provide 2-benzene-sulfonyl pyridine (149 g, 676 mmol) in 86 % yield: 1H NMR (500 MHz, CDCl3) δ 8.66 (d, J = 5.5 Hz, IH), 8.19 (d, J = 1.1 Hz, IH), 8.05 (m, 2H), 7.92 (ddd, J= 9.3, 7.7, 1.6 Hz, IH), 7.60 (m, IH), 7.54 (m, 2H), 7.44 (m, IH); IR (KBr) 788, 984, 1124, 1166, 1306, 1424, 1446, 1575, 3085 cm“1; MS (TOF) mlz 220.0439 (220.0427 calcd for C11H10NO2S, MH); Anal, calcd for C11H9NO2S: C, 60.26; H, 4.14; N, 6.39; S, 14.62. Found: C, 60.40; H, 4.02; N, 6.40; S, 14.76.

As noted above, use of the improved process of the present invention results in an improved habit of the crystalline Form IV compound of Formula I. The improved habit reduces surface area of the crystal, improves the filtration, and washing, and improves the efficiency of azide mutagen rejection. These improvements are described in greater detail below.

In patent application WO2005/042515, the polish filtration is carried out in 7 volumes (L/kg) of isopropanol near its boiling point (65-83 0C), a process that is difficult and hazardous to execute in commercial manufacturing because of the high risk of crystallization on the filter and/or vessel transfer lines due to supersaturation. In the preferred crystallization solvent, isopropyl acetate, the polish filtration is conducted in four volumes of isopropyl acetate at temperatures from 45 to 55 0C. This temperature range is 35 to 45 0C lower than the boiling point of isopropyl acetate, which provides a key safety advantage.

PATENT

WO 2005042515

PATENT

WO 2017031215

EXAMPLES

Example 1: Preparation of Compound (I) via Negishi Coupling Route

Example 1 provides a scheme including preparations 1A-1D, described below, for the synthesis of the compound of Formula (I) and intermediates used in the route. An overview of the scheme is as follows:

80 on ma s ale

Example 1A: Preparation of Compound (I)

Zinc dust (200 mg, 3.06 mmol) combined with 2.0 mL of dimethylformamide was treated with 0.010 mL of 1,2-dibromoethane and heated to 65°C for 3 minutes. The mixture was cooled to ambient temperature and treated with 0.010 mL of trimethylsilyl chloride. After 5 minutes, 1.26 mL of 1M zinc chloride in diethyl ether was added to the mixture followed by Compound (Ila) (600 mg, 1.20 mmol). The mixture was heated to 65°C and further treated with 0.020 mL each of 1,2-dibromoethane and trimethylsilyl chloride. After 2.5 hours, via HPLC chromatogram, the reaction showed some formation of the zincate and was allowed to stir at ambient temperature for 16 hours. At this time

tetrakis(triphenylphosphine)palladium(0) (70 mg, 0.06 mmol), Compound (Ilia) (357 mg, 1.20 mmol) were added to the reaction and the mixture heated to 65°C. HPLC analysis showed the formation of Compound (I) in the reaction.

IB: Preparation of Comp

To a solution of Compound (IV) (8.00 g, 18 mmol) in 40 mL of 1,2-dichloroethane was added a solution of iodine monochloride (10.7 g, 65.9 mmol) in 40 mL of 1,2-dichloroethane resulting in a slurry. The slurry was heated to 75°C for 4 hours then cooled to ambient temperature. The solids were collected by filtration, washed with heptane, then combined with 90 mL of ethyl acetate and 80 mL of saturated sodium thiosulfate solution. The organic phase was washed with saturated sodium chloride solution and dried with sodium sulfate. The mixture was concentrated to yield 7.80 g (87%) of Compound (Ila) as a yellow solid. The product could be further purified by silica gel chromatography. Thus 2.0 g of yellow solid was dissolved in dichloromethane and charged onto a silica gel column. The product was eluted using tert-butyl methyl ether to provide 1.87 g (93% recovery) of Compound (Ila) as a white powder. Analytical data: Iodine monochloride complex: ¾ NMR (500 MHz, DMSO-de) δ 8.80 (2 H), 8.05 (1 H), 7.77 (2 H), 7.59 (2 H), 5.86 (2 H).

Uncomplexed: ¾ NMR (500 MHz, DMSO-de) δ 8.71 (2 H), 8.03 (1 H), 7.74 (2 H), 7.44 (2 H), 5.86 (2 H).

It was observed that the iodination proceeded smoothly as a suspension in 1,2-dichloroethane with IC1 (4.0 equiv) at 75°C. An ICl-Compound (Ila) complex was initially isolated by filtration. Compound (Ila) was then obtained in approximately 85% yield by treatment of the ICl-Compound (Ila) complex with sodium thiosulfate. This protocol provided a viable means of isolation of Compound (Ila) without the use of DMF.

Example 1C: Preparation of silyl substituted triazole (Compound IV)

A mixture of Compound (V) (8.07 g, 30.0 mmol) and Compound (VI) (5.12 g, 29.2 mmol) was heated to 100°C for 18 hours. To the mixture was added 40 mL of heptane and the reaction was allowed to cool with rapid stirring. After 1 hour the solids were collected by filtration and washed with heptane then dried to 9.30 g (72%) of Compound (IV) as a tan solid. Analytical data: ¾ NMR (500 MHz, DMSO-de) δ 8.66 (2 H), 8.04 (1 H), 7.67 (2 H), 7.32 (2 H), 5.72 (2 H), 0.08 (9 H).

It was further found that combining Compound (V) and Compound (VI) (neat) and heating at 95 – 105°C afforded a 92: 8 mixture of regioisomers as shown below:

Crystallization of the mixture from heptane afforded Compound (IV) in 62-72% yield, thus obviating the need for chromatography to isolate Compound (IV).

Example ID: Preparation of starting material Compound (VI)

Zinc bromide (502 g, 2.23 mole) was added in approximately 100 g portions to 2.0 L of tetrahydrofuran cooled to between 0 and 10°C. To this cooled solution was added 4-bromopyridine hydrochloride (200 g, 1.02 mol), triphenylphosphine (54 g, 0.206 mol), and palladium (II) chloride (9.00 g, 0.0508 mol). Triethylamine (813 g, 8.03 mol) was then added at a rate to maintain the reaction temperature at less than 10°C, and finally

trimethylsilylacetylene (202 g, 2.05 mol) was added. The mixture was heated to 60°C for 4.5 hours. The reaction was cooled to -5°C and combined with 2.0 L of hexanes and treated with 2 L of 7.4 M NH4OH. Some solids were formed and were removed as much as possible with the aqueous phase. The organic phase was again washed with 2.0 L of 7.4 M NH4OH, followed by 2 washes with 500 mL of water, neutralized with 1.7 L of 3 M hydrochloric acid, dried with sodium sulfate, and concentrate to a thick slurry. The slurry was combined with 1.0 L of hexanes to give a precipitate. The precipitate was removed by filtration and the filtrate was concentrated to 209 g of dark oil. The product was purified by distillation (0.2 torr, 68°C) to give 172 g (96%) of Compound (VI) as colorless oil. Analytical data: ¾ NMR (500 MHz, DMDO-de) δ 8.57 (2 H), 7.40 (2 H), 0.23 (9 H).

EXAMPLE 2 – Preparation of Compound (Ilia)

Example 2 provides a morpholine amide route for the synthesis of Compound (Ilia). In this approach, morpholine amide (Compound VII) was prepared from 2-chlorobenzoyl chloride (Preparation 2A). Metallation of 2-bromopyridine with LDA (1.09 equiv.) in THF at -70°C followed by addition of (Compound VII) afforded Compound (Ilia) in 37% yield after crystallization from IP A/heptane (Preparation 2B). This sequence provides a direct route to Compound (Ilia), and a means to isolate Compound (Ilia) without the use of

chromatography. Compound (Ilia) may then be used to form Compound (I) as shown in Example 1A above (Preparation 2C).

