AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

INCB24360 (epacadostat)

 phase 2, Uncategorized  Comments Off on INCB24360 (epacadostat)
Apr 182016
 

 ChemSpider 2D Image | epacadostat | C11H13BrFN7O4S

Epacadostat
(Z)-N-(3-bromo-4-fluorophenyl)-N’-hydroxy-4-[2-(sulfamoylamino)ethylamino]-1,2,5-oxadiazole-3-carboxamidine
1,2,5-Oxadiazole-3-carboximidamide, 4-[[2-[(aminosulfonyl)amino]ethyl]amino]-N-(3-bromo-4-fluorophenyl)-N’-hydroxy-
1204669-58-8
INCB024360
N-(3-Brom-4-fluorphenyl)-N’-hydroxy-4-{[2-(sulfamoylamino)ethyl]amino}-1,2,5-oxadiazol-3-carboximidamid
UNII 71596A9R13
(Z)-N-(3-bromo-4-fluorophenyl)-N’-hydroxy-4-(2-(sulfamoylamino)ethylamino)-1,2,5-oxadiazole-3-carboximidamide
1,2,5-Oxadiazole-3-carboximidamide, 4-[[2-[(aminosulfonyl)amino]ethyl]amino]-N’-(3-bromo-4-fluorophenyl)-N-hydroxy-

Molecular Formula, C11H13BrFN7O4S

Average mass438.233 Da

cas 1204669-58-8 (or 1204669-37-3)

Synonym: IDO1 inhibitor INCB024360
indoleamine-2,3-dioxygenase inhibitor INCB024360
Code name: INCB 024360
INCB024360
Chemical structure: 1,2,5-Oxadiazole-3-carboximidamide, 4-((2-((Aminosulfonyl)amino)ethyl)amino)-N-(3-bromo-4-fluorophenyl)-N’-hydroxy-, (C(Z))-
Company Incyte Corp.
Description Indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor
Molecular Target Indoleamine 2,3-dioxygenase 1 (IDO1)
Mechanism of Action Indoleamine 2,3-dioxygenase (INDO) inhibitor
Therapeutic Modality Small molecule

 

  • OriginatorIncyte Corporation
  • DeveloperFred Hutchinson Cancer Research Center; Incyte Corporation; Merck AG
  • ClassAmides; Antineoplastics; Imides; Oxadiazoles; Small molecules
    • Phase IIFallopian tube cancer; Malignant melanoma; Non-small cell lung cancer; Ovarian cancer; Peritoneal cancer; Solid tumours

    Most Recent Events

    • 15 Jan 2016Phase-II clinical trials in Solid tumours (Combination therapy, Late-stage disease, Second-line therapy or greater) in USA (PO)
    • 11 Jan 2016Phase-II clinical trials in Non-small cell lung cancer (Combination therapy, Late-stage disease, Second-line therapy or greater) in USA (PO)
    • 11 Jan 2016The US FDA and Health Canada approve IND application and Clinical Trial Application, respectively, for a phase Ib trial in Ovarian cancer (Combination therapy, Recurrent, Second-line therapy or greater)

In 2016, orphan drug designation was assigned to the compound in the US. for the treatment of stage IIB-IV melanoma

EpacadostatAn orally available hydroxyamidine and inhibitor of indoleamine 2,3-dioxygenase (IDO1), with potential immunomodulating and antineoplastic activities. epacadostat targets and binds to IDO1, an enzyme responsible for the oxidation of tryptophan into kynurenine. By inhibiting IDO1 and decreasing kynurenine in tumor cells, epacadostat increases and restores the proliferation and activation of various immune cells, including dendritic cells (DCs), NK cells, and T-lymphocytes, as well as interferon (IFN) production, and a reduction in tumor-associated regulatory T cells (Tregs). Activation of the immune system, which is suppressed in many cancers, may inhibit the growth of IDO1-expressing tumor cells. IDO1 is overexpressed by a variety of tumor cell types and DCsINCB24360 (epacadostat), An Agent For Cancer Immunotherapy

Incyte and Merck Expand Clinical Collaboration to Include Phase 3 Study Investigating the Combination of Epacadostat with Keytruda® (pembrolizumab) as First-line Treatment for Advanced Melanoma

Pivotal study to evaluate Incyte’s IDO1 inhibitor in combination with Merck’s anti-PD-1 therapy in patients with advanced or metastatic melanoma

WILMINGTON, Del. and KENILWORTH, N.J. — October 13, 2015 — Incyte Corporation (Nasdaq: INCY) and Merck (NYSE:MRK), known as MSD outside the United States and Canada, today announced the expansion of the companies’ ongoing clinical collaboration to include a Phase 3 study evaluating the combination of epacadostat, Incyte’s investigational selective IDO1 inhibitor, with Keytruda® (pembrolizumab), Merck’s anti-PD-1 therapy, as first-line treatment for patients with advanced or metastatic melanoma. The Phase 3 study, which is expected to begin in the first half of 2016, will be co-funded by Incyte and Merck.

“We are very pleased to expand our collaboration with Merck and to move the clinical development program for epacadostat in combination with Keytruda into Phase 3,” said Hervé Hoppenot, President and Chief Executive Officer of Incyte. “We believe the combination of these two immunotherapies shows promise and, if successfully developed, may help to improve clinical outcomes for patients with metastatic melanoma.”

“The initiation of this large Phase 3 study with Incyte in the first-line advanced melanoma treatment setting is an important addition to our robust immunotherapy clinical development program for Keytruda,” said Dr. Roger Dansey, senior vice president and therapeutic area head, oncology late-stage development, Merck Research Laboratories. “We continue to explore the benefit that Keytruda brings to patients suffering from advanced melanoma when used alone, and we are pleased to be able to add this important combination study with epacadostat to our Keytruda development program.”

Under the terms of the agreement Incyte and Merck have also agreed, for a period of two years, not to initiate new pivotal studies of an IDO1 inhibitor in combination with a PD-1/PD-L1 antagonist as first-line therapy in advanced or metastatic melanoma with any third party. During this time, the companies will each offer the other the opportunity to collaborate on any new pivotal study involving an IDO1 inhibitor in combination with a PD-1/PD-L1 antagonist for types of melanoma and lines of therapy outside of the current collaboration agreement.

The agreement is between Incyte and certain subsidiaries and Merck through its subsidiaries.

Epacadostat and Keytruda are part of a class of cancer treatments known as immunotherapies that are designed to enhance the body’s own defenses in fighting cancer; the two therapies target distinct regulatory components of the immune system. IDO1 is an immunosuppressive enzyme that has been shown to induce regulatory T cell generation and activation, and allow tumors to escape immune surveillance. Keytruda is a humanized monoclonal antibody that blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2. Preclinical evidence suggests that the combination of these two agents may lead to an enhanced anti-tumor immune response compared with either agent alone.

Safety and efficacy data from the ongoing Phase 1/2 study evaluating the combination of epacadostat with Keytruda in patients with advanced malignancies is scheduled to be highlighted as a late-breaking oral presentation (Abstract #142) at the upcoming Society for Immunotherapy of Cancer 30th Anniversary Annual Meeting & Associated Programs, November 4–8, 2015 at the Gaylord National Resort & Convention Center in National Harbor, MD.

Metastatic Melanoma

Melanoma, the most serious form of skin cancer, strikes adults of all ages and accounts for approximately five percent of all new cases of cancer in the United States each year. The number of new cases of melanoma continues to rise by almost three percent each year which translates to 76,000 new cases yearly in the U.S. alone.[i] The 5-year survival rate for late-stage or metastatic disease is 15 percent.[ii] 

About Epacadostat (INCB024360)

Indoleamine 2,3-dioxygenase 1 (IDO1) is an immunosuppressive enzyme that has been shown to induce regulatory T cell generation and activation, and allow tumors to escape immune surveillance. Epacadostat is an orally bioavailable small molecule inhibitor of IDO1 that has nanomolar potency in both biochemical and cellular assays and has demonstrated potent activity in enhancing T lymphocyte, dendritic cell and natural killer cell responses in vitro, with a high degree of selectivity. Epacadostat has shown proof-of-concept clinical data in patients with unresectable or metastatic melanoma in combination with the CTLA-4 inhibitor ipilimumab, and is currently in four proof-of-concept clinical trials with PD-1 and PD-L1 immune checkpoint inhibitors in a variety of cancer histologies.

PATENT

WO 2014066834

https://www.google.com/patents/WO2014066834A1?cl=en

EXAMPLE 1

4-({2-[(Aminosulfonyl)amino]ethyl}amino)- V-(3-bromo-4-fluorophenyl)- V -hydroxy- l,2,5-oxadiazole-3-carboximidamide

Figure imgf000055_0001

Step 1: 4-Amino-N’-hydroxy-l,2,5-oxadiazole-3-carboximidamide

[00184] Malononitrile (320.5 g, 5 mol) was added to water (7 L) preheated to 45 °C and stirred for 5 min. The resulting solution was cooled in an ice bath and sodium nitrite (380 g, 5.5 mol) was added. When the temperature reached 10 °C, 6 N hydrochloric acid (55 mL) was added. A mild exothermic reaction ensued with the temperature reaching 16 °C. After 15 min the cold bath was removed and the reaction mixture was stirred for 1.5 hrs at 16-18 °C. The reaction mixture was cooled to 13 °C and 50% aqueous hydroxylamine (990 g, 15 mol) was added all at once. The temperature rose to 26 °C. When the exothermic reaction subsided the cold bath was removed and stirring was continued for 1 hr at 26-27 °C, then it was slowly brought to reflux. Reflux was maintained for 2 hrs and then the reaction mixture was allowed to cool overnight. The reaction mixture was stirred in an ice bath and 6 N hydrochloric acid (800 mL) was added in portions over 40 min to pH 7.0. Stirring was continued in the ice bath at 5 °C. The precipitate was collected by filtration, washed well with water and dried in a vacuum oven (50 °C) to give the desired product (644 g, 90%). LCMS for C3H6N5O2

(M+H)+: m/z = 144.0. 13C MR (75 MHz, CD3OD): δ 156.0, 145.9, 141.3. Step 2: 4-Amino-N-hydroxy-l,2,5-oxadiazole-3-carboximidoyl chloride [00185] 4-Amino-N,-hydroxy-l ,2,5-oxadiazole-3-carboximidamide (422 g, 2.95 mol) was added to a mixture of water (5.9 L), acetic acid (3 L) and 6 Ν hydrochloric acid (1.475 L, 3 eq.) and this suspension was stirred at 42 – 45 °C until complete solution was achieved. Sodium chloride (518 g, 3 eq.) was added and this solution was stirred in an ice/water/methanol bath. A solution of sodium nitrite (199.5 g, 0.98 eq.) in water (700 mL) was added over 3.5 hrs while maintaining the temperature below 0 °C. After complete addition stirring was continued in the ice bath for 1.5 hrs and then the reaction mixture was allowed to warm to 15 °C. The precipitate was collected by filtration, washed well with water, taken in ethyl acetate (3.4 L), treated with anhydrous sodium sulfate (500 g) and stirred for 1 hr. This suspension was filtered through sodium sulfate (200 g) and the filtrate was concentrated on a rotary evaporator. The residue was dissolved in methyl i-butyl ether (5.5 L), treated with charcoal (40 g), stirred for 40 min and filtered through Celite. The solvent was removed in a rotary evaporator and the resulting product was dried in a vacuum oven (45 °C) to give the desired product (256 g, 53.4%). LCMS for C3H4CIN4O2 (M+H)+: m/z = 162.9. 13C NMR (100 MHz, CD3OD): 5 155.8, 143.4, 129.7.