Preparation 2A: Preparation of Compound (VII)

Toluene (1.5 L) was added to Compound (IX) (150 g, 0.86 mol) and cooled to 10°C. Morpholine (82 mL, 0.94 mol) was added to the clear solution over 10 minutes. The resulting white slurry was stirred for 20 minutes then pyridine (92 mL, 1.2 mol) was added dropwise over 20 minutes. The cloudy white mixture was stirred in a cold bath for 1 hour. Water (600 mL) was added in a single portion and the cold bath removed. The mixture was stirred for 20 minutes and the layers are separated. The organic layer was washed with a mixture of 1 N HC1 and water (2: 1, 500 mL:250 mL). The pH of the aqueous layer was ~ 2. The organic layer was washed with a mixture of saturated NaHCCb and water (1 : 1, 100 mL: 100 mL). The pH of the aqueous layer was ~ 9. The layers were separated. The organic layer was concentrated in vacuo to an oil. The oil was dissolved in IPA (70 mL) and heated at 60°C for 30 min. The clear solution was allowed to cool to 30°C, then heptane (700 mL, 4.7 v) was added dropwise. The resulting slurry was stirred at RT for 2 hours then cooled to 0°C for 1 hour. The slurry was filtered at RT, washed with heptane then dried under vacuum at 30°C overnight. Compound (VII) (156.2 g, 81%) was obtained as a white solid. Analytical data: ¾ NMR (500 MHz, CDCh) δ 7.42-7.40 (m, 1 H), 7.35-7.29 (m, 3 H), 3.91-3.87 (m, 1 H), 3.80-3.76 (m, 3 H), 3.71 (ddd, J= 11.5, 6.8, 3.3 Hz, 1 H), 3.60 (ddd, J = 11.2, 6.4, 3.4 Hz, 1 H), 3.28 (ddd, J= 13.4, 6.3, 3.2 Hz, 1 H), 3.22 (ddd, J= 13.7, 6.8, 3.3 Hz, 1 H); LRMS (ES+) calcd for CnHi3F6ClN02 (M+H)+ 226.1, found 225.9 m/z.

Preparation 2B: Preparation of Compound (Ilia)

THF (75 mL) was added to diisopropyl amine (4.9 mL, 34.8 mmol) and cooled to a

temperature of -70°C under N2 atmosphere. 2.5 M w-BuLi in hexanes (13.9 mL, 34.8 mmol) was added in a single portion (a 30-40°C exotherm) to the clear solution and cooled back to -70°C. Compound (VIII) (5.0 g, 31.6 mmol) was added neat to the LDA solution (a 2 to 5°C exotherm) followed by a THF (10 mL) rinse, keeping T< -65°C. This clear yellow solution was stirred at -70°C for 15 min. Compound (VII) (7.1 g, 31.6 mmol) in THF (30 mL) was added keeping T< -65°C. The resulting clear orange solution was stirred at -70°C for 3 hours. MeOH (3 mL) was added to quench reaction mixture and the cold bath was removed. 5 N HC1 (25 mL) was added to the reaction solution. MTBE (25 mL) was added, and the layers were separated. The organic layer was washed with water (25 mL X 2). The organic layer was dried over MgS04 and filtered. The organic layer was concentrated in vacuo to an orange oil. The oil was dissolved in IPA (15 mL, 3 vol) at ambient temperature. Heptane (25 mL) was added dropwise and the resulting slurry was stirred at RT for 1 hour. The slurry was cooled to 0°C for 1 hour and filtered. The filter cake was rinsed with chilled heptane (20 mL) and dried under vacuum at 30°C overnight. Compound (Ilia) (4.25 g, 45%) was obtained as a yellow solid.

Several reactions were run at different temperatures and with different addition rates of Compound (VII). If the reaction temperature was maintained below -65°C and Compound (VII) was added in <5 min, it was found that the reaction worked well. If the temperature was increased and/or the addition time of Compound (VII) was increased, then yields suffered, and the work-up was complicated by emulsions.

Preparation 2C: Preparation of Compound (I)

Compound (Ilia) may then reacted with Compound (Ila) to produce Compound (I) as shown in Preparation 1A.

EXAMPLE 3

Example 3 describes a new route for the synthesis of an intermediate free base, which may be used to form Compound (I) as described further below.

Example 3A: Preparation of starting material (Compound X) from 2-Chloronicotinonitrile

A mixture of NaH (40.0 g, 1 mol, 60% dispersion in mineral oil) and 2-chloronicotinonitrile (69.3 g, 500 mmol) in THF (1 L) was heated to reflux. A solution of 4-acetylpyridine (60.6 g, 500 mmol) in THF (400 mL) was added over a period of 40 min. The resulting dark brown mixture was stirred at reflux for ~ 2 h. The heating mantle was then removed, and AcOH (58 mL, 1 mol) was added. EtOAc (1 L) and H2O (1 L) were then added, and the layers were separated. The organic layer was concentrated to afford an oily solid. CH3CN (500 mL) was added, and the mixture was stirred for 30 min. H2O (1 L) was then added. The mixture was stirred for 1 h then filtered. The solid was rinsed with 2: 1

CH3CN-H2O (900 mL) and hexanes (400 mL) then dried under vacuum at 45°C overnight to afford 61.4 g (55% yield) of Compound (X) as yellow solid. Compound (X) exists as an approximate 95:5 enol-ketone mixture in CDCI3. Analytical data for enol: IR (CHCI3): 3024, 2973, 2229, 1631, 1597, 1579, 1550, 1497; ¾ NMR (500 MHz, CDCI3) δ 8.69 (dd, J= 4.4,

1.7 Hz, 2H), 8.55 (dd, J = 5.2, 1.8 Hz, 1H), 7.97 (dd, J= 7.9, 1.8 Hz, 1H), 7.70 (dd, J= 4.6, 1.5 Hz, 2H, 7.17 (dd, J = 7.8, 5.0 Hz, 1H), 6.59 (s, 1H); LRMS (ES+) calcd for C13H10N3O (M+H)+ 224.1, found 224.0 m/z.

Preparation 3B: Preparation of Compound (XI)

Preparation 3B(1):

(X) (XI)

Compound (XI) may be prepared using Compound (X).

Preparation 3B(2):

Alternatively, the following procedure for the conversion of nitrile into an acid which may also yield compound (XI). A mixture of Compound (X) (1 eq) and NaOH (1.5 eq) in 1 : 1 fhO-EtOH (3.5 mL/g of Compound (X)) was heated at 65°C overnight. The reaction mixture was cooled to RT then added to CH2C12 (12.5 mL/g of Compound (X)) and H20 (12.5 mL/g of Compound (X)). Cone. HC1 (2.5 mL/g of Compound (X)) was then added, and the layers were separated. The aqueous layer was extracted with CH2CI2 (10 mL/g of Compound (X)). The combined organic extracts were washed with H2O (12.5 ml/g of Compound (X)), dried (MgS04), filtered and concentrated to afford Compound (XI).

Preparation 3C

Compound Compound (XI) may then be converted into a Stage C intermediate free base, with observed 87% conversion in Grignard reaction as shown above. A complete synthesis route for Com ound (I) starting from compound Compound (XI) is depicted below.

Detailed experimental procedures for the synthesis of benzoate salt and final step are given in

International Patent Application Publication WO 2008/079600 Al .

References

  1.  “Company Overview of Eli Lilly & Co., Worldwide License to Develop and Commercialize VLY-686”. Bloomberg Business. Retrieved 16 November 2015.
  2.  [1]
  3.  “Vanda Pharmaceuticals Announces Tradipitant Phase II Proof of Concept Study Results for Chronic Pruritus in Atopic Dermatitis”. PR Newswire. Retrieved 16 November 2015.
  4.  Schmidt, B (2006). “Proof of principle studies”. Epilepsy Res. 68 (1): 48–52. doi:10.1016/j.eplepsyres.2005.09.019. PMID 16377153.
  5.  George, DT; Gilman, J; Hersh, J; et al. (2008). “Neurokinin 1 receptor antagonism as a possible therapy for alcoholism.”. Science. 6: 1536–1539. doi:10.2147/SAR.S70350. PMC 4567173Freely accessible. PMID 26379454.
  6.  Tauscher, J; Kielbasa, W; Iyengar, S; et al. (2010). “Development of the 2nd generation neurokinin-1 receptor antagonist LY686017 for social anxiety disorder”. European Neuropsychopharmacology. 20 (2): 80–87. doi:10.1016/j.euroneuro.2009.10.005. PMID 20018493.