Step 3: 4-Amino-N’-hydroxy-N-(2-methoxyethyl)-l,2,5-oxadiazole-3-carboximidamide [00186] 4-Amino-N-hydroxy-l ,2,5-oxadiazole-3-carboximidoyl chloride (200.0 g, 1.23 mol) was mixed with ethyl acetate (1.2 L). At 0-5 °C 2-methoxyethylamine [Aldrich, product # 143693] (119.0 mL, 1.35 mol) was added in one portion while stirring. The reaction temperature rose to 41 °C. The reaction was cooled to 0 – 5 °C. Triethylamine (258 mL, 1.84 mol) was added. After stirring 5 min, LCMS indicated reaction completion. The reaction solution was washed with water (500 mL) and brine (500 mL), dried over sodium sulfate, and concentrated to give the desired product (294 g, 1 19%) as a crude dark oil.

LCMS for C6Hi2 503 (M+H)+: m/z = 202.3. 1H NMR (400 MHz, DMSO- ): δ 10.65 (s, 1 H), 6.27 (s, 2 H), 6.10 (t, J = 6.5 Hz, 1 H), 3.50 (m, 2 H), 3.35 (d, J = 5.8 Hz, 2 H), 3.08 (s, 3 H).

Step 4: N’-Hydroxy-4-[(2-methoxyethyl)amino]-l,2,5-oxadiazole-3-carboximidamide

[00187] 4-Amino-N-hydroxy-N-(2-methoxyethyl)-l,2,5-oxadiazole-3- carboximidamide (248.0 g, 1.23 mol) was mixed with water (1 L). Potassium hydroxide (210 g, 3.7 mol) was added. The reaction was refluxed at 100 °C overnight (15 hours). TLC with 50% ethyl acetate (containing 1% ammonium hydroxide) in hexane indicated reaction completed (product Rf = 0.6, starting material Rf = 0.5). LCMS also indicated reaction completion. The reaction was cooled to room temperature and extracted with ethyl acetate (3 x 1 L). The combined ethyl acetate solution was dried over sodium sulfate and concentrated to give the desired product (201 g, 81%) as a crude off-white solid. LCMS for C6H12N5O3 (M+H)+: m/z = 202.3 LH NMR (400 MHz, OMSO-d6): δ 10.54 (s, 1 H), 6.22 (s, 2 H), 6.15 (t, J = 5.8 Hz, 1 H), 3.45 (t, J= 5.3 Hz, 2 H), 3.35 (m, 2 H), 3.22 (s, 3 H). Step 5: N-Hydroxy-4-[(2-methoxyethyl)amino]-l,2,5-oxadiazole-3-carboximidoyl chloride

[00188] At room temperature N’-hydroxy-4-[(2-methoxyethyl)amino]- 1 ,2,5- oxadiazole-3-carboximidamide (50.0 g, 0.226 mol) was dissolved in 6.0 M hydrochloric acid aqueous solution (250 mL, 1.5 mol). Sodium chloride (39.5 g, 0.676 mol) was added followed by water (250 mL) and ethyl acetate (250 mL). At 3-5 °C a previously prepared aqueous solution (100 mL) of sodium nitrite (15.0 g, 0.217 mol) was added slowly over 1 hr. The reaction was stirred at 3 – 8 °C for 2 hours and then room temperature over the weekend. LCMS indicated reaction completed. The reaction solution was extracted with ethyl acetate (2 x 200 mL). The combined ethyl acetate solution was dried over sodium sulfate and concentrated to give the desired product (49.9 g, 126%) as a crude white solid. LCMS for

C6HioClN403 (M+H)+: m/z = 221.0. !H NMR (400 MHz, DMSO-d6): δ 13.43 (s, 1 H), 5.85 (t, J= 5.6 Hz, 1 H), 3.50 (t, J= 5.6 Hz, 2 H), 3.37(dd, J= 10.8, 5.6 Hz, 2 H), 3.25 (s, 3 H).

Step 6 : N-(3-Bromo-4-fluorophenyl)-N’-hydroxy-4- [(2-methoxyethyl)amino] – 1 ,2,5- oxadiazole-3-carboximidamide [00189] N-Hydroxy-4-[(2-methoxyethyl)amino]- 1 ,2,5-oxadiazole-3-carboximidoyl chloride (46.0 g, 0.208 mol) was mixed with water (300 mL). The mixture was heated to 60 °C. 3-Bromo-4-fluoroaniline [Oakwood products, product # 013091] (43.6 g, 0.229 mol) was added and stirred for 10 min. A warm sodium bicarbonate (26.3 g, 0.313 mol) solution (300 mL water) was added over 15 min. The reaction was stirred at 60 °C for 20 min. LCMS indicated reaction completion. The reaction solution was cooled to room temperature and extracted with ethyl acetate (2 x 300 mL). The combined ethyl acetate solution was dried over sodium sulfate and concentrated to give the desired product (76.7 g, 98%) as a crude brown solid. LCMS for Ci2Hi4BrF503 (M+H)+: m/z = 374.0, 376.0. 1H NMR (400 MHz, DMSO- tf): δ 11.55 (s, 1 H), 8.85 (s, 1 H), 7.16 (t, J= 8.8 Hz, 1 H), 7.08 (dd, J= 6.1, 2.7 Hz, 1 H), 6.75 (m, 1 H), 6.14 (t, J= 5.8 Hz, 1 H), 3.48 (t, J = 5.2 Hz, 2 H), 3.35 (dd, J= 10.8, 5.6 Hz, 2 H), 3.22 (s, 3 H).

Step 7: 4-(3-Bromo-4-fluorophenyl)-3-{4- [(2-methoxyethyl)amino]-l,2,5-oxadiazol-3- yl}-l,2,4-oxadiazol-5(4H)-one

[00190] A mixture of N-(3-bromo-4-fluorophenyl)-N’-hydroxy-4-[(2- methoxyethyl)amino]-l,2,5-oxadiazole-3-carboximidamide (76.5 g, 0.204 mol), 1,1 ‘- carbonyldiimidazole (49.7 g, 0.307 mol), and ethyl acetate (720 mL) was heated to 60 °C and stirred for 20 min. LCMS indicated reaction completed. The reaction was cooled to room temperature, washed with 1 N HC1 (2 x 750 mL), dried over sodium sulfate, and concentrated to give the desired product (80.4 g, 98%) as a crude brown solid. LCMS for

Figure imgf000058_0001

(M+H)+: m/z = 400.0, 402.0. 1H NMR (400 MHz, DMSO-c½): δ 7.94 (t, J = 8.2 Hz, 1 H), 7.72 (dd, J = 9.1, 2.3 Hz, 1 H), 7.42 (m, 1 H), 6.42 (t, J= 5.7 Hz, 1 H), 3.46 (t, J = 5.4 Hz, 2 H), 3.36 (t, J= 5.8 Hz, 2 H), 3.26 (s, 3 H).

Step 8: 4-(3-Bromo-4-fluorophenyl)-3-{4-[(2-hydroxyethyl)amino]-l,2,5-oxadiazol-3- yl}-l,2,4-oxadiazol-5(4H)-one

[00191] 4-(3-Bromo-4-fluoroplienyl)-3-{4-[(2-metlioxyethyl)amino]-l,2,5-oxadiazol- 3-yl}-l,2,4-oxadiazol-5(4H)-one (78.4 g, 0.196 mol) was dissolved in dichloromethane (600 mL). At -67 °C boron tribromide (37 mL, 0.392 mol) was added over 15 min. The reaction was warmed up to -10 °C in 30 min. LCMS indicated reaction completed. The reaction was stirred at room temperature for 1 hour. At 0 – 5 °C the reaction was slowly quenched with saturated sodium bicarbonate solution (1.5 L) over 30 min. The reaction temperature rose to 25 °C. The reaction was extracted with ethyl acetate (2 x 500 mL, first extraction organic layer is on the bottom and second extraction organic lager is on the top). The combined organic layers were dried over sodium sulfate and concentrated to give the desired product (75 g, 99%) as a crude brown solid. LCMS for Ci2HioBrFN504 (M+H)+: m/z = 386.0, 388.0.

1H NMR (400 MHz, DMSO-^): δ 8.08 (dd, J = 6.2, 2.5 Hz, 1 H), 7.70 (m, 1 H), 7.68 (t, J = 8.7 Hz, 1 H), 6.33 (t, J = 5.6 Hz, 1 H), 4.85 (t, J= 5.0 Hz, 1 H), 3.56 (dd, J= 10.6, 5.6 Hz, 2 H), 3.29 (dd, J= 11.5, 5.9 Hz, 2 H).

Step 9 : 2-({4- [4-(3-Bromo-4-fluorophenyl)-5-oxo-4,5-dihydro- 1 ,2,4-oxadiazol-3-yl] – l,2,5-oxadiazol-3-yl}amino)ethyl methanesulfonate

[00192] To a solution of 4-(3-bromo-4-fluorophenyl)-3-{4-[(2-hydroxyethyl)amino]- l,2,5-oxadiazol-3-yl}-l,2,4-oxadiazol-5(4H)-one (1.5 kg, 3.9 mol, containing also some of the corresponding bromo-compound) in ethyl acetate (12 L) was added methanesulfonyl chloride (185 mL, 2.4 mol) dropwise over 1 h at room temperature. Triethylamine (325 mL, 2.3 mol) was added dropwise over 45 min, during which time the reaction temperature increased to 35 °C. After 2 h, the reaction mixture was washed with water (5 L), brine (1 L), dried over sodium sulfate, combined with 3 more reactions of the same size, and the solvents removed in vacuo to afford the desired product (7600 g, quantitative yield) as a tan solid. LCMS for C HnBrFNsOeS a (M+Na)+: m/z = 485.9, 487.9. !H NMR (400 MHz, DMSO- d6): δ 8.08 (dd, J = 6.2, 2.5 Hz, 1 H), 7.72 (m, 1 H), 7.58 (t, J = 8.7 Hz, 1 H), 6.75 (t, J = 5.9 Hz, 1 H), 4.36 (t, J = 5.3 Hz, 2 H), 3.58 (dd, J = 11.2, 5.6 Hz, 2 H), 3.18 (s, 3 H).

Step 10: 3-{4-[(2-Azidoethyl)amino]-l,2,5-oxadiazol-3-yl}-4-(3-bromo-4-fluorophenyl)- l,2,4-oxadiazol-5(4H)-one

To a solution of 2-({4-[4-(3-bromo-4-f uorophenyl)-5-oxo-4,5-dihydro-l ,2,4- oxadiazol-3-yl]-l ,2,5-oxadiazol-3-yl}amino)ethyl methanesulfonate (2.13 kg, 4.6 mol, containing also some of the corresponding bromo-compound) in dimethylformamide (4 L) stirring in a 22 L flask was added sodium azide (380 g, 5.84 mol). The reaction was heated at 50 °C for 6 h, poured into ice/water (8 L), and extracted with 1 : 1 ethyl acetate:heptane (20 L). The organic layer was washed with water (5 L) and brine (5 L), and the solvents removed in vacuo to afford the desired product (1464 g, 77%) as a tan solid. LCMS for CnHgBrFNsOs a

(M+Na)+: m/z = 433.0, 435.0. !H NMR (400 MHz, DMSO-J6): δ 8.08 (dd, J = 6.2, 2.5 Hz, 1 H), 7.72 (m, 1 H), 7.58 (t, J= 8.7 Hz, 1 H), 6.75 (t, J = 5.7 Hz, 1 H), 3.54 (t, J = 5.3 Hz, 2 H), 3.45 (dd, J= 1 1.1 , 5.2 Hz, 2 H).