George, D.T.; Gilman, J.; Hersh, J.; Thorsell, A.; Herion, D.; Geyer, C.; Peng, X.; Kielbasa, W.; Rawlings, R.; Brandt, J.E.; Gehlert, D.R.; Tauscher, J.T.; Hunt, S.P.; Hommer, D.; Heilig, M. Neurokinin 1 receptor antagonism as a possible therapy for alcoholism, Science 2008, 319(5869): 1536

Gackenheimer, S.L.; Gehlert, D.R.In vitro and in vivo autoradiography of the NK-1 antagonist (3H)-LY686017 in guinea pig brain39th Annu Meet Soc Neurosci (October 17-21, Chicago) 2009, Abst 418.16

Tonnoscj, K.; Zopey, R.; Labus, J.S.; Naliboff, B.D.; Mayer, E.A.
The effect of chronic neurokinin-1 receptor antagonism on sympathetic nervous system activity in irritable bowel syndrome (IBS) Dig Dis Week (DDW) (May 30-June 4, Chicago) 2009, Abst T1261

Kopach, M.E.; Kobierski, M.E.; Coffey, D.S.; et al.  
Process development and pilot-plant synthesis of (2-chlorophenyl)[2-(phenylsulfonyl)pyridin-3-yl]methanone
Org Process Res Dev 2010, 14(5): 1229

1 to 7 of 7
Patent ID Patent Title Submitted Date Granted Date
US2016060250 NOVEL INTERMEDIATE AND PROCESS USEFUL IN THE PREPARATION OF -(2-CHLOROPHENYL)-METHANONE 2015-11-10 2016-03-03
US2015320866 PHARMACEUTICAL COMPOSITION COMPRISING ANTIEMETIC COMPOUNDS AND POLYORTHOESTER 2013-12-13 2015-11-12
US2014206877 NOVEL INTERMEDIATE AND PROCESS USEFUL IN THE PREPARATION OF -(2-CHLOROPHENYL)-METHANONE 2014-03-27 2014-07-24
US2012225904 New 7-Phenyl-[1, 2, 4]triazolo[4, 3-a]Pyridin-3(2H)-One Derivatives 2010-11-09 2012-09-06
US2010056795 NOVEL INTERMEDIATE AND PROCESS USEFUL IN THE PREPARATION OF -(2-CHLOROPHENYL)-METHANONE 2010-03-04
US7381826 Crystalline forms of {2-[1-(3, 5-bis-trifluoromethyl-benzyl)-5-pyridin-4-yl-1H-[1, 2, 3]triazol-4-yl]-pyridin-3-yl}-(2-chlorophenyl)-methanone 2007-04-05 2008-06-03
US7320994 Triazole derivatives as tachykinin receptor antagonists 2005-10-27 2008-01-22
Tradipitant
LY686017.svg
Legal status
Legal status
  • Investigational
Identifiers
CAS Number
PubChem CID
ChemSpider
Chemical and physical data
Formula C28H16ClF6N5O
Molar mass 587.90 g/mol
3D model (Jmol)

TRADIPITANT

Overview

Tradipitant

Tradipitant is being evaluated in a Phase II study in treatment resistant pruritus in atopic dermatitis.

Tradipitant is an NK-1 receptor antagonist licensed from Eli Lilly in 2012. Tradipitant has demonstrated proof-of-concept in alcohol dependence in a study published by the NIH1. In that study tradipitant was shown to reduce alcohol cravings and voluntary alcohol consumption among patients with alcohol dependence. NK-1R antagonists have been evaluated in a number of indications including chemotherapy-induced nausea and vomiting (CINV), post-operative nausea and vomiting (PONV), alcohol dependence, anxiety, depression, and pruritus.

The NK-1R is expressed throughout different tissues of the body, with major activity found in neuronal tissue. Substance P (SP) and NK-1R interactions in neuronal tissue regulate neurogenic inflammation locally and the pain perception pathway through the central nervous system. Other tissues, including endothelial cells and immune cells, have also exhibited SP and NK-1R activity2. The activation of NK-1R by the natural ligand SP is involved in numerous physiological processes, including the perception of pain, behavioral stressors, cravings, and the processes of nausea and vomiting1,2,3. An inappropriate over-expression of SP either in nervous tissue or peripherally could result in pathological conditions such as substance dependence, anxiety, nausea/vomiting, and pruritus1,2,3,4. An NK-1R antagonist may possess the ability to reduce this over-stimulation of the NK-1R, and as a result address the underlying pathophysiology of the symptoms in these conditions.

References

  1. George DT, Gilman J, Hersh J, Thorsell A, Herion D, Geyer C, Peng X, Keilbasa W, Rawlings R, Brandt JE, Gehlert DR, Tauscher JT, Hunt SP, Hommer D, Heilig M. Neurokinin 1 receptor antagonism as a possible therapy for alcoholism. Science. 2008; 319(5869):1536-9
  2. Almeida TA, Rojo J, Nieto PM, Pinto FM, Hernandez M, et al. Tachykinins and tachykinin receptors: structure and activity relationships. Current Medicinal Chemistry. 2004;11:2045-2081.
  3. Hargreaves R, Ferreira JC, Hughes D, Brands J, Hale J, Mattson B, Mill S. Development of aprepitant, the first neurokinin-1 receptor antagonist for the prevention of chemotherapy-induced nausea and vomiting. Annals of the New York Academy of Sciences. 2011; 1222:40-48.
  4. Stander S, Weisshaar E, Luger A. Neurophysiological and neurochemical basis of modern pruritus treatment. Experimental Dermatology. 2007;17:161-69.

///////////////////tradipitant, PHASE 2, VLY-686,  LY686017, традипитант , تراديبيتانت , 曲地匹坦 , VANDA, ELI LILLY, Gastroparesis Pruritus


Filed under: Phase2 drugs, Uncategorized Tagged: eli lilly, Gastroparesis Pruritus, LY686017, традипитант, phase 2, tradipitant, VANDA, VLY-686, 曲地匹坦, تراديبيتانت

FDA approves Xermelo (telotristat ethyl) for carcinoid syndrome diarrhea

$
0
0

ChemSpider 2D Image | Telotristat ethyl | C27H26ClF3N6O3Image result for telotristat ethyl

 

Telotristat ethyl

Molecular Formula, C27-H26-Cl-F3-N6-O3,

Molecular Weight, 574.9884,

RN: 1033805-22-9
UNII: 8G388563M

LX 1032

(2S)-2-Amino-3-[4-[2-amino-6-[[(1R)-1-[4-chloro-2-(3-methylpyrazol-1-yl)phenyl]-2,2,2-trifluoroethyl]oxy]pyrimidin-4-yl]phenyl]propionic acid ethyl ester

Ethyl-4-(2-amino-6-{(1R)-1-[4-chlor-2-(3-methyl-1H-pyrazol-1-yl)phenyl]-2,2,2-trifluorethoxy}-4-pyrimidinyl)-L-phenylalaninat

L-Phenylalanine, 4-[2-amino-6-[(1R)-1-[4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl]-2,2,2-trifluoroethoxy]-4-pyrimidinyl]-, ethyl ester
SEE……………
Image result for Telotristat etiprate,LX1606 Hippurate.png
Telotristat etiprate,
(S)-ethyl 2-amino-3-(4-(2-amino-6-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate 2-benzamidoacetate .
CAS: 1137608-69-5 (etiprate), LX 1606
Chemical Formula: C36H35ClF3N7O6
Molecular Weight: 754.16
L-Phenylalanine, 4-[2-amino-6-[(1R)-1-[4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl]-2,2,2-trifluoroethoxy]-4-pyrimidinyl]-, ethyl ester, compd. with N-benzoylglycine (1:1)
  • LX 1032 hippurate
  • LX 1606
SEE ALSO………….
Telotristat, also known as LX1033, 1033805-28-5 CAS OF ACID FORM
 Arokiasamy Devasagayaraj
02/28/2017
The U.S. Food and Drug Administration today approved Xermelo (telotristat ethyl) tablets in combination with somatostatin analog (SSA) therapy for the treatment of adults with carcinoid syndrome diarrhea that SSA therapy alone has inadequately controlled.
February 28, 2017
The U.S. Food and Drug Administration today approved Xermelo (telotristat ethyl) tablets in combination with somatostatin analog (SSA) therapy for the treatment of adults with carcinoid syndrome diarrhea that SSA therapy alone has inadequately controlled.

Carcinoid syndrome is a cluster of symptoms sometimes seen in people with carcinoid tumors. These tumors are rare, and often slow-growing. Most carcinoid tumors are found in the gastrointestinal tract. Carcinoid syndrome occurs in less than 10 percent of patients with carcinoid tumors, usually after the tumor has spread to the liver. The tumors in these patients release excess amounts of the hormone serotonin, resulting in diarrhea. Complications of uncontrolled diarrhea include weight loss, malnutrition, dehydration, and electrolyte imbalance.

“Today’s approval will provide patients whose carcinoid syndrome diarrhea is not adequately controlled with another treatment option,” said Julie Beitz, M.D., director of the Office of Drug Evaluation III in the FDA’s Center for Drug Evaluation and Research.