Step 11: 3-{4-[(2-Aminoethyl)amino]-l,2,5-oxadiazol-3-yl}-4-(3-bromo-4-fluorophenyl)-

1.2.4- oxadiazol-5(4H)-one hydrochloride

[00194] Sodium iodide (1080 g, 7.2 mol) was added to 3-{4-[(2-azidoethyl)amino]-

1.2.5- oxadiazol-3-yl}-4-(3-bromo-4-fluorophenyl)-l ,2,4-oxadiazol-5(4H)-one (500 g, 1.22 mol) in methanol (6 L). The mixture was allowed to stir for 30 min during which time a mild exotherm was observed. Chlorotrimethylsilane (930 mL, 7.33 mol) was added as a solution in methanol (1 L) dropwise at a rate so that the temperature did not exceed 35 °C, and the reaction was allowed to stir for 3.5 h at ambient temperature. The reaction was neutralized with 33 wt% solution of sodium thiosulfate pentahydrate in water (-1.5 L), diluted with water (4 L), and the pH adjusted to 9 carefully with solid potassium carbonate (250 g – added in small portions: watch foaming). Di-ieri-butyl dicarbonate (318 g, 1.45 mol) was added and the reaction was allowed to stir at room temperature. Additional potassium carbonate (200 g) was added in 50 g portions over 4 h to ensure that the pH was still at or above 9. After stirring at room temperature overnight, the solid was filtered, triturated with water (2 L), and then MTBE (1.5 L). A total of 11 runs were performed (5.5 kg, 13.38 mol). The combined solids were triturated with 1 : 1 THF:dichloromethane (24 L, 4 runs in a 20 L rotary evaporator flask, 50 °C, 1 h), filtered, and washed with dichloromethane (3 L each run) to afford an off- white solid. The crude material was dissolved at 55 °C tetrahydrofuran (5 mL/g), treated with decolorizing carbon (2 wt%) and silica gel (2 wt%), and filtered hot through celite to afford the product as an off-white solid (5122 g). The combined MTBE, THF, and dichloromethane filtrates were concentrated in vacuo and chromatographed (2 kg silica gel, heptane with a 0-100% ethyl acetate gradient, 30 L) to afford more product (262 g). The combined solids were dried to a constant weight in a convection oven (5385 g, 83%).

In a 22 L flask was charged hydrogen chloride (4 N solution in 1 ,4-dioxane, 4 L, 16 mol). tert-Butyl [2-({4-[4-(3-bromo-4-fluorophenyl)-5-oxo-4,5-dihydro-l ,2,4- oxadiazol-3-yl]-l ,2,5-oxadiazol-3-yl}amino)ethyl]carbamate (2315 g, 4.77 mol) was added as a solid in portions over 10 min. The slurry was stirred at room temperature and gradually became a thick paste that could not be stirred. After sitting overnight at room temperature, the paste was slurried in ethyl acetate (10 L), filtered, re-slurried in ethyl acetate (5 L), filtered, and dried to a constant weight to afford the desired product as a white solid (combined with other runs, 5 kg starting material charged, 41 13 g, 95%). LCMS for

Ci2HnBrFN603 (M+H)+: m/z = 384.9, 386.9. 1H NMR (400 MHz, DMSO-^): δ 8.12 (m, 4 H), 7.76 (m, 1 H), 7.58 (t, J = 8.7 Hz, 1 H), 6.78 (t, J = 6.1 Hz, 1 H), 3.51 (dd, J = 1 1.8, 6.1 Hz, 2 H), 3.02 (m, 2 H).

Step 12: tert-Butyl ({[2-({4-[4-(3-bromo-4-nuorophenyl)-5-oxo-4,5-dihydro-l,2,4- oxadiazol-3-yl]-l,2,5-oxadiazol-3-yl}amino)ethyl]amino}sulfonyl)carbamate

A 5 L round bottom flask was charged with chlorosulfonyl isocyanate [Aldrich, product # 142662] (149 mL, 1.72 mol) and dichloromethane (1.5 L) and cooled using an ice bath to 2 °C. teri-Butanol (162 mL, 1.73 mol) in dichloromethane (200 mL) was added dropwise at a rate so that the temperature did not exceed 10 °C. The resulting solution was stirred at room temperature for 30-60 min to provide tert-bvAy\ [chlorosulfonyl]carbamate.

A 22 L flask was charged with 3- {4-[(2-aminoethyl)amino]- 1 ,2,5-oxadiazol-3- yl}-4-(3-bromo-4-fluorophenyl)-l,2,4-oxadiazol-5(4H)-one hydrochloride (661 g, 1.57 mol) and 8.5 L dichloromethane. After cooling to -15 °C with an ice/salt bath, the solution oi tert- Vmtvl i Vi 1 r>rosulfonyl]carbamate (prepared as above) was added at a rate so that the temperature did not exceed -10 °C (addition time 7 min). After stirring for 10 min, triethylamine (1085 mL, 7.78 mol) was added at a rate so that the temperature did not exceed -5 °C (addition time 10 min). The cold bath was removed, the reaction was allowed to warm to 10 °C, split into two portions, and neutralized with 10% cone HC1 (4.5 L each portion). Each portion was transferred to a 50 L separatory funnel and diluted with ethyl acetate to completely dissolve the white solid (-25 L). The layers were separated, and the organic layer was washed with water (5 L), brine (5 L), and the solvents removed in vacuo to afford an off- white solid. The solid was triturated with MTBE (2 x 1.5 L) and dried to a constant weight to afford a white solid. A total of 4113 g starting material was processed in this manner (5409 g, 98%). 1H NMR (400 MHz, DMSO-^): δ 10.90 (s, 1 H), 8.08 (dd, J = 6.2, 2.5 Hz, 1 H), 7.72 (m, 1 H), 7.59 (t, J = 8.6 Hz, 1 H), 6.58 (t, J = 5.7 Hz, 1 H), 3.38 (dd, J= 12.7, 6.2 Hz, 2 H), 3.10 (dd, J= 12.1 , 5.9 Hz, 2 H), 1.41 (s, 9 H).

Step 13: N-[2-({4-[4-(3-Bromo-4-fluorophenyl)-5-oxo-4,5-dihydro-l,2,4-oxadiazol-3-yl]- l,2,5-oxadiazol-3-yl}amino)ethyl]sulfamide

[00198] To a 22 L flask containing 98:2 trifluoroacetic acid:water (8.9 L) was added tert-bvXyl ({[2-({4-[4-(3-bromo-4-fluorophenyl)-5-oxo-4,5-dihydro-l,2,4-oxadiazol-3-yl]- l,2,5-oxadiazol-3-yl}amino)ethyl]amino}sulfonyl)carbamate (1931 g, 3.42 mol) in portions over 10 minutes. The resulting mixture was stirred at room temperature for 1.5 h, the solvents removed in vacuo, and chased with dichloromethane (2 L). The resulting solid was treated a second time with fresh 98:2 trifluoroacetic acid:water (8.9 L), heated for 1 h at 40- 50 °C, the solvents removed in vacuo, and chased with dichloromethane (3 x 2 L). The resulting white solid was dried in a vacuum drying oven at 50 °C overnight. A total of 5409 g was processed in this manner (4990 g, quant, yield). LCMS for C12H12BrFN705S (M+H)+: m/z = 463.9, 465.9. 1H NMR (400 MHz, DMSO- ): δ 8.08 (dd, J = 6.2, 2.5 Hz, 1 H), 7.72 (m, 1 H), 7.59 (t, J= 8.7 Hz, 1 H), 6.67 (t, J = 5.9 Hz, 1H), 6.52 (t, J= 6.0 Hz, 1 H), 3.38 (dd, J = 12.7, 6.3 Hz, 2 H), 3.11 (dd, J = 12.3, 6.3 Hz). Step 14: 4-({2-[(Aminosulfonyl)amino]ethyl}amino)-N-(3-bromo-4-fluorophenyl)-N’- hydroxy-l,2,5-oxadiazole-3-carboximidamide

Figure imgf000063_0001

[00199] To a crude mixture of N-[2-({4-[4-(3-bromo-4-fluorophenyl)-5-oxo-4,5- dihydro-l,2,4-oxadiazol-3-yl]-l,2,5-oxadiazol-3-yl}amino)ethyl]sulfamide (2.4 mol) containing residual amounts of trifluoroacetic acid stirring in a 22 L flask was added THF (5 L). The resulting solution was cooled to 0 °C using an ice bath and 2 N NaOH (4 L) was added at a rate so that the temperature did not exceed 10 °C. After stirring at ambient temperature for 3 h (LCMS indicated no starting material remained), the pH was adjusted to 3-4 with concentrated HC1 (-500 mL). The THF was removed in vacuo, and the resulting mixture was extracted with ethyl acetate (15 L). The organic layer was washed with water (5 L), brine (5 L), and the solvents removed in vacuo to afford a solid. The solid was triturated with MTBE (2 x 2 L), combined with three other reactions of the same size, and dried overnight in a convection oven to afford a white solid (3535 g). The solid was recrystallized (3 x 22 L flasks, 2:1 watenethanol, 14.1 L each flask) and dried in a 50 °C convection oven to a constant weight to furnish the title compound as an off-white solid (3290 g, 78%). LCMS for CnHnBrF yC S (M+H)+: m/z = 437.9, 439.9. i NMR (400 MHz, DMSO-J^): δ 11.51 (s, 1 H), 8.90 (s, 1 H), 7.17 (t, J= 8.8 Hz, 1 H), 7.11 (dd, J= 6.1, 2.7 Hz, 1 H), 6.76 (m, 1 H), 6.71 (t, J = 6.0 Hz, 1 H), 6.59 (s, 2 H), 6.23 (t, J= 6.1 Hz, 1 H), 3.35 (dd, J= 10.9, 7.0 Hz, 2 H), 3.10 (dd, J= 12.1, 6.2 Hz, 2 H).

PATENT

WO 2010005958

https://www.google.com/patents/WO2010005958A2?cl=en

EXAMPLES Example 1

4-({2-[(Aminosulfonyl)amino]ethyl}amino)-7V-(3-bromo-4-fluorophenyl)-iV’-hydroxy- l,2,5-oxadiazole-3-carboximidamide

Figure imgf000043_0001

Step A: 4-Amino-N’-hydroxy-l,2,5-oxadiazole-3-carboximidamide

Figure imgf000043_0002

Malononitrile [Aldrich, product # M1407] (320.5 g, 5 mol) was added to water (7 L) preheated to 45 0C and stirred for 5 min. The resulting solution was cooled in an ice bath and sodium nitrite (380 g, 5.5 mol) was added. When the temperature reached 10 0C, 6 N hydrochloric acid (55 mL) was added. A mild exothermic reaction ensued with the temperature reaching 16 0C. After 15 min the cold bath was removed and the reaction mixture was stirred for 1.5 hrs at 16-18 0C. The reaction mixture was cooled to 13 0C and 50% aqueous hydroxylamine (990 g, 15 mol) was added all at once. The temperature rose to 26 0C. When the exothermic reaction subsided the cold bath was removed and stirring was continued for 1 hr at 26-270C, then it was slowly brought to reflux. Reflux was maintained for 2 hrs and then the reaction mixture was allowed to cool overnight. The reaction mixture was stirred in an ice bath and 6 N hydrochloric acid (800 mL) was added in portions over 40 min to pH 7.0. Stirring was continued in the ice bath at 5 0C. The precipitate was collected by filtration, washed well with water and dried in a vacuum oven (50 0C) to give the desired product (644 g, 90%). LCMS for C3H6N5O2 (M+H)+: m/z = 144.0. 13C NMR (75 MHz, CD3OD): δ 156.0, 145.9, 141.3. Step B: 4-Amino-N-hydroxy-l,2,5-oxadiazole-3-carboximidoyl chloride

Figure imgf000044_0001

4-Amino-N’-hydroxy-l,2,5-oxadiazole-3-carboximidamide (422 g, 2.95 mol) was added to a mixture of water (5.9 L), acetic acid (3 L) and 6 Ν hydrochloric acid (1.475 L, 3 eq.) and this suspension was stirred at 42 – 45 0C until complete solution was achieved. Sodium chloride (518 g, 3 eq.) was added and this solution was stirred in an ice/water/methanol bath. A solution of sodium nitrite (199.5 g, 0.98 eq.) in water (700 mL) was added over 3.5 hrs while maintaining the temperature below 0 0C. After complete addition stirring was continued in the ice bath for 1.5 hrs and then the reaction mixture was allowed to warm to 15 0C. The precipitate was collected by filtration, washed well with water, taken in ethyl acetate (3.4 L), treated with anhydrous sodium sulfate (500 g) and stirred for 1 hr. This suspension was filtered through sodium sulfate (200 g) and the filtrate was concentrated on a rotary evaporator. The residue was dissolved in methyl f-butyl ether (5.5 L), treated with charcoal (40 g), stirred for 40 min and filtered through Celite. The solvent was removed in a rotary evaporator and the resulting product was dried in a vacuum oven (45 0C) to give the desired product (256 g, 53.4%). LCMS for C3H4ClN4O2(M+H)+: m/z = 162.9. 13c NMR (100 MHz, CD3OD): δ 155.8, 143.4, 129.7.