Xermelo, in a regimen with SSA therapy, is approved in tablet form to be taken orally three times daily with food. Xermelo inhibits the production of serotonin by carcinoid tumors and reduces the frequency of carcinoid syndrome diarrhea.

The safety and efficacy of Xermelo were established in a 12-week, double-blind, placebo-controlled trial in 90 adult participants with well-differentiated metastatic neuroendocrine tumors and carcinoid syndrome diarrhea. These patients were having between four to 12 daily bowel movements despite the use of SSA at a stable dose for at least three months. Participants remained on their SSA treatment, and were randomized to add placebo or treatment with Xermelo three times daily. Those receiving Xermelo added on to their SSA treatment experienced a greater reduction in average bowel movement frequency than those on SSA and placebo. Specifically, 33 percent of participants randomized to add Xermelo on to SSA experienced an average reduction of two bowel movements per day compared to 4 percent of patients randomized to add placebo on to SSA.

The most common side effects of Xermelo include nausea, headache, increased levels of the liver enzyme gamma-glutamyl transferase, depression, accumulation of fluid causing swelling (peripheral edema), flatulence, decreased appetite and fever. Xermelo may cause constipation, and the risk of developing constipation may be increased in patients whose bowel movement frequency is less than four bowel movements per day. Patients treated with a higher than recommended dosage of Xermelo developed severe constipation in clinical trials. One patient required hospitalization and two other patients developed complications of either intestinal perforation or intestinal obstruction. Patients should be monitored for severe constipation. If a patient experiences severe constipation or severe, persistent or worsening abdominal pain, they should discontinue Xermelo and contact their healthcare provider.

The FDA granted this application fast track designation and priority review. The drug also received orphan drug designation, which provides incentives to assist and encourage the development of drugs for rare diseases.

Xermelo is manufactured by Woodlands, Texas-based Lexicon Pharmaceuticals, Inc.

SYNTHESIS…….WO 2011100285

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2011100285&recNum=142&docAn=US2011024141&queryString=((serotonin)%2520OR%2520(HT2C)%2520OR%2520(&

5.67. Synthesis of (S)-2-Amino-3-[4-(2-amino-6-{R-l-[4-chloro-2-(3-methyl-pyrazol-l-yll- phenyll-2,2,2-trifluoro-ethoxy)-pyrimidin-4-yl)-phenyll-propionic acid ethyl ester

The title compound was prepared stepwise, as described below:

Step 1: Synthesis of l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanone. To a 500 ml 2 necked RB flask containing anhydrous methanol (300 ml) was added thionyl chloride (29.2 ml, 400 mmol) dropwise at 0-5°C (ice water bath) over 10 minutes. The ice water bath was removed, and 2-bromo-4-chloro-benzoic acid (25 g, 106 mmol) was added. The mixture was heated to mild reflux for 12h. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was concentrated. Crude product was dissolved in dichloromethane (DCM, 250 ml), washed with water (50 ml), sat. aq. NaHC03 (50 ml), brine (50 ml), dried over sodium sulfate, and concentrated to give the 2- bromo-4-chloro-benzoic acid methyl ester (26 g, 99 %), which was directly used in the following step.

2-Bromo-4-chloro-benzoic acid methyl ester (12.4 g, 50 mmol) in toluene (200 ml) was cooled to -70°C, and trifluoromethyl trimethyl silane (13 ml, 70 mmol) was added.

Tetrabutylamonium fluoride (1M, 2.5 ml) was added dropwise, and the mixture was allowed to warm to room temperature over 4h, after which it was stirred for 10 hours at room temperature. The reaction mixture was concentrated to give the crude [l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-l-methoxy-ethoxy]-trimethyl-silane. The crude intermediate was dissolved in methanol (100 ml) and 6N HCI (100 ml) was added. The mixture was kept at 45-50°C for 12h. Methanol was removed, and the crude was extracted with dichloromethane (200 ml). The combined DCM layer was washed with water (50 ml), NaHC03 (50 ml), brine (50 ml), and dried over sodium sulfate. Removal of solvent gave a crude product, which was purified by ISCO column chromatography, using 1-2% ethyl acetate in hexane as solvent, to afford l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanone (10 g, 70%). !H-NMR (300 MHz, CDC ): δ (ppm) 7.50 (d,lH), 7.65(d,lH), 7.80(s,lH).

Step 2: Synthesis of R-l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanol. To catechol borane (1M in THF 280 ml, 280 mmol) in a 2L 3-necked RB flask was added S-2-methyl-CBS oxazaborolidine (7.76 g, 28 mmol) under nitrogen, and the resulting mixture was stirred at room temperature for 20 min. The reaction mixture was cooled to -78°C (dry ice/acetone bath), and 1-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanone (40 g, 139 mmol) in THF (400 ml) was added dropwise over 2 hours. The reaction mixture was allowed to warm to -36°C, and was stirred at that temperature for 24 hours, and further stirred at -32 °C for another 24h. 3N NaOH (250 ml) was added, and the cooling bath was replaced by ice-water bath. Then 30 % hydrogen peroxide in water (250 ml) was added dropwise over 30 minutes. The ice water bath was removed, and the mixture was stirred at room temperature for 4 hours. The organic layer was separated, concentrated and re-dissolved in ether (200 ml). The aqueous layer was extracted with ether (2 x 200 ml). The combined organic layers were washed with IN aq. NaOH (4 x 100 ml), brine, and dried over sodium sulfate. Removal of solvent gave crude product which was purified by column chromatography using 2 to 5% ethyl acetate in hexane as solvent to give desired alcohol 36.2 g (90 %, e.e. >95%). The alcohol (36.2 g) was crystallized from hexane (80 ml) to obtain R-l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanol 28.2 g (70 %; 99-100 % e.e.). !H-NMR (400 MHz, CDCIs) δ (ppm) 5.48 (m, 1H), 7.40 (d, 1H), 7.61 (d, 2H).

Step 3: Synthesis of R-l-[4-chloro-2-(3-methyl-pyrazol-l-yl)-phenyll-2.2.2-trifluoro-ethanol. R-l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanol (15.65 g, 54.06 mmol), 3-methylpyrazole (5.33 g, 65 mmol), Cul (2.06 g, 10.8 mmol), 2CO3 (15.7 g, 113.5 mmol), (lR,2R)-N,N’-dimethyl-cyclohexane-l,2-diamine (1.54 g, 10.8 mmol) and toluene (80 ml) were combined in a 250 ml pressure tube and heated to 130°C (oil bath temperature) for 12 hours. The reaction mixture was diluted with ethyl acetate and washed with H2O (4 x 100 ml), brine, and dried over sodium sulfate. Removal of solvent gave a crude product, which was purified by ISCO column chromatography using 5-10 % ethyl acetate in hexane as solvent to get R-l-[4-chloro-2-(3-methyl-pyrazol-l-yl)-phenyl]-2,2,2-trifluoro-ethanol (13.5 g; 86 %). i-H-NMR (400 MHz, CDC ): δ (ppm) 2.30(s, 3H), 4.90(m, 1H), 6.20(s, 1H), 6.84(d, 1H), 7.20(s, 1H), 7.30(d, 1H), 7.50(d, 1H).

Step 4: Synthesis of (S)-2-Amino-3- 4-(2-amino-6-fR-l-[4-chloro-2-(3-methyl-pyrazol-l-yl)-phenyll^^^-trifluoro-ethoxyl-pyrimidin^-yll-phenvD-propionic acid ethyl ester. R-l-[4-chloro-2-(3-methyl-pyrazol-l-yl)-phenyl]-2,2,2-trifluoro-ethanol (17.78 g, 61.17 mmol), (S)-3-[4-(2-amino-6-chloro-pyrimidine-4-yl)-phenyl]-2-tert-butoxycarbonylamino-propionic acid (20.03 g, 51 mmol), 1,4-dioxane (250 ml), and CS2CO3 (79.5 g, 244 mmol) were combined in a 3-necked 500 ml RB flask and heated to 100°C (oil bath temperature) for 12-24 hours. The progress of reaction was monitored by LCMS. After the completion of the reaction, the mixture was cooled to 60°C, and water (250 ml) and THF (400 ml) were added. The organic layer was separated and washed with brine (150 ml). The solvent was removed to give crude BOC protected product, which was taken in THF (400 ml), 3N HCI (200 ml). The mixture was heated at 35-40 °C for 12 hours. THF was removed in vacuo. The remaining aqueous layer was extracted with isopropyl acetate (2x 100 ml) and concentrated separately to recover the unreacted alcohol (3.5 g). Traces of remaining organic solvent were removed from the aqueous fraction under vacuum.