Step C: 4-Amino-N’-hydroxy-N-(2-methoxyethyl)- 1 ,2,5-oxadiazole-3-carboximidamide

Figure imgf000044_0002

4-Amino-N-hydroxy-l,2,5-oxadiazole-3-carboximidoyl chloride (200.0 g, 1.23 mol) was mixed with ethyl acetate (1.2 L). At 0-50C 2-methoxyethylamine [Aldrich, product # 143693] (119.0 mL, 1.35 mol) was added in one portion while stirring. The reaction temperature rose to 41 0C. The reaction was cooled to 0 – 5 °C. Triethylamine (258 mL, 1.84 mol) was added. After stirring 5 min, LCMS indicated reaction completion. The reaction solution was washed with water (500 mL) and brine (500 mL), dried over sodium sulfate, and concentrated to give the desired product (294 g, 119%) as a crude dark oil. LCMS for C6Hi2N5O3 (M+H)+: m/z = 202.3. 1H NMR (400 MHz, DMSO-J6): δ 10.65 (s, 1 H), 6.27 (s, 2 H), 6.10 (t, J= 6.5 Hz, 1 H), 3.50 (m, 2 H), 3.35 (d, J= 5.8 Hz, 2 H), 3.08 (s, 3 H).

Step D: N’-Hydroxy-4-[(2-methoxyethyl)amino]-l ,2,5-oxadiazole-3-carboximidamide

Figure imgf000045_0001

4-Amino-N’-hydroxy-N-(2-methoxyethyl)-l,2,5-oxadiazole-3-carboximidaniide (248.0 g, 1.23 mol) was mixed with water (1 L). Potassium hydroxide (210 g, 3.7 mol) was added. The reaction was refluxed at 100 0C overnight (15 hours). TLC with 50% ethyl acetate (containing 1% ammonium hydroxide) in hexane indicated reaction completed (product Rf= 0.6, starting material Rf = 0.5). LCMS also indicated reaction completion. The reaction was cooled to room temperature and extracted with ethyl acetate (3 x 1 L). The combined ethyl acetate solution was dried over sodium sulfate and concentrated to give the desired product (201 g, 81%) as a crude off-white solid. LCMS for C6H12N5O3 (M+H)+: m/z = 202.3 1H NMR (400 MHz, DMSO-Gk): δ 10.54 (s, 1 H), 6.22 (s, 2 H), 6.15 (t, J= 5.8 Hz, 1 H), 3.45 (t, J= 5.3 Hz, 2 H), 3.35 (m, 2 H), 3.22 (s, 3 H).

Step E: N-Hydroxy-4-[(2-methoxyethyl)amino]-l,2,5-oxadiazole-3-carboximidoyl chloride

Figure imgf000045_0002

Ν. ,Ν O

At room temperature N’-hydroxy-4-[(2-methoxyethyl)amino]-l,2,5-oxadiazole-3- carboximidamide (50.0 g, 0.226 mol) was dissolved in 6.0 M hydrochloric acid aqueous solution (250 mL, 1.5 mol). Sodium chloride (39.5 g, 0.676 mol) was added followed by water (250 mL) and ethyl acetate (250 mL). At 3-5 0C a previously prepared aqueous solution (100 mL) of sodium nitrite (15.0 g, 0.217 mol) was added slowly over 1 hr. The reaction was stirred at 3 – 8 0C for 2 hours and then room temperature over the weekend. LCMS indicated reaction completed. The reaction solution was extracted with ethyl acetate (2 x 200 mL). The combined ethyl acetate solution was dried over sodium sulfate and concentrated to give the desired product (49.9 g, 126%) as a crude white solid. LCMS for C6Hi0ClN4O3 (M+H)+: m/z = 221.0. 1H NMR (400 MHz, DMSO-J6): δ 13.43 (s, 1 H), 5.85 (t, J= 5.6 Hz, 1 H), 3.50 (t, J= 5.6 Hz, 2 H), 3.37(dd, J= 10.8, 5.6 Hz, 2 H), 3.25 (s, 3 H).

Step F: N-(3-Bromo-4-fluorophenyl)-N’-hydroxy-4-[(2-methoxyethyl)amino]- 1 ,2,5- oxadiazole-3 -carboximidamide

Figure imgf000046_0001

N-Hydroxy-4-[(2-methoxyethyl)amino]-l,2,5-oxadiazole-3-carboximidoyl chloride (46.0 g, 0.208 mol) was mixed with water (300 mL). The mixture was heated to 60 °C. 3-Bromo-4- fluoroaniline [Oakwood products, product # 013091] (43.6 g, 0.229 mol) was added and stirred for 10 nrnn. A warm sodium bicarbonate (26.3 g, 0.313 mol) solution (300 mL water) was added over 15 min. The reaction was stirred at 60 0C for 20 min. LCMS indicated reaction completion. The reaction solution was cooled to room temperature and extracted with ethyl acetate (2 x 300 mL). The combined ethyl acetate solution was dried over sodium sulfate and concentrated to give the desired product (76.7 g, 98%) as a crude brown solid. LCMS for Ci2Hi4BrFN5O3 (M+H)+: m/z = 374.0, 376.0. 1H NMR (400 MHz, DMSO-J6): δ 11.55 (s, 1 H), 8.85 (s, 1 H), 7.16 (t, J= 8.8 Hz, 1 H), 7.08 (dd, J= 6.1, 2.7 Hz, 1 H), 6.75 (m, 1 H), 6.14 (t, J= 5.8 Hz, 1 H), 3.48 (t, J= 5.2 Hz, 2 H), 3.35 (dd, J= 10.8, 5.6 Hz, 2 H), 3.22 (s, 3 H).

Step G: 4-(3-Bromo-4-fluorophenyl)-3-{4-[(2-methoxyethyl)amino]-l,2,5-oxadiazol-3-yl}- 1 ,2,4-oxadiazol-5(4H)-one

Figure imgf000046_0002

A mixture of N-(3-bromo-4-fluorophenyl)-N’-hydroxy-4-[(2-methoxyethyl)amino]-l,2,5- oxadiazole-3-carboximidamide (76.5 g, 0.204 mol), l,r-carbonyldiimidazole (49.7 g, 0.307 mol), and ethyl acetate (720 mL) was heated to 60 0C and stirred for 20 min. LCMS indicated reaction completed. The reaction was cooled to room temperature, washed with 1 Ν HCl (2 x 750 mL), dried over sodium sulfate, and concentrated to give the desired product (80.4 g, 98%) as a crude brown solid. LCMS for C13H12BrFN5O4 (M+H)+: m/z = 400.0, 402.0. 1H NMR (400 MHz, OMSO-d6): δ 7.94 (t, J= 8.2 Hz, 1 H), 7.72 (dd, J= 9.1, 2.3 Hz, 1 H), 7.42 (m, 1 H), 6.42 (t, J= 5.7 Hz, 1 H), 3.46 (t, J= 5.4 Hz, 2 H), 3.36 (t, J= 5.8 Hz, 2 H), 3.26 (s, 3 H).

Step H: 4-(3-Bromo-4-fluorophenyl)-3-{4-[(2-liydroxyethyl)amino]-l,2,5-oxadiazol-3-yl}- 1 ,2,4-oxadiazol-5(4H)-one

Figure imgf000047_0001

4-(3-Bromo-4-fluorophenyl)-3-{4-[(2-methoxyetliyl)amino]-l,2,5-oxadiazol-3-yl}-l,2,4- oxadiazol-5(4H)-one (78.4 g, 0.196 mol) was dissolved in dichloromethane (600 mL). At -67 0C boron tribromide (37 mL, 0.392 mol) was added over 15 min. The reaction was warmed up to -10 0C in 30 min. LCMS indicated reaction completed. The reaction was stirred at room temperature for 1 hour. At 0 – 5 0C the reaction was slowly quenched with saturated sodium bicarbonate solution (1.5 L) over 30 min. The reaction temperature rose to 25 0C. The reaction was extracted with ethyl acetate (2 x 500 mL, first extraction organic layer is on the bottom and second extraction organic lager is on the top). The combined organic layers were dried over sodium sulfate and concentrated to give the desired product (75 g, 99%) as a crude brown solid. LCMS for C12H10BrFN5O4 (M+H)+: m/z = 386.0, 388.0. 1H NMR (400 MHz, DMSO-^6): δ 8.08 (dd, J= 6.2, 2.5 Hz, 1 H), 7.70 (m, 1 H), 7.68 (t, J= 8.7 Hz, 1 H), 6.33 (t, J= 5.6 Hz, 1 H), 4.85 (t, J= 5.0 Hz, 1 H), 3.56 (dd, J= 10.6, 5.6 Hz, 2 H), 3.29 (dd, J= 11.5, 5.9 Hz, 2 H).

Step I: 2-({4-[4-(3-Bromo-4-fluorophenyl)-5-oxo-4,5-dihydro-l,2,4-oxadiazol-3-yl]-l,2,5- oxadiazol-3-yl}amino)ethyl methanesulfonate

Figure imgf000047_0002

To a solution of 4-(3-bromo-4-fluorophenyl)-3-{4-[(2-hydroxyethyl)amino]-l,2,5-oxadiazol- 3-yl}-l,2,4-oxadiazol-5(4H)-one (1.5 kg, 3.9 mol, containing also some of the corresponding bromo-compound) in ethyl acetate (12 L) was added methanesulfonyl chloride (185 mL, 2.4 mol) dropwise over 1 h at room temperature. Triethylamine (325 mL, 2.3 mol) was added dropwise over 45 min, during which time the reaction temperature increased to 35 0C. After 2 h, the reaction mixture was washed with water (5 L), brine (I L), dried over sodium sulfate, combined with 3 more reactions of the same size, and the solvents removed in vacuo to afford the desired product (7600 g, quantitative yield) as a tan solid. LCMS for

Ci3HnBrFN5O6SNa (M+Na)+: m/z = 485.9, 487.9. 1H NMR (400 MHz, DMSCW6): δ 8.08 (dd, J= 6.2, 2.5 Hz, 1 H), 7.72 (m, 1 H), 7.58 (t, J= 8.7 Hz, 1 H), 6.75 (t, J- 5.9 Hz, 1 H), 4.36 (t, J= 5.3 Hz, 2 H), 3.58 (dd, J= 11.2, 5.6 Hz, 2 H), 3.18 (s, 3 H).

Step J: 3-{4-[(2-Azidoethyl)amino]-l,2,5-oxadiazol-3-yl}-4-(3-bromo-4-fluorophenyl)- 1 ,2,4-oxadiazol-5(4H)-one

Figure imgf000048_0001

To a solution of 2-({4-[4-(3-bromo-4-fluorophenyl)-5-oxo-4,5-dihydro-l,2,4-oxadiazol-3-yl]- l,2,5-oxadiazol-3-yl}amino)ethyl methanesulfonate (2.13 kg, 4.6 mol, containing also some of the corresponding bromo-compound) in dimethylformamide (4 L) stirring in a 22 L flask was added sodium azide (380 g, 5.84 mol). The reaction was heated at 500C for 6 h, poured into ice/water (8 L), and extracted with 1 : 1 ethyl acetate:heptane (20 L). The organic layer was washed with water (5 L) and brine (5 L), and the solvents removed in vacuo to afford the desired product (1464 g, 77%) as a tan solid. LCMS for C12H8BrFN8O3Na (M+Na)+: m/z =

433.0, 435.0. 1H NMR (400 MHz, DMSO-*/*): δ 8.08 (dd, J= 6.2, 2.5 Hz, 1 H), 7.72 (m, 1 H), 7.58 (t, J= 8.7 Hz, 1 H), 6.75 (t, J= 5.7 Hz, 1 H), 3.54 (t, J= 5.3 Hz, 2 H), 3.45 (dd, J= 11.1, 5.2 Hz, 2 H).

Step K: 3-{4-[(2-Aminoethyl)amino]-l,2,5-oxadiazol-3-yl}-4-(3-bromo-4-fluorophenyl)- 1 ,2,4-oxadiazol-5(4H)-one hydrochloride

Figure imgf000049_0001

Sodium iodide (1080 g, 7.2 mol) was added to 3-{4-[(2-azidoethyl)amino]-l,2,5-oxadiazol-3- yl}-4-(3-bromo-4-fluorophenyl)-l,2,4-oxadiazol-5(4H)-one (500 g, 1.22 mol) in methanol (6 L). The mixture was allowed to stir for 30 min during which time a mild exotherm was observed. Chlorotrimethylsilane (930 mL, 7.33 mol) was added as a solution in methanol (1 L) dropwise at a rate so that the temperature did not exceed 35 0C, and the reaction was allowed to stir for 3.5 h at ambient temperature. The reaction was neutralized with 33 wt% solution of sodium thiosulfate pentahydrate in water (~1.5 L), diluted with water (4 L), and the pΗ adjusted to 9 carefully with solid potassium carbonate (250 g – added in small portions: watch foaming). Di-fe/t-butyl dicarbonate (318 g, 1.45 mol) was added and the reaction was allowed to stir at room temperature. Additional potassium carbonate (200 g) was added in 50 g portions over 4 h to ensure that the pΗ was still at or above 9. After stirring at room temperature overnight, the solid was filtered, triturated with water (2 L), and then MTBE (1.5 L). A total of 11 runs were performed (5.5 kg, 13.38 mol). The combined solids were triturated with 1 : 1 TΗF:dichloromethane (24 L, 4 runs in a 20 L rotary evaporator flask, 50 0C, 1 h), filtered, and washed with dichloromethane (3 L each run) to afford an off- white solid. The crude material was dissolved at 55 0C tetrahydrofuran (5 mL/g), treated with decolorizing carbon (2 wt%) and silica gel (2 wt%), and filtered hot through celite to afford the product as an off-white solid (5122 g). The combined MTBE, THF, and dichloromethane filtrates were concentrated in vacuo and chromatographed (2 kg silica gel, heptane with a 0-100% ethyl acetate gradient, 30 L) to afford more product (262 g). The combined solids were dried to a constant weight in a convection oven (5385 g, 83%).

In a 22 L flask was charged hydrogen chloride (4 N solution in 1,4-dioxane, 4 L, 16 mol). fert-Butyl [2-({4-[4-(3-bromo-4-fluorophenyl)-5-oxo-4,5-dihydro-l,2,4-oxadiazol-3-yl]- l,2,5-oxadiazol-3-yl}amino)ethyl]carbamate (2315 g, 4.77 mol) was added as a solid in portions over 10 min. The slurry was stirred at room temperature and gradually became a thick paste that could not be stirred. After sitting overnight at room temperature, the paste was slurried in ethyl acetate (10 L), filtered, re-slurried in ethyl acetate (5 L), filtered, and dried to a constant weight to afford the desired product as a white solid (combined with other runs, 5 kg starting material charged, 4113 g, 95%). LCMS for C12HnBrFN6O3 (M+H)+: m/z

= 384.9, 386.9. 1H NMR (400 MHz, DMSO-J6): δ 8.12 (m, 4 H), 7.76 (m, 1 H), 7.58 (t, J= 8.7 Hz, 1 H), 6.78 (t, J= 6.1 Hz, 1 H), 3.51 (dd, J= 11.8, 6.1 Hz, 2 H), 3.02 (m, 2 H).

Step L: tert-Butyl ({[2-({4-[4-(3-bromo-4-fluorophenyl)-5-oxo-4,5-diliydro-l,2,4-oxadiazol- 3-yl]-l,2,5-oxadiazol-3-yl}amino)ethyl]amino}sulfonyl)carbamate

Figure imgf000050_0001

A 5 L round bottom flask was charged with chlorosulfonyl isocyanate [Aldrich, product #

142662] (149 mL, 1.72 mol) and dichloromethane (1.5 L) and cooled using an ice bath to 2 0C. tert-Butanol (162 mL, 1.73 mol) in dichloromethane (200 mL) was added dropwise at a rate so that the temperature did not exceed 10 0C. The resulting solution was stirred at room temperature for 30-60 min to provide tert-butyl [chlorosulfonyljcarbamate.

A 22 L flask was charged with 3-{4-[(2-aminoethyl)amino]-l,2,5-oxadiazol-3-yl}-4-(3- bromo-4-fluorophenyl)-l,2,4-oxadiazol-5(4H)-one hydrochloride (661 g, 1.57 mol) and 8.5 L dichloromethane. After cooling to -15 0C with an ice/salt bath, the solution of tert-butyl [chlorosulfonyl]carbamate (prepared as above) was added at a rate so that the temperature did not exceed -10 0C (addition time 7 min). After stirring for 10 min, triethylamine (1085 mL, 7.78 mol) was added at a rate so that the temperature did not exceed -5 0C (addition time 10 min). The cold bath was removed, the reaction was allowed to warm to 10 0C, split into two portions, and neutralized with 10% cone HCl (4.5 L each portion). Each portion was transferred to a 50 L separatory funnel and diluted with ethyl acetate to completely dissolve the white solid (~25 L). The layers were separated, and the organic layer was washed with water (5 L), brine (5 L), and the solvents removed in vacuo to afford an off-white solid. The solid was triturated with MTBE (2 x 1.5 L) and dried to a constant weight to afford a white solid. A total of 4113 g starting material was processed in this manner (5409 g, 98%). *Η NMR (400 MHz, OMSO-d6): δ 10.90 (s, 1 H), 8.08 (dd, J= 6.2, 2.5 Hz, 1 H), 7.72 (m, 1 H), 7.59 (t, J= 8.6 Hz, 1 H), 6.58 (t, J= 5.7 Hz, 1 H), 3.38 (dd, J= 12.7, 6.2 Hz, 2 H), 3.10 (dd, J = 12.1, 5.9 Hz, 2 H), 1.41 (s, 9 H). Step M: N-[2-({4-[4-(3-Bromo-4-fluorophenyl)-5-oxo-4,5-dmydro-l ,2,4-oxadiazol-3-yl]- l,2,5-oxadiazol-3-yl}amino)ethyl]sulfamide

Figure imgf000051_0001

To a 22 L flask containing 98:2 trifluoroacetic acid:water (8.9 L) was added tert-butyl ({[2- ({4-[4-(3-bromo-4-fluorophenyl)-5-oxo-4,5-diliydro-l,2,4-oxadiazol-3-yl]-l,2,5-oxadiazol-3- yl}amino)ethyl]amino}sulfonyl)carbamate (1931 g, 3.42 mol) in portions over 10 minutes. The resulting mixture was stirred at room temperature for 1.5 h, the solvents removed in vacuo, and chased with dichloromethane (2 L). The resulting solid was treated a second time with fresh 98:2 trifluoroacetic acid:water (8.9 L), heated for 1 h at 40-50 0C, the solvents removed in vacuo, and chased with dichloromethane (3 x 2 L). The resulting white solid was dried in a vacuum drying oven at 50 0C overnight. A total of 5409 g was processed in this manner (4990 g, quant, yield). LCMS for C]2H12BrFN7O5S (M+H)+: m/z = 463.9, 465.9.

1H NMR (400 MHz, OM$>O-d6): δ 8.08 (dd, J= 6.2, 2.5 Hz, 1 H), 7.72 (m, 1 H), 7.59 (t, J= 8.7 Hz, 1 H), 6.67 (t, J= 5.9 Hz, IH), 6.52 (t, J= 6.0 Hz, 1 H), 3.38 (dd, J= 12.7, 6.3 Hz, 2 H), 3.11 (dd, J= 12.3, 6.3 Hz).

Step N: 4-( {2-[(Aminosulfonyl)amino]ethyl} amino)-N-(3-bromo-4-fluorophenyl)-N- hydroxy-l,2,5-oxadiazole-3-carboximidamide

To a crude mixture of N-[2-({4-[4-(3-bromo-4-fluorophenyl)-5-oxo-4,5-dihydro-l,2,4- oxadiazol-3-yl]-l,2,5-oxadiazol-3-yl}amino)ethyl]sulfamide (2.4 mol) containing residual amounts of trifluoroacetic acid stirring in a 22 L flask was added THF (5 L). The resulting solution was cooled to 0 °C using an ice bath and 2 Ν NaOH (4 L) was added at a rate so that the temperature did not exceed 10 0C. After stirring at ambient temperature for 3 h (LCMS indicated no starting material remained), the pH was adjusted to 3-4 with concentrated HCl (-500 mL). The THF was removed in vacuo, and the resulting mixture was extracted with ethyl acetate (15 L). The organic layer was washed with water (5 L), brine (5 L), and the solvents removed in vacuo to afford a solid. The solid was triturated with MTBE (2 x 2 L), combined with three other reactions of the same size, and dried overnight in a convection oven to afford a white solid (3535 g). The solid was recrystallized (3 x 22 L flasks, 2: 1 water: ethanol, 14.1 L each flask) and dried in a 50 0C convection oven to a constant weight to furnish the title compound as an off-white solid (3290 g, 78%). LCMS for CnH14BrFN7O4S (M+H)+: m/z = 437.9, 439.9. 1H NMR (400 MHz, DMSO-J6): δ 11.51 (s, 1 H), 8.90 (s, 1 H), 7.17 (t, J= 8.8 Hz, 1 H), 7.11 (dd, J= 6.1, 2.7 Hz, 1 H), 6.76 (m, 1 H), 6.71 (t, J= 6.0 Hz, 1 H), 6.59 (s, 2 H), 6.23 (t, J= 6.1 Hz, 1 H), 3.35 (dd, J= 10.9, 7.0 Hz, 2 H), 3.10 (dd, J= 12.1, 6.2 Hz, 2 H).

The final product was an anhydrous crystalline solid. The water content was determined to be less than 0.1% by Karl Fischer titration.