To a 1L beaker equipped with a temperature controller and pH meter, was added H3PO4 (40 ml, 85 % in water) and water (300 ml) then 50 % NaOH in water to adjust pH to 6.15. The temperature was raised to 58 °C and the above acidic aqueous solution was added dropwise into the buffer with simultaneous addition of 50 % NaOH solution in water so that the pH was maintained between 6.1 to 6.3. Upon completion of addition, precipitated solid was filtered and washed with hot water (50-60°C) (2 x 200 ml) and dried to give crude (S)-2-amino-3-[4-(2-amino-6-[R-l-[4-chloro-2-(3-methyl-pyrazol-l-yl)-phenyl]-2,2,2-trifluoro-ethoxy}-pyrimidin-4-yl)-phenyl}^ propionic acid (26.8 g; 95 %). LCMS and HPLC analysis indicated the compound purity was about 96-97 %.

To anhydrous ethanol (400 ml) was added SOC (22 ml, 306 mmol) dropwise at 0-5°C.

Crude acid (26.8 ) from the above reaction was added. The ice water bath was removed, and the reaction mixture was heated at 40-45°C for 6-12 hours. After the reaction was completed, ethanol was removed in vacuo. To the residue was added ice water (300 ml), and extracted with isopropyl acetate (2 x 100 ml). The aqueous solution was neutralized with saturated Na2C03 to adjust the pH to 6.5. The solution was extracted with ethyl acetate (2 x 300 ml). The combined ethyl acetate layer was washed with brine and concentrated to give 24 g of crude ester (HPLC purity of 96-97 %). The crude ester was then purified by ISCO column chromatography using 5 % ethanol in DCM as solvent to give (S)-2-amino-3-[4-(2-amino-6-{R-l-[4-chloro-2-(3-methyl-pyrazol-l-yl)-phenyl]-2,2,2-trifluoro-ethoxy}-pyrimidin-4-yl)-phenyl}-propionic acid ethyl ester (20.5g; 70 %; HPLC purity of 98 %). LCMS M+l = 575. !H-NMR (400 MHz, CDsOD): δ (ppm) 1.10 (t, 3H), 2.25 (s, 3H), 2.85 (m, 2H), 3.65 (m, IH), 4.00 (q, 2H), 6.35 (s, IH), 6.60 (s, IH), 6.90 (m, IH), 7.18 (d, 2H), 7.45 (m, 2H), 7.70 (d, IH), 7.85 (m, 3H).

SYNTHESIS OF INTERMEDIATE

WO 2009048864

https://google.com/patents/WO2009048864A1?cl=en

6.15. Preparation of 6SV3-(4-(2-Amino-6-chloropyrimidin-4-yl)phenyl)-2- (fert-butoxycarbonylamino)propanoic Acid Using the Lithium Salt of (S)-2-(te^-butoxycarbonylamino)-3-(4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)phenyl)propanoic Acid

Figure imgf000021_0001

During preparation of compound 7, the isolation of the free acid can be optionally omitted. Thus, an aqueous solution of the lithium salt of compound 7 in 100 ml water, prepared from 5.0 g of Boc-Tyr-OMe (4, 17 mmol), was mixed 2-amino-4,6- dichloropyrimidine (3.3 g, 1.2 eq), potassium bicarbonate (5.0 g, 3 eq), bis(triphenylphosphine)palladium(II) dichloride (60 mg, 0.5 mol%), and 100 ml ethanol. The resulting mixture was heated at 700C for 5 hours. Additional 2-amino-4,6- dichloropyrimidine (1.1 g, 0.4 eq) was added and heating was continued at 7O0C for an additional 2 hours. HPLC analysis showed about 94% conversion. Upon cooling and filtration, the filtrate was analyzed by HPLC against a standard solution of compound 8. The assay indicated 3.9 g compound 8 was contained in the solution (59% yield from compound 4).

6.16. Alternative Procedure for Preparation of (S)-3-(4-f2-Amino-6- chloropyrimidin-4-yl)phenyl)-2-(fe^-butoxycarbonylamino)propanoic Acid Using Potassium Carbonate as Base

Figure imgf000021_0002

The boronic acid compound 11 (Ryscor Science, Inc., North Carolina, 1.0 g, 4.8 mmol) and potassium carbonate (1.32 g, 2 eq) were mixed in aqueous ethanol (15 ml ethanol and 8 ml water). Di-ter£-butyldicarbonate (1.25 g, 1.2 eq) was added in one portion. After 30 minutes agitation at room temperature, HPLC analysis showed complete consumption of the starting compound 11. The 2-amino-4,6- dichloropyrimidine (1.18 g, 1.5 eq) and the catalyst bis(triphenylphosphine)palladium(II) dichloride (34 mg, 1 mol%) were added and the resulting mixture was heated at 65-700C for 3 hours. HPLC analysis showed complete consumption of compound 12. After concentration and filtration, HPLC analysis of the resulting aqueous solution against a standard solution of compound 8 showed 1.26 g compound 8 (67% yield).

6.17. Alternative procedure for preparation of (5)-3-(4-(2-Amino-6-

Figure imgf000022_0001

The boronic acid compound 11 (10 g, 48 mmol) and potassium bicarbonate (14.4 g, 3 eq) were mixed in aqueous ethanol (250 ml ethanol and 50 ml water). Oi-tert- butyldicarbonate (12.5 g, 1.2 eq) was added in one portion. HPLC analysis indicated that the reaction was not complete after overnight stirring at room temperature. Potassium carbonate (6.6 g, 1.0 eq) and additional di-te/t-butyldicarbonate (3.1 g, 0.3 eq) were added. After 2.5 hours agitation at room temperature, HPLC analysis showed complete consumption of the starting compound 11. The 2-amino-4,6-dichloropyrimidine (11.8 g, 1.5 eq) and the catalyst bis(triphenylphosphine)-palladium(II) dichloride (0.34 g, 1 mol%” were added and the resulting mixture was heated at 75-8O0C for 2 hours. HPLC analysis showed complete consumption of compound 12. The mixture was concentrated under reduced pressure and filtered. The filtrate was washed with ethyl acetate (200 ml) and diluted with 3 : 1 THF/MTBE (120 ml). This mixture was acidified to pH about 2.4 by 6 N hydrochloric acid. The organic layer was washed with brine and concentrated under reduced pressure. The residue was precipitated in isopropanol, filtered, and dried at 500C under vacuum to give compound 8 as an off-white solid (9.0 g, 48% yield). Purity: 92.9% by HPLC analysis. Concentration of the mother liquor yielded and additional 2.2 g off-white powder (12% yield). Purity: 93.6% by HPLC analysis

PATENT

https://www.google.com/patents/WO2013059146A1?cl=en

This invention is directed to solid pharmaceutical dosage forms in which an active pharmaceutical ingredient (API) is (S)-ethyl 2-amino-3-(4-(2-amino-6-((R)-l-(4-chloro-2-(3- methyl-lH-pyrazol-l-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate

(telotristat):

Figure imgf000004_0001

or a pharmaceutically acceptable salt thereof. The compound, its salts and crystalline forms can be obtained by methods known in the art. See, e.g., U.S. patent no. 7,709,493.

PATENT

http://www.google.co.in/patents/WO2008073933A2?cl=en

6.19. Synthesis of (S)-2-Amino-3-r4-q-amino-6-{R-l-r4-chloro-2-(3-methyl- Pyrazol-l-yl)-phenyll-2,2,2-trifluoro-ethoxy}-pyrimidin-4-yl)-phenyll- propionic acid ethyl ester

Figure imgf000042_0001

The title compound was prepared stepwise, as described below: Step 1 : Synthesis of l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanone. To a 500 ml 2 necked RB flask containing anhydrous methanol (300 ml) was added thionyl chloride (29.2 ml, 400 mmol) dropwise at 0-50C (ice water bath) over 10 min. The ice water bath was removed, and 2-bromo-4-chloro-benzoic acid (25 g, 106 mmol) was added. The mixture was heated to mild reflux for 12h. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was concentrated. Crude product was dissolved in dichloromethane (DCM, 250 ml), washed with water (50 ml), sat. aq. NaHCO3 (50 ml), brine (50 ml), dried over sodium sulfate, and concentrated to give the 2- bromo-4-chloro-benzoic acid methyl ester (26 g, 99 %), which was directly used in the following step.