 

 

CLIP

 

Print
INCB24360
Company:Incyte Corp.
Target: IDO1
Disease: Cancer

Incyte’s Andrew P. Combs presented the company’s clinical candidate for cancer immunotherapy. The basic tenet of this burgeoning field is that the human body’s immune system is a tremendous resource for fighting disease; scientists just need to figure out how to unleash it. One target that’s proven to be particularly attractive for this purpose in recent years is indoleamine-2,3-dioxygenase-1, or IDO1 (C&EN, April 6, page 10).

IDO1 plays a role in signaling the immune system to stand down from attacking foreign bodies it might otherwise go after, such as fetuses. Tumors also produce IDO1 to evade the immune system, so molecules that can inhibit this enzyme could bring the full force of the body’s defenses to bear on these deadly invaders.

Incyte’s search for an IDO1 inhibitor began with a high-throughput screen, which led to a proof-of-concept compound. But the compound had poor oral bioavailability. What’s more, the molecule and its analogs underwent glucuronidation during its metabolism: Enzymes tacked on a glucuronic acid group to the structure’s amidoxime, which was key to its activity.

The chemists reasoned they could block this metabolism by sterically hindering that position. Making such molecules proved to be more difficult than they expected. But then they unearthed a Latvian paper from 1993 that gave them the synthetic method they needed to make the series of compounds that would lead to their clinical candidate INCB24360 (epacadostat).

With its furazan core, as well as its amidoxime, bromide, and sulfuric diamide functional groups, INCB24360 is something of an odd duck, Combs acknowledged. “Some of you in the audience may be looking at this and saying, ‘That molecule does not look like something I would bring forward or maybe even make,’ ” he said, noting that the structure breaks many medicinal chemistry rules. “We’re a data-centric company, and we followed the data, not the rules,” Combs told C&EN.

The compound has completed Phase I clinical trials and is now being used in collaborative studies with several other pharmaceutical companies that combine INCB24360 with other cancer immunotherapy agents.

 

09338-scitech1-Incytecxd
TEAMWORK
Incyte’s team (from left): Andrew Combs, Dilip Modi, Joe Glenn, Brent Douty, Padmaja Polam, Brian Wayland, Rick Sparks, Wenyu Zhu, and Eddy Yue.
Credit: Incyte
WO2007113648A2 * Mar 26, 2007 Oct 11, 2007 Pfizer Products Inc. Ctla4 antibody combination therapy
US20070185165 * Dec 19, 2006 Aug 9, 2007 Combs Andrew P N-hydroxyamidinoheterocycles as modulators of indoleamine 2,3-dioxygenase
US20100055111 * Feb 14, 2008 Mar 4, 2010 Med. College Of Georgia Research Institute, Inc. Indoleamine 2,3-dioxygenase, pd-1/pd-l pathways, and ctla4 pathways in the activation of regulatory t cells
US20120058079 * Nov 11, 2011 Mar 8, 2012 Incyte Corporation, A Delaware Corporation 1,2,5-Oxadiazoles as Inhibitors of Indoleamine 2,3-Dioxygenase

REFERENCES

1: Vacchelli E, Aranda F, Eggermont A, Sautès-Fridman C, Tartour E, Kennedy EP, Platten M, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology. 2014 Dec 15;3(10):e957994. eCollection 2014 Nov. Review. PubMed PMID: 25941578; PubMed Central PMCID: PMC4292223.

2: Liu X, Shin N, Koblish HK, Yang G, Wang Q, Wang K, Leffet L, Hansbury MJ, Thomas B, Rupar M, Waeltz P, Bowman KJ, Polam P, Sparks RB, Yue EW, Li Y, Wynn R, Fridman JS, Burn TC, Combs AP, Newton RC, Scherle PA. Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity. Blood. 2010 Apr 29;115(17):3520-30. doi: 10.1182/blood-2009-09-246124. Epub 2010 Mar 2. PubMed PMID: 20197554.

3: Koblish HK, Hansbury MJ, Bowman KJ, Yang G, Neilan CL, Haley PJ, Burn TC, Waeltz P, Sparks RB, Yue EW, Combs AP, Scherle PA, Vaddi K, Fridman JS. Hydroxyamidine inhibitors of indoleamine-2,3-dioxygenase potently suppress systemic tryptophan catabolism and the growth of IDO-expressing tumors. Mol Cancer Ther. 2010 Feb;9(2):489-98. doi: 10.1158/1535-7163.MCT-09-0628. Epub 2010 Feb 2. PubMed PMID: 20124451.

//////////1204669-58-8 , INCB024360, INCB24360, epacadostat, PHASE 2, CANCER, orphan drug designation
Fc1ccc(cc1Br)N/C(=N\O)c2nonc2NCCNS(N)(=O)=O
Share

AMG 337

 phase 2, Uncategorized  Comments Off on AMG 337
Apr 182016
 

str1.

PIC CREDIT.BETHANY HALFORD

str1

 

Name: AMG-337(AMG337; AMG 337)
Cas 1173699-31-4
Formula: C23H22FN7O3
M.Wt: 463.46
Chemical Name: 6-[(1R)-1-[8-fluoro-6-(1-methylpyrazol-4-yl)-[1,2,4]triazolo[4,3-a]pyridin-3-yl]ethyl]-3-(2-methoxyethoxy)-5-methylidene-1,6-naphthyridine

(R)-6-(1-(8-fluoro-6-(1-methyl-1H-pyrazol-4-yl)-[1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)-1,6-naphthyridin-5(6H)-one

(R)-6-(1-(8-Fluoro-6-(1-methyl-1H-pyrazol-4-yl)-[1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)-1,6-naphthyridin-5(6H)-one

6-{ (lR)-l-[8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)[l,2,4]triazolo[4,3-a]pyridin-3-yl]ethyl}-3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one (“Compound M”),

PHASE 2 CANCER OF ESOPHAGUS

AMG-337 is a potent and highly selective small molecule ATP-competitive MET kinase inhibitor. AMG 337 inhibits MET kinase activity with an IC50 of < 5nM in enzymatic assays.
IC50 value: < 5nM [1]
Target: MET
in vitro: AMG-337 demonstrates exquisite selectivity for MET when profiled against a diverse panel of over 400 protein and lipid kinases in a competitive binding assay. In cellular assays, AMG 337 inhibits HGF-dependent MET phosphorylation with an IC50 of < 10 nM. [1] AMG 337 is a selective inhibitor of Met, which inhibits multiple mechanisms of Met activation. [2]
in vivo: AMG-337 demonstrates robust activity in MET-dependent cancer models. Oral administration of AMG 337 results in robust dose-dependent anti-tumor efficacy in MET amplified gastric cancer xenograft models, with inhibition of tumor growth consistent with the pharmacodynamic modulation of MET signaling

AMG 337 is a potent and highly selective small molecule ATP-competitive MET kinase inhibitor that demonstrates robust activity in MET-dependent cancer models. In enzymatic assays, AMG 337 inhibited MET kinase activity with an IC50 less than 5 nM. AMG 337 demonstrated exquisite selectivity for MET when profiled against a diverse panel of over 400 protein and lipid kinases in a competitive binding assay. In cellular assays, AMG 337 inhibited HGF-dependent MET phosphorylation with an IC50 of less than 10 nM [1].

AMG 337 was profiled in cell viability assays using a diverse panel of over 200 cancer cell lines where on treatment with AMG 337 affected the viability of only two gastric cancer cell lines (SNU-5 and Hs746T), both of which harbor amplification of the MET gene. The AMG 337 IC50 in the two sensitive cell lines was less than 50 nM, and greater than 10 µM in all other tested cell lines.

The receptor tyrosine kinase c-Met and its natural ligand, hepatocyte growth factor (HGF), are involved in cell proliferation, migration, and invasion and are essential for normal embryonic development. Deregulation of c-Met/HGF signaling can lead to tumorigenesis and metastasis and has been implicated in a variety of cancers. Several mechanisms lead to deregulation, including overexpression of c-Met and/or HGF, amplification of the MET gene, or activating mutations of c-Met, all of which have been found in human cancers.

AMG 337 is a potent and highly selective inhibitor of wild-type and some mutant forms of MET. In a competitive binding assay conducted on 402 human kinases, AMG 337 bound only to MET. In a cell viability study, the only cell lines that responded to an AMG 337 analog were gastric cancer cells harboring MET gene amplification. None of the other cell lines were sensitive to the AMG 337 analog and none harbored MET gene amplification. In secondary pharmacology assays with transporters, enzymes, ion channels, and receptors, binding to the adenosine transporter was the only activity inhibited.

In vivo, oral administration of AMG 337 resulted in robust dose-dependent anti-tumor efficacy in MET amplified gastric cancer xenograft models, with inhibition of tumor growth consistent with the pharmacodynamic modulation of MET signaling. Further studies in an expanded panel of additional cancer cell lines derived from gastric, NSCLC, and esophageal cancer confirmed that the in-vitro anti-proliferative activity of AMG 337 correlated with amplification of MET. In those cell lines, treatment with AMG 337 inhibited downstream PI3K and MAPK signaling pathways, which translated into growth arrest as evidenced by an accumulation of cells in the G1 phase of the cell cycle, a concomitant reduction in DNA synthesis, and the induction of apoptosis [1].

In a small subset of patients with MET-amplified gastrointestinal (GI) tumors, monotherapy with the investigational agent AMG 337 produced a “dramatic” response. Of the 13 patients with MET-amplified gastric and esophageal cancers, eight experienced a response. The overall response rate in this group of patients was 62%. Response was rapid, with time to response being 4 weeks in most cases. Patients achieved tumor shrinkage and symptomatic improvement. One patient achieved a complete response and is still on treatment at 155 weeks; the others achieved partial responses or stable disease. This has led to further trials, including Phase II trials MET amplified gastric/esophageal adenocarcinoma or other solid tumors.

PAPER

Discovery of (R)-6-(1-(8-Fluoro-6-(1-methyl-1H-pyrazol-4-yl)-[1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)-1,6-naphthyridin-5(6H)-one (AMG 337), a Potent and Selective Inhibitor of MET with High Unbound Target Coverage and Robust In Vivo Antitumor Activity.

Boezio, A.A.Copeland, K.W.Rex, K.K Albrecht, B.Bauer, D.Bellon, S.F.Boezio, C.Broome, M.A.Choquette, D.Coxon, A.Dussault, I.Hirai, S.Lewis, R.Lin, M.H.Lohman, J.Liu, J.Peterson, E.A.Potashman, M.Shimanovich, R.Teffera, Y.Whittington, D.A.Vaida, K.R.Harmange, J.C.

(2016) J.Med.Chem. 59: 2328-2342

http://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.5b01716

Abstract Image

Deregulation of the receptor tyrosine kinase mesenchymal epithelial transition factor (MET) has been implicated in several human cancers and is an attractive target for small molecule drug discovery. Herein, we report the discovery of compound 23 (AMG 337), which demonstrates nanomolar inhibition of MET kinase activity, desirable preclinical pharmacokinetics, significant inhibition of MET phosphorylation in mice, and robust tumor growth inhibition in a MET-dependent mouse efficacy model.