2-Bromo-4-chloro-benzoic acid methyl ester (12.4 g, 50 mmol) in toluene (200 ml) was cooled to -700C, and trifluoromethyl trimethyl silane (13 ml, 70 mmol) was added. Tetrabutylamonium fluoride (IM, 2.5 ml) was added dropwise, and the mixture was allowed to warm to room temperature over 4h, after which it was stirred for 1Oh at room temperature. The reaction mixture was concentrated to give the crude [l-(2-bromo-4-chloro-phenyl)-2,2,2- trifluoro-l-methoxy-ethoxy]-trimethyl-silane. The crude intermediate was dissolved in methanol (100 ml) and 6N HCl (100 ml) was added. The mixture was kept at 45-500C for 12h. Methanol was removed, and the crude was extracted with dichloromethane (200 ml). The combined DCM layer was washed with water (50 ml), NaHCO3 (50 ml), brine (50 ml), and dried over sodium sulfate. Removal of solvent gave a crude product, which was purified by ISCO column chromatography, using 1-2% ethyl acetate in hexane as solvent, to afford 1- (2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanone (10 g, 70%). 1H-NMR (300 MHz, CDCl3): δ (ppm) 7.50 (d,lH), 7.65(d,lH), 7.80(s,lH).

Step 2: Synthesis of R-l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanol. To catechol borane (IM in THF 280 ml, 280 mmol) in a 2L 3-necked RB flask was added S-2- methyl-CBS oxazaborolidine (7.76 g, 28 mmol) under nitrogen, and the resulting mixture was stirred at room temperature for 20 min. The reaction mixture was cooled to -78°C (dry ice/acetone bath), and l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanone (40 g, 139 mmol) in THF (400 ml) was added dropwise over 2h. The reaction mixture was allowed to warm to -36°C, and was stirred at that temperature for 24 h, and further stirred at -32°C for another 24h. 3N NaOH (250 ml) was added, and the cooling bath was replaced by ice-water bath. Then 30 % hydrogen peroxide in water (250 ml) was added dropwise over 30 minutes. The ice water bath was removed, and the mixture was stirred at room temperature for 4h. The organic layer was separated, concentrated and re-dissolved in ether (200 ml). The aqueous layer was extracted with ether (2 x 200 ml). The combined organic layers were washed with IN aq. NaOH (4 x 100 ml), brine, and dried over sodium sulfate. Removal of solvent gave crude product which was purified by column chromatography using 2 to 5% ethyl acetate in hexane as solvent to give desired alcohol 36.2 g (90 %, e.e. >95%). The alcohol (36.2 g) was crystallized from hexane (80 ml) to obtain R-l-(2-bromo-4-chloro- phenyl)-2,2,2-trifiuoro-ethanol 28.2 g (70 %; 99-100 % e.e.). 1H-NMR (400 MHz, CDCl3) δ (ppm) 5.48 (m, IH), 7.40 (d, IH), 7.61 (d, 2H). Step 3: Synthesis of R-l-r4-chloro-2-(3-methyl-pyrazol-l-vπ-phenyl1-2.2.2-trifluoro- ethanol. R-l-(2-bromo-4-chloro-phenyl)-2,2,2-trifluoro-ethanol (15.65g, 54.06 mmol), 3- methylpyrazole (5.33 g, 65 mmol), CuI (2.06 g, 10.8 mmol), K2CO3 (15.7 g, 113.5 mmol), (lR,2R)-N,N’-dimethyl-cyclohexane-l,2-diamine (1.54 g, 10.8 mmol) and toluene (80 ml) were combined in a 250 ml pressure tube and heated to 1300C (oil bath temperature) for 12 h. The reaction mixture was diluted with ethyl acetate and washed with H2O (4 x 100 ml), brine, and dried over sodium sulfate. Removal of solvent gave a crude product, which was purified by ISCO column chromatography using 5-10 % ethyl acetate in hexane as solvent to get R-I- [4-chloro-2-(3-methyl-pyrazol-l-yl)-phenyl]-2,2,2-trifluoro-ethanol (13.5 g; 86 %). 1H-NMR (400 MHz, CDCl3): δ (ppm) 2.30(s, 3H), 4.90(m, IH), 6.20(s, IH), 6.84(d, IH), 7.20(s, IH), 7.30(d, IH), 7.50(d, IH).

Step 4: Synthesis of (S)-2-Amino-3- r4-(2-amino-6- (R-I- r4-chloro-2-(3-methyl- pyrazol- 1 -ylVphenyl~|-2,2.,2-trifluoro-ethoxy| -pyrimidin-4-yl)-phenyU -propionic acid ethyl ester. R-l-[4-chloro-2-(3-methyl-pyrazol-l-yl)-phenyl]-2,2,2-trifluoro-ethanol (17.78 g, 61.17 mmol), (S)-3-[4-(2-amino-6-chloro-pyrimidine-4-yl)-phenyl]-2-tert- butoxycarbonylamino-propionic acid (20.03 g, 51 mmol), 1,4-dioxane (250 ml), and Cs2CO3 (79.5 g, 244 mmol) were combined in a 3-necked 500 ml RB flask and heated to 1000C (oil bath temperature) for 12-24 h. The progress of reaction was monitored by LCMS. After the completion of the reaction, the mixture was cooled to 600C, and water (250 ml) and THF (400 ml) were added. The organic layer was separated and washed with brine (150 ml). The solvent was removed to give crude BOC protected product, which was taken in THF (400 ml), 3N HCl (200 ml). The mixture was heated at 35-400C for 12h. THF was removed in vacuo. The remaining aqueous layer was extracted with isopropyl acetate (2x 100 ml) and concentrated separately to recover the unreacted alcohol (3.5 g). Traces of remaining organic solvent were removed from the aqueous fraction under vacuum.

To a IL beaker equipped with a temperature controller and pH meter, was added H3PO4 (40 ml, 85 % in water) and water (300 ml) then 50 % NaOH in water to adjust pH to 6.15. The temperature was raised to 58°C and the above acidic aqueous solution was added dropwise into the buffer with simultaneous addition of 50 % NaOH solution in water so that the pH was maintained between 6.1 to 6.3. Upon completion of addition, precipitated solid was filtered and washed with hot water (50-600C) (2 x 200 ml) and dried to give crude (S)-2- amino-3-[4-(2-amino-6-{R-l-[4-chloro-2-(3-methyl-pyrazol-l-yl)-phenyl]-2,2,2-trifluoro- ethoxy}-pyrimidin-4-yl)-phenyl} -propionic acid (26.8 g; 95 %). LCMS and HPLC analysis indicated the compound purity was about 96-97 %. To anhydrous ethanol (400 ml) was added SOCl2 (22 ml, 306 mmol) dropwise at 0-

5°C. Crude acid (26.8 g ) from the above reaction was added. The ice water bath was removed, and the reaction mixture was heated at 40-450C for 6-12h. After the reaction was completed, ethanol was removed in vacuo. To the residue was added ice water (300 ml), and extracted with isopropyl acetate (2 x 100 ml). The aqueous solution was neutralized with saturated Na2CO3 to adjust the pH to 6.5. The solution was extracted with ethyl acetate (2 x 300 ml). The combined ethyl acetate layer was washed with brine and concentrated to give 24 g of crude ester (HPLC purity of 96-97 %). The crude ester was then purified by ISCO column chromatography using 5 % ethanol in DCM as solvent to give (S)-2-amino-3-[4-(2- amino-6- (R- 1 -[4-chloro-2-(3-methyl-pyrazol- 1 -yl)-phenyl]-2,2,2-trifluoro-ethoxy} – pyrimidin-4-yl)-phenyl} -propionic acid ethyl ester (20.5g; 70 %; HPLC purity of 98 %). LCMS M+l = 575. 1H-NMR (400 MHz, CD3OD): δ (ppm) 1.10 (t, 3H), 2.25 (s, 3H), 2.85 (m, 2H), 3.65 (m, IH), 4.00 (q, 2H), 6.35 (s, IH), 6.60 (s, IH), 6.90 (m, IH), 7.18 (d, 2H), 7.45 (m, 2H), 7.70 (d, IH), 7.85 (m, 3H).

PATENT

WO 2011056916

https://www.google.com/patents/WO2011056916A1?cl=en

PATENT

WO 2010065333

https://www.google.com/patents/WO2010065333A1?cl=en

CLIP,……..PL CHECK ERROR

CONFUSION ON CODES, CLEAR PIC BELOW……LINK
Description of Telotristat Etiprate
Telotristat etiprate is the hippurate salt of telotristat ethyl.
Telotristat ethyl, also known as LX1032, has the chemical name, CAS identifier, and chemical structure shown below:
Chemical name: (S)-ethyl 2-amino-3-(4-(2-amino-6-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate
CAS Registry number: 1033805-22-9
Chemical structure:
Telotristat etiprate, also known as LX1606, is the hippurate salt of telotristat ethyl, and has the chemical name, CAS identifier, and chemical structure shown below:
Chemical Name: (S)-ethyl 2-amino-3-(4-(2-amino-6-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate 2-benzamidoacetate
CAS Registry number: 1137608-69-5
Chemical Structure:
Description of LX1033
Telotristat, also known as LX1033, has the chemical name, CAS identifier and chemical structure shown below:
Chemical Name: (S)-2-amino-3-(4-(2-amino-6-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoic acid
CAS Registry number: 1033805-28-5
Chemical Structure:

REFERENCES

Kulke, M.H.; Hoersch, D.; Caplin, M.E.; et al.
Telotristat ethyl, a tryptophan hydroxylase inhibitor for the treatment of carcinoid syndrome
J Clin Oncol 2017, 35(1): 14

WO2010056992A1 * Nov 13, 2009 May 20, 2010 The Trustees Of Columbia University In The City Of New York Methods of preventing and treating low bone mass diseases
US7709493 May 20, 2009 May 4, 2010 Lexicon Pharmaceuticals, Inc. 4-phenyl-6-(2,2,2-trifluoro-1-phenylethoxy)pyrimidine-based compounds and methods of their use
US20090088447 * Sep 25, 2008 Apr 2, 2009 Bednarz Mark S Solid forms of (s)-ethyl 2-amino-3-(4-(2-amino-6-((r)-1-(4-chloro-2-(3-methyl-1h-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)-pyrimidin-4-yl)phenyl)propanoate and methods of their use
Citing Patent Filing date Publication date Applicant Title
US9199994 Sep 5, 2014 Dec 1, 2015 Karos Pharmaceuticals, Inc. Spirocyclic compounds as tryptophan hydroxylase inhibitors
US9512122 Sep 1, 2015 Dec 6, 2016 Karos Pharmaceuticals, Inc. Spirocyclic compounds as tryptophan hydroxylase inhibitors

///////////telotristat ethyl, fast track designation,priority review,orphan drug designation, Xermelo ,  Woodlands, Texas-based,  Lexicon Pharmaceuticals, Inc, fda 2017, LX 1606, LX 1032

O=C(OCC)[C@@H](N)Cc1ccc(cc1)c2cc(nc(N)n2)O[C@H](c3ccc(Cl)cc3n4ccc(C)n4)C(F)(F)F

O=C(OCC)[C@@H](N)CC1=CC=C(C2=NC(N)=NC(O[C@H](C3=CC=C(Cl)C=C3N4N=C(C)C=C4)C(F)(F)F)=C2)C=C1.O=C(O)CNC(C5=CC=CC=C5)=O


Filed under: 0rphan drug status, FAST TRACK FDA, FDA 2017, Priority review, Uncategorized Tagged: Fast Track Designation, FDA 2017, Inc., Lexicon Pharmaceuticals, LX 1032, LX 1606, Orphan Drug Designation, Priority review, telotristat ethyl, Texas-based, Woodlands, Xermelo

Award for me, 100 Most Impactful Health care Leaders, Global listing

FDA approves Odactra for house dust mite allergies

$
0
0
Image result for fda approved
03/01/2017
The U.S. Food and Drug Administration today approved Odactra, the first allergen extract to be administered under the tongue (sublingually) to treat house dust mite (HDM)-induced nasal inflammation (allergic rhinitis), with or without eye inflammation (conjunctivitis), in people 18 through 65 years of age.

March 1, 2017

Release

The U.S. Food and Drug Administration today approved Odactra, the first allergen extract to be administered under the tongue (sublingually) to treat house dust mite (HDM)-induced nasal inflammation (allergic rhinitis), with or without eye inflammation (conjunctivitis), in people 18 through 65 years of age.

“House dust mite allergic disease can negatively impact a person’s quality of life,” said Peter Marks, M.D., Ph.D., director of the FDA’s Center for Biologics Evaluation and Research. “The approval of Odactra provides patients an alternative treatment to allergy shots to help address their symptoms.”

House dust mite allergies are a reaction to tiny bugs that are commonly found in house dust. Dust mites, close relatives of ticks and spiders, are too small to be seen without a microscope. They are found in bedding, upholstered furniture and carpeting. Individuals with house dust mite allergies may experience a cough, runny nose, nasal itching, nasal congestion, sneezing, and itchy and watery eyes.

Odactra exposes patients to house dust mite allergens, gradually training the immune system in order to reduce the frequency and severity of nasal and eye allergy symptoms. It is a once-daily tablet, taken year round, that rapidly dissolves after it is placed under the tongue. The first dose is taken under the supervision of a health care professional with experience in the diagnosis and treatment of allergic diseases. The patient is to be observed for at least 30 minutes for potential adverse reactions. Provided the first dose is well tolerated, patients can then take Odactra at home. It can take about eight to 14 weeks of daily dosing after initiation of Odactra for the patient to begin to experience a noticeable benefit.

The safety and efficacy of Odactra was evaluated in studies conducted in the United States, Canada and Europe, involving approximately 2,500 people. Some participants received Odactra, while others received a placebo pill. Participants reported their symptoms and the need to use symptom-relieving allergy medications. During treatment, participants taking Odactra experienced a 16 to 18 percent reduction in symptoms and the need for additional medications compared to those who received a placebo.

The most commonly reported adverse reactions were nausea, itching in the ears and mouth, and swelling of the lips and tongue. The prescribing information includes a boxed warning that severe allergic reactions, some of which can be life-threatening, can occur. As with other FDA-approved allergen extracts administered sublingually, patients receiving Odactra should be prescribed auto-injectable epinephrine. Odactra also has a Medication Guide for distribution to the patient.

Odactra is manufactured for Merck, Sharp & Dohme Corp., (a subsidiary of Merck and Co., Inc., Whitehouse Station, N.J.) by Catalent Pharma Solutions Limited, United Kingdom.

(sublingually) to treat house dust mite (HDM)-induced nasal inflammation (allergic rhinitis), with or without eye inflammation (conjunctivitis), in people 18 through 65 years of age

/////////////Odactra,  Merck, Sharp & Dohme Corp,  Catalent Pharma Solutions Limited, United Kingdom, FDA 2017, approves,  house dust mite allergies


Filed under: FDA 2017, Uncategorized Tagged: Approves, Catalent Pharma Solutions Limited, FDA 2017, house dust mite allergies, MERCK, Odactra, Sharp & Dohme Corp, United Kingdom

Oxymetazoline, оксиметазолин , أوكسيميتازولين , 羟甲唑啉

$
0
0

Oxymetazoline.svg

Oxymetazoline

1491-59-4   CAS NUMBER

  • Molecular FormulaC16H24N2O
  • Average mass260.375 Da
Phenol, 3-[(4,5-dihydro-1H-imidazol-2-yl)methyl]-6-(1,1-dimethylethyl)-2,4-dimethyl-
оксиметазолин
أوكسيميتازولين
羟甲唑啉

Oxymetazoline is a selective α1 adrenergic receptor agonist and α2 adrenergic receptorpartial agonist. It is a topical decongestant, used in the form of oxymetazoline hydrochloride. It was developed from xylometazoline at E. Merck Darmstadt by Fruhstorfer in 1961.[1]Oxymetazoline is generally available as a nasal spray.

Oxymetazoline HCl; CAS 2315-02-8

Oxymetazoline hydrochloride is a vasoconstrictor. Chemically it is 3-[(4,5-dihydro-1H-imidazol-2-yl)methyl]6-(1,1,-dimethylethyl)-2,4-dimethylphenolmono-hydrochloride. Its molecular weight is 296.8 for the hydrochloride salt and 260.4 for the free base. It is freely soluble in water and ethanol and has a partition coefficient of 0.1 in octanol/water. Its structural formula is:

 

Oxymetazoline hydrochloride - Structural Formula Illustration

 

Medical uses

Oxymetazoline is available over-the-counter as a topical decongestant in the form of oxymetazoline hydrochloride in nasal sprays such as Afrin, Operil, Dristan, Dimetapp, oxyspray, Facimin, Nasivin, Nostrilla, Sudafed OM, Vicks Sinex, Zicam, SinuFrin, and Mucinex Full Force.[2]

Due to its vasoconstricting properties, oxymetazoline is also used to treat nose bleeds[3][4]and eye redness due to minor irritation (marketed as Visine L.R. in the form of eye drops).[5]

Company:

Allergan

Approval Status:

Approved January 2017

Specific Treatments:

facial erythema associated with rosacea

Therapeutic Areas

Dermatology

Find Related Trials for The Following Conditions

Rosacea

General Information

Rhofade (oxymetazoline hydrochloride) is an alpha1A adrenoceptor agonist. Oxymetazoline acts as a vasoconstrictor.