(R)-6-(1-(8-Fluoro-6-(1-methyl-1H-pyrazol-4-yl)-[1,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)-1,6-naphthyridin-5(6H)-one (23)

Step 1: Coupling 9c and 13c in MeCN for 30 min at room temperature resulted in 86% yield. LRMS (ESI): m/z (M + H) 482.2. Step 2: THF for 50 min at room temperature resulted in 48% yield. The racemate was purified by supercritical fluid chromatography (SFC) by repeating 0.75 mL injections of a 30 mg/mL solution onto a Chiralpak AS-H, 2 cm × 15 cm (i.d. × length) column, eluting with 20% i-PrOH and 80% CO2 at a flow rate of 50 mL/min to provide 120 mg peak 1 (23) with >99% ee and 150 mg of peak 2 (ent-23) with >99% ee.(29) 1H NMR (400 MHz, Chloroform-d): δ 8.72 (d, J = 2.93 Hz, 1H), 8.31 (d, J = 0.78 Hz, 1H), 8.15 (d, J = 2.84 Hz, 1H), 7.72 (s, 1H), 7.61 (s, 1H), 7.42 (d, J = 7.82 Hz, 1H), 7.09 (dd, J = 0.73, 10.61 Hz, 1H), 7.05 (q, J= 7.00 Hz, 1H), 6.82 (d, J = 7.82 Hz, 1H), 4.26–4.37 (m, 2H), 3.97 (s, 3H), 3.80–3.88 (m, J = 3.80, 5.10 Hz, 2H), 3.49 (s, 3H), 2.15 (d, J = 7.14 Hz, 3H). HRMS (ESI): m/z (M + H) calcd, 464.1859; found, 464.1841. The solid was recrystallized in EtOH followed by the addition of H2O to form crystalline free base monohydrate form I with a dehydration event at 40–55 °C followed by a melt at 151–153 °C. The solid could also be recrystallized in EtOH under anhydrous conditions to form crystalline anhydrous free base form I with a melting point of 151–153 °C.

PATENT

WO 2009091374

http://www.google.com/patents/WO2009091374A2?cl=en

Example 515

(SV6-(l-f8-fluoro-6-(3-methvIisoxazol-5-vn-|l,2,41triazoIo[4,3-a1pyridin-3-vncthvn-3-(f2- methoxyethoxy)methv.)-l,6-naphthyridin-5(6HVone Synthesized in the same general manner as that previously described for example 509 using General Method N. Chiral separation by preparative SFC (Chiralpak® AD-H (20 x 150 mm, 5Dm), 25% MeOH, 75% CO2, 0.2% DEA; 100 bar system pressure; 75 mL/min; tr 4.75min). On the basis of previous crystallographic data and potency recorded for related compound in the same program, the absolute stereochemistry has been assigned to be the S enantiomer. M/Z – 465.2 [M+H], calc 464.16 for C23H2iFN6O4

Figure imgf000165_0002

Example 516 ri?)-6-ri-(8-fluoro-6-(l-methyl-lH-pyrazol-4-vn-H.2.41triazolo[4,3-alpyridin-3-yl)ethyl)- 3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one The title compound was synthesized using General Method N. Chiral separation by preparative SFC (Chiralpak® AS-H (20 x 150 mm, 5 Dm), 20% iPrOH, 80% CO2; 100 bar system pressure, 50 mL/min; tr 1.67 min). On the basis of previous crystallographic data and potency recorded for related compound in the same program, the absolute stereochemistry has been assigned to be the R enantiomer. M/Z = 464.2 [M+H], calc 463.18 for C23H22FN7O3. 1H NMR (400 MHz, CHLOROFORM-^ D ppm 2.15 (d, J=7.14 Hz, 3 H) 3.49 (s, 3 H) 3.80 – 3.90 (m, 2 H) 3.97 (s, 3 H) 4.27 – 4.39 (m, 2 H) 6.83 (d, J=7.73 Hz, 1 H) 7.00 – 7.13 (m, 2 H) 7.42 (d, J=7.82 Hz, 1 H) 7.61 (s, 1 H) 7.72 (s, 1 H) 8.15 (d, J=2.84 Hz, 1 H) 8.31 (s, 1 H) 8.72 (d, J=3.03 Hz, 1 H).

Figure imgf000166_0001
PATENT
WO 2015161152

6-{ (lR)-l-[8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)[l,2,4]triazolo[4,3-a]pyridin-3-yl]ethyl}-3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one (“Compound M”), which is a selective inhibitor of the c-Met receptor, and useful in the treatment, prevention, or amelioration of cancer:

PATENT

https://www.google.com/patents/WO2014210042A2?cl=en

The overall scheme for the preparation of Compound A is shown below. The optical purity of Compound A is controlled during the synthetic process by both the quality of the incoming starting materials and the specific reagents used for the transformations. Chiral purity is preserved during both the coupling reaction (the second step) and the dehydration reaction (the third step).

NAPH (S)-halopropionic NAPA

acid/ester

PREPARATION OF COMPOUND A

In one aspect, provided herein is a method for preparing Compound A, salts of Compound A, and the monohydrate form of Compound A. Compound A can be prepared from the NAPH, PYRH, and S-propionic acid/ester starting materials in three steps. First, NAPH and ^-propionic acid/ester undergo an S 2 alkylation reaction to result in (R)-2-(3-(2-methoxyethoxy)-5-oxo-l,6-naphthyridin-6(5H)-yl)propanoic acid/ester. The ^-propionic acid starting material produces (R)-2-(3-(2-methoxyethoxy)-5-oxo-l,6-naphthyridin-6(5H)-yl)propanoic acid (“NAPA”) in one step. The ^-propionic ester starting material first produces the ester analog of NAPA, and is subsequently hydrolyzed to form NAPA. During workup, the acid can optionally form a salt (e.g., HC1 or 2-naphthalenesulfonic acid).

Step 1:

NAPH (S)-2-halopropionic

acid/ester

1 2

wherein R is Br, CI, I, or OTf; and R is COOH or Ci-salkyl ester, and

when R is Ci^alkyl ester, the method of forming the NAPA or salt thereof further comprises hydrolyzing the Ci-salkyl ester to form an acid.

Second, NAPA and PYRH are coupled together to form (R)-N’-(3-fluoro-5-(lmethyl-lH-pyrazol-4-yl)pyridin-2-yl)-2-(3-(2-methoxyethoxy)-5-oxo- l,6-naphthyridin- 6(5H)yl)propanehydrazide (“HYDZ”).

Step 2:

Third, HYDZ is dehydrated to form Compound A.

The free base form of Compound A can be crystallized as a salt or a monohydrate.

Step 1: Alkylation of NAPH to form NAPA

The first step in the preparation of Compound A is the alkylation of NAPH to form NAPA. The NAPA product of the alkylation reaction is produced as a free base and is advantageously stable.

Thus, one aspect of the disclosure provides a method for preparing NAPA comprising admixing 3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one (“NAPH”):

Me

1 R2 , and a base, under conditions sufficient to form NAPA:

wherein R1 is Br, CI, I, or OTf; and

R2 is COOH or C^alkyl ester;

and when R2 is Ci_3alkyl ester, the method of forming the NAPA or salt thereof further comprises hydrolyzing the Ci-3alkyl ester to form an acid.

Me

The compound, R1 R2 , represents an (^-propionic acid and/or (S)- propionic ester

Me

(“(S)-propionic acid/ester”). When R1 R2 is an acid (i.e., R2 is COOH), NAPA is formed in one step:

-prop on c ac

Me

When R1 R2 is an ester (i.e., R2 is C1-3 alkyl ester), then the NAPA ester analog is formed, which can be hydrolyzed to form NAPA.

The SN2 alkylation of NAPH to form NAPA occurs with an inversion of

EXAMPLE 1

SYNTHESIS OF (R)-2-(3-(2-METHOXYETHOXY)-5-OXO-l,6-NAPHTHYRIDIN-6(5H)- YL)PROPANOIC ACID NAPHTHALENE-2-SULFONATE (NAPA)

Scheme 1: Synthesis of naphthyridinone acid 2-napsylate (NAPA)

NAPA was synthesized according to Scheme 1 by the following procedure. A jacket reactor (60 L) was charged with 3000 g (1.0 equivalent) of 3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one and 4646 g (2.0 equivalents) of magnesium ie/t-butoxide. 12 L (4.0 Vol) tetrahydrofuran was added to the reactor and an N2sweep and stirring were initiated. 2213 g (1.5 equivalents) of S-2-bromopropionic acid was added over at least 30 min, controlling the addition such that the batch temperature did not rise above 30 °C. The charge port was rinsed with tetrahydrofuran (0.5 Vol) after addition. The batch was then aged for at least 5 min at 25 °C. 1600 g (1.05 equivalents) of potassium iert-butoxide was added to the reactor in four portions (approximately equal) such that the batch temperature did not rise above 30 °C. The charge port was again rinsed with tetrahydrofuran (1.5 L, 0.5 Vol). The batch temperature was adjusted to 35+5 °C and the batch was aged for at least 12 h.

A separate 100 L reactor was charged with 6 L of 2-Metetrahydrofuran (2-MeTHF) (2.0 Vol), 8.4 L of water (1.5 Vol) and 9.08 L (4.0 equivalents) of 6 N HC1. The mixture from the 60 L reactor was pumped into the 100 L reactor, while maintaining the batch temperature at less than 45 °C.

The batch temperature was then adjusted to 20+5°C. The pH of the batch was adjusted with 6N HC1 (or 2N NaOH) solution until the pH was 1.4 to 1.9. The aqueous layer was separated from the product-containing organic layer. The aqueous layer was extracted with 2-MeTHF (2 Vol), and the 2-MeTHF was combined with the product stream in the reactor. The combined organic stream was washed with 20% brine (1 Vol). The organic layer was polish-filtered through a < ΙΟμιη filter into a clean vessel.

In a separate vessel, 1.1 equivalents of 2-Naphthalenesulfonic acid hydrate was dissolved in THF (2 Vol). The solution was polish-filtered prior to use. The 2-naphthalenesulfonic acid hydrate THF solution was added into the product organic solution in the vessel over at least 2 h at 25+5 °C. The batch temperature was adjusted to 60+5 °C and the batch was aged for 1+0.5 h. The batch temperature was adjusted to 20+5 °C over at least 2 h. The batch was filtered to collect the product. The collected filter cake was washed with THF (5.0 Vol) by displacement. The product cake was dried on a frit under vacuum/nitrogen stream until the water content was < lwt% by LOD.

The yield of the product (R)-2-(3-(2-methoxyethoxy)-5-oxo- l,6-naphthyridin-6(5H)-yl)propanoic acid naphthalene-2-sulfonate, was 87%. The chiral purity was determined using chiral HPLC and was found to be 98-99% ee. The purity was determined using HPLC, and was found to be > 98%.

Thus, Example 1 shows the synthesis of NAPA according to the disclosure.

EXAMPLE 2

SYNTHESIS OF (R)-N’-(3-FLUORO-5-(l-METHYL-lH-PYRAZOL-4-YL)PYRIDIN-2- YL)-2-(3-(2-METHOXYETHOXY)-5-OXO-l,6-NAPHTHYRIDIN-6(5H)- YL)PROPANEHYDRAZIDE (HYDZ)

Scheme 2: Synthesis of (R)-N’-(3-fluoro-5-(l-methyl- lH-pyrazol-4-yl)pyridin-2-yl)-2-(3-(2-methoxyethoxy)-5-oxo-l,6-naphthyridin-6(5H)-yl)propanehydrazide

HYDZ was synthesized according to Scheme 2 by the following procedure. A 60 L jacket reactor was charged with 2805.0 g (1.0 equivalent) of (R)-2-(3-(2-methoxyethoxy)-5-oxo-l,6-naphthyridin-6(5H)-yl)propanoic acid 2-napsylate (NAPA) and N,N-dimethylacetamide (DMAC) (4.6 mL DMAC per gram of NAPA). Stirring and an N2 sweep were initiated. 1.05 equivalents of N,N-diisopropylethylamine (DIPEA) was added while maintaining the batch temperature at less than 35°C. Initially the NAPA dissolves. A white precipitate formed while aging, but the precipitate had no impact on the reaction performance. 2197 g (1.10 equivalents) of 3-fluoro-2-hydrazinyl-5-(l-methyl- lH-pyrazol-4-yl)pyridine (PYRH) was added to the batch. The batch temperature was adjusted to 10+5 °C. 2208 g (1.2 equivalents) of N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (EDC) was added in four portions (approximately equal) over at least 1 h (about 20 min interval per portion) at 10+5 °C.