Rhofade is spccifically indicated for the topical treatment of persistent facial erythema associated with rosacea in adults.

Rhofade is supplied as a cream for topical administration. Apply a pea-sized amount of Rhofade cream, once daily in a thin layer to cover the entire face (forehead, nose, each cheek, and chin) avoiding the eyes and lips. Wash hands immediately after applying Rhofade cream.

Image result for oxymetazoline hydrochloride uses

Side effects and special considerations

Rebound congestion

It is recommended that oxymetazoline not be used for more than three days, as rebound congestion, or rhinitis medicamentosa, may occur.[6] Patients who continue to use oxymetazoline beyond this point may become dependent on the medication to relieve their chronic congestion.

Effects of benzalkonium chloride

Some studies have found that benzalkonium chloride, a common additive to oxymetazoline nasal sprays, may damage nasal epithelia and exacerbate rhinitis medicamentosa. However, the majority of studies find benzalkonium chloride to be a safe preservative.[7]

Use in pregnancy

The Food and Drug Administration places oxymetazoline in category C, indicating risk to the fetus cannot be ruled out. While it has been shown that a single dose does not significantly alter either maternal or fetal circulation,[8] this subject has not been studied extensively enough to draw reliable conclusions.

Overdose

If accidentally ingested, standard methods to remove unabsorbed drugs should be considered.[clarification needed] There is no specific antidote for oxymetazoline, although its pharmacological effects may be reversed by α adrenergic antagonists such as phentolamine. In the event of a possibly life-threatening overdose (such as a hypertensive crisis), benzodiazepines should be considered to decrease the likelihood of seizures and convulsions, as well as reduce anxiety and to lower blood pressure. In children, oxymetazoline may produce profound central nervous system depression due to stimulation of central α2receptors and imidazoline receptors, much like clonidine.

Pharmacology

Mechanism of action

Oxymetazoline is a sympathomimetic that selectively agonizes α1 and, partially, α2 adrenergic receptors.[9] Since vascular beds widely express α1 receptors, the action of oxymetazoline results in vasoconstriction. In addition, the local application of the drug also results in vasoconstriction due to its action on endothelial postsynaptic α2 receptors; systemic application of α2 agonists, in contrast, causes vasodilation because of centrally-mediated inhibition of sympathetic tone via presynaptic α2 receptors.[10] Vasoconstriction of vessels results in relief of nasal congestion in two ways: first, it increases the diameter of the airway lumen; second, it reduces fluid exudation from postcapillary venules.[11] It can reduce nasal airway resistance (NAR) up to 35.7% and nasal mucosal blood flow up to 50%.[12]

Pharmacokinetics

Imidazolines are sympathomimetic agents, with primary effects on α adrenergic receptors and little if any effect on β adrenergic receptors. Oxymetazoline is readily absorbed orally. Effects on α receptors from systemically absorbed oxymetazoline hydrochloride may persist for up to 7 hours after a single dose. The elimination half-life in humans is 5–8 hours. It is excreted unchanged both by the kidneys (30%) and in feces (10%).

History

The oxymetazoline brand Afrin was first sold as a prescription medication in 1966. After finding substantial early success as a prescription medication, it became available as an over-the-counter drug in 1975. Schering-Plough did not engage in heavy advertising until 1986.[13]From the late 1980s to mid 1990s, Afrin featured in many notable television advertisements. Some of these commercials showed men, women, and children using other brands of nasal sprays, and then standing upside down or hanging upside down from playground equipment to prevent their nasal spray from dripping out. This was juxtaposed with Afrin users having no problems.

Society and culture

Brand names

Brand names include Afrin, Dristan, Nasivin, Nezeril, Nostrilla, Logicin, Vicks Sinex, Visine L.R., Sudafed OM, Zicam, SinuFrin and Mucinex Sinus-Max.

Image result for oxymetazoline SYNTHESIS

 

Image result for oxymetazoline hydrochloride

References

  1. Jump up^ German Patent 1,117,588
  2. Jump up^ “Oxymetazoline: Drug Information Provided by Lexi-Comp: Merck Manual Professional”. Merck.com. Retrieved 2013-04-15.
  3. Jump up^ Katz, Robert I.; Hovagim, Alec R.; Finkelstein, Harvey S.; Grinberg, Yair; Boccio, Remigio V.; Poppers, Paul J. (1990). “A comparison of cocaine, lidocaine with epinephrine, and oxymetazoline for prevention of epistaxis on nasotracheal intubation”. Journal of Clinical Anesthesia. 2 (1): 16–20. doi:10.1016/0952-8180(90)90043-3. PMID 2310576.
  4. Jump up^ Krempl, G. A.; Noorily, A. D. (1995). “Use of oxymetazoline in the management of epistaxis”. The Annals of otology, rhinology, and laryngology. 104 (9 Pt 1): 704–6.PMID 7661519.
  5. Jump up^ “VISINE® Original Red Eye Drops | VISINE® products”. Visine.com. Retrieved2013-04-15.
  6. Jump up^ Ramey, J. T.; Bailen, E; Lockey, R. F. (2006). “Rhinitis medicamentosa”. Journal of investigational allergology & clinical immunology. 16 (3): 148–55. PMID 16784007.
  7. Jump up^ Marple, B; Roland, P; Benninger, M (2004). “Safety review of benzalkonium chloride used as a preservative in intranasal solutions: An overview of conflicting data and opinions”. Otolaryngology – Head and Neck Surgery. 130 (1): 131–41.doi:10.1016/j.otohns.2003.07.005. PMID 14726922.
  8. Jump up^ Rayburn, W. F.; Anderson, J. C.; Smith, C. V.; Appel, L. L.; Davis, S. A. (1990).“Uterine and fetal Doppler flow changes from a single dose of a long-acting intranasal decongestant”. Obstetrics and gynecology. 76 (2): 180–2. PMID 2196495.
  9. Jump up^ Westfall Thomas C, Westfall David P, “Chapter 6. Neurotransmission: The Autonomic and Somatic Motor Nervous Systems” (Chapter). Brunton LL, Lazo JS, Parker KL: Goodman & Gilman’s The Pharmacological Basis of Therapeutics, 11e: http://www.accessmedicine.com/content.aspx?aID=954433.
  10. Jump up^ Biaggioni Italo, Robertson David, “Chapter 9. Adrenoceptor Agonists & Sympathomimetic Drugs” (Chapter). Katzung BG: Basic & Clinical Pharmacology, 11e: http://www.accessmedicine.com/content.aspx?aID=4520412.
  11. Jump up^ Widdicombe, John (1997). “Microvascular anatomy of the nose”. Allergy. 52 (40 Suppl): 7–11. doi:10.1111/j.1398-9995.1997.tb04877.x. PMID 9353554.
  12. Jump up^ Bende, M.; Löth, S. (2007). “Vascular effects of topical oxymetazoline on human nasal mucosa”. The Journal of Laryngology & Otology. 100 (3): 285–8.doi:10.1017/S0022215100099151. PMID 3950497.
  13. Jump up^ Dougherty, Phillip H. (20 October 1986). “Advertising; Afrin Goes After Users Of Nasal Decongestants”. The New York Times. The New York Times Company. Retrieved 2015-03-30.
Oxymetazoline
Oxymetazoline.svg
Clinical data
Trade names Afrin, Ocuclear, Drixine
AHFS/Drugs.com Monograph
Pregnancy
category
  • C
Dependence
liability
Moderate
Routes of
administration
Intranasal
ATC code
Legal status
Legal status
Pharmacokinetic data
Metabolism Kidney (30%), fecal (10%)
Biological half-life 5–6 hours
Identifiers
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
ECHA InfoCard 100.014.618
Chemical and physical data
Formula C16H24N2O
Molar mass 260.375 g·mol−1
3D model (Jmol)
Melting point 301.5 °C (574.7 °F)

/////////Oxymetazoline, оксиметазолин , أوكسيميتازولين , 羟甲唑啉 , Rhofade, oxymetazoline hydrochloride, alpha1A adrenoceptor agonist, vasoconstrictor

CC1=CC(=C(C(=C1CC2=NCCN2)C)O)C(C)(C)C.Cl


Filed under: FDA 2017, Uncategorized Tagged: alpha1A adrenoceptor agonist, 羟甲唑啉, оксиметазолин, Oxymetazoline, oxymetazoline hydrochloride, Rhofade, vasoconstrictor, أوكسيميتازولين
Viewing all 2871 articles
Browse latest View live


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