The batch was aged until the amide conversion target was met. If the amide conversion target was not reached within 2 h, additional EDC was added until the conversion target was met. Once the target was met, the batch was heated to 55 °C until the solution was homogeneous. The batch was filtered through a <20 μ in-line filter into a reactor. The vessel and filter were rinsed with DMAC (0.2 mL DMAC/g of NAPA). The batch temperature was adjusted to 45+5 °C.

The reactor was charged with a seed slurry of (R)-N’-(3-fluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridin-2-yl)-2-(3-(2-methoxyethoxy)-5-oxo-l,6-naphthyridin-6(5H)-yl)propanehydrazide (HYDZ) (0.01 equivalents) in water (0.3 mL/g).

The batch was aged at 50+5 °C for at least 30 min. The batch temperature was adjusted to 20+5°C over at least 2 h. The batch was aged at 20+5°C for at least 30 min. 2.90 mL water per g was added at 25+5 °C over at least 2 h. The batch was aged at 20+5 °C for at least 1 h. The batch slurry was filtered to collect the product. The product was washed with 30% DMAC/H20 (0.5 Vol) by displacement. The product cake was washed with water (3 Vol) by displacement. The product cake was dried on the frit under vacuum/nitrogen stream until the water content was < 0.2 wt% as determined by Karl Fischer titration (KF). The product was a white, crystalline solid. The yield was about 83-84%. The ee was measured by HPLC and was found to be > 99.8%ee. The purity was determined by HPLC and was found to be >99.8 LCAP (purity by LC area percentage).

Thus, Example 2 demonstrates the synthesis of HYDZ according to the disclosure.

EXAMPLE 3

SYNTHESIS OF (R)-6-(l-(8-FLUORO-6-(l-METHYL-lH-PYRAZOL-4-YL)- [l,2,4]TRIAZOLO[4,3-A]PYRIDIN-3-YL)ETHYL)-3-(2-METHOXYETHOXY)-l,6- NAPHTHYRIDIN-5(6H)-ONE HYDROCHLORIDE SALT (COMPOUND A-HCL) – ROUTE 1

Scheme 3 Route 1 – Synthesis of (R)-6-(l-(8-fluoro-6-(l-methyl- lH-pyrazol-4-yl)- [l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)- l,6-naphthyridin-5(6H)-one hydrochloride

(R)-6-(l-(8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)- l,6-naphthyridin-5(6H)-one hydrochloride salt (Compound A-

HC1) was synthesized according to Scheme 3, Route 1 by the following procedure. A 15 L reactor, Reactor 1, was charged with 750 g HYDZ and the reactor jacket temperature was adjusted to 20+5 °C. A nitrogen sweep was initiated in Reactor 1 and the condenser coolant (at 5+5 °C) was started. Acetonitrile (3.4 L, 4.5 Vol) was added to Reactor 1 and stirring was initiated. 420 g (2.5 equivalents) of 2,6-lutidine was added to the reactor.

A solution of diphenylphosphinyl chloride Ph2P(0)(Cl) was prepared by combining 850 g (2.3 equivalents) of Ph2P(0)(Cl) and 300 g acetonitrile in an appropriate container. The contents of the PH2P(0)(C1) solution were added to Reactor 1. The jacket temperature was adjusted over 60+30 min until the reflux temperature of the batch (approximately 85 °C) was reached. The reaction was stirred for 14+6 h. The batch temperature was reduced to 75+5 °C and the batch was sampled for IPT analysis. The expected result was < 2% HYDZ remaining. If the target was not met, the heating at reflux temperature was continued for 9+6 h. Sampling, analysis, and heating was repeated until a satisfactory conversion assay result was obtained (< 10% HYDZ was considered satisfactory, < 1% was actually achieved). The final sample was assayed for optical purity by HPLC, and was found to be > 99.5% ee.

A K2CO3/KCI quench solution (5.0 Vol) was prepared in advance by combining 555 g (3.1 equivalents) of potassium carbonate with 335 g (2.9 equivalents) of potassium chloride and 3450 g of water in an appropriate container. The quench solution was added to Reactor 1 over at least 15 min, maintaining the batch temperature at 60+5 °C. As the aqueous base reacted with excess acid some bubbling (C02) occurred. 3.0 L (4.0 Vol) of toluene was added to Reactor 1 at 65+5 °C. A sample of the batch was taken for IPT analysis. The lower (aqueous) phase of the sample was assayed by pH probe (glass electrode). The pH was acceptable if in the range of pH 8-11. The upper (organic) phase of the sample was assayed by HPLC.

The batch was agitated for 20+10 min at 65+5 °C. Stirring was stopped and the suspension was allowed to settle for at least 20 min. The aqueous phase was drained from Reactor 1 via a closed transfer into an appropriate inerted container. The remaining organic phase was drained from Reactor 1 via a closed transfer to an appropriate inerted container. The aqueous phase was transferred back into Reactor 1.

An aqueous cut wash was prepared in advance by combining 2.3 L (3.0 Vol) acetonitrile and 2.3 L(3.0 Vol) toluene in an appropriate container. The aqueous cut wash was added to Reactor 1. The batch was agitated for 20+10 min at 65+5 °C. The stirring was stopped and the suspension was allowed to settle for at least 20 min. The lower (aqueous) phase was drained from Reactor 1 via a closed transfer into an appropriate inerted container. The organic phase was drained from Reactor 1 via a closed transfer to the inerted container containing the first organic cut. The combined mass of the two organic cuts was measured and the organic cuts were transferred back to Reactor 1. Agitation was initiated and the batch temperature was adjusted to 60+10 °C. A sample of the batch was taken and tested for Compound A content by HPLC. The contents of Reactor 1 were distilled under vacuum (about 300-450 mmHg) to approximately 8 volumes while maintaining a batch temperature of 60+10 °C and a jacket temperature of less than 85 °C. The final volume was between 8 and 12 volumes.

The nitrogen sweep in Reactor 1 was resumed and the batch temperature adjusted to 70+5 °C. A sample of the batch was taken to determine the toluene content by GC. If the result was not within 0-10% area, the distillation was continued and concomitantly an equal volume of 2-propanol, up to 5 volumes, was added to maintain constant batch volume. Sampling, analysis, and distillation was repeated until the toluene content was within the 0-10% area window. After the distillation was complete, 540 g (450 mL, 3.5 equivalents) of hydrochloric acid was added to Reactor 1 over 45+15 min while maintaining a batch temperature at 75+5 °C.

A Compound A-HC1 seed suspension was prepared in advance by combining 7.5 g of Compound A-HC1 and 380 mL (0.5 Vol) of 3 propanol in an appropriate container. The seed suspension was added to Reactor 1 at 75+5 °C. The batch was agitated for 60+30 min at 75+5 °C. The batch was cooled to 20+5 °C over 3+1 h. The batch was agitated for 30+15 min at 20+5 °C. 2.6 L (3.5 Vol) of heptane was added to the batch over 2+1 h. The batch was then agitated for 60+30 min at 20+5 °C. A sample of the batch was taken and filtered for IPT analysis. The filtrate was assayed for Compound A-HC1. If the amount of Compound A-HC1 in the filtrate was greater than 5.0 mg/mL the batch was held at 20 °C for at least 4 h prior to filtration. If the amount of Compound A-HC1 in the filtrate was in the range of 2-5 mg.ML, the contents of Reactor 1 were filtered through a < 25 μιη PTFE or PP filter cloth, sending the filtrate to an appropriate container.

A first cake wash was prepared in advance by combining 1.5L (2.0 Vol) of 2-propanol and 1.5L (2.0 Vol) of heptane in an appropriate container. The first cake wash was added to Reactor 1 and the contents were agitated for approximately 5 min at 20+5 °C. The contents of Reactor 1 were transferred to the cake and filter. A second cake wash of 3.0L (4.0 Vol) of heptane was added to Reactor 1 and the contents were agitated for approximately 5 min at 20+5 °C. The contents of Reactor 1 were transferred to the cake and filter. The wet cake was dried under a flow of nitrogen and vacuum until the heptane content was less than 0.5 wt% as determined by GC. The dried yield was 701g, 85% as a yellow powder. The dried material was assayed for chemical purity and potency by HPLC and for residual solvent content by GC. The isolated product was 88.8% Compound A-HC1, having 99.8% ee and 0.6% water.

Thus, Example 3 shows the synthesis of Compound A-HCL according to the disclosure.

EXAMPLE 4

SYNTHESIS OF (R)-6-(l-(8-FLUORO-6-(l-METHYL-lH-PYRAZOL-4-YL)- [l,2,4]TRIAZOLO[4,3-A]PYRIDIN-3-YL)ETHYL)-3-(2-METHOXYETHOXY)-l,6- NAPHTHYRIDIN-5(6H)-ONE HYDROCHLORIDE SALT (COMPOUND A-HCL) – ROUTE 2

HYDZ A HCI

Scheme 4: Route 2 – Synthesis of (R)-6-(l-(8-fluoro-6-(l-methyl- lH-pyrazol-4-yl)- [l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)- l,6-naphthyridin-5(6H)-one hydrochloride

(R)-6-(l-(8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)- l,6-naphthyridin-5(6H)-one hydrochloride salt was synthesized according to Scheme 4, Route 2, by the following procedure. A clean and dry 60 L reactor was fitted with a reflux condenser, nitrogen inlet, and vented to a scrubber (Reactor 1). The jacket temperature of Reactor 1 was set to 20 °C. A scrubber was set up to the vent of Reactor 1, and aqueous bleach solution was charged to the scrubber. The circulating pump (commercial 5.25% NaOCl) was initiated. The scrubber pump was turned on and N2 sweep on Reactor 1 was started. Reactor 1 was charged with 2597 g (0.52 equivalents) of Lawesson’s reagent. Reactor 1 was then charged with 6000 g (1.0 equivalent) of HYDZ and 30 L (5.0 vol) acetonitrile (MeCN). Agitation of Reactor 1 was initiated. The reactor was heated to 50+5 °C and aged until an LC assay showed consumption of HYDZ (> 99% conversion).

The jacket temperature of a second clean and dry reactor, Reactor 2, was set to 50 °C. The contents of Reactor 1 were transferred to Reactor 2 through a 5 micron inline filter. Reactor 1 was rinsed with MeCN, and the rinse was transferred through the inline filter to Reactor 2. Reactor 2 was charged with toluene. (31.7 Kg)

In a separate container a solution of 16.7% K2C03 was prepared by adding 7200 g K2C03 and 36 L water to the container and shaking the container well until all the solid was dissolved. Half of the contents of the K2C03 solution was added to Reactor 2 over at least 10 min. The batch temperature of Reactor 2 was adjusted to 50+5 °C. The batch in Reactor 2 was agitated at 50+5 °C for at least 1 h. The agitation was stopped and the batch in Reactor 2 was allowed to phase separate. The aqueous phase was removed. The remaining contents of the K2C03 solution was added to Reactor 2 over at least 10 min. The batch temperature in Reactor 2 was adjusted to 50+5 °C. The batch in Reactor 2 was agitated at 50+5 °C for at least 1 h. The agitation was stopped and the batch in Reactor 2 was allowed to phase separate. The aqueous phase was removed.

The jacket temperature of a clean and dry reactor, Reactor 3, was set to 50 °C. The contents of Reactor 2 were transferred to Reactor 3 through a 5 micron in-line filter. The contents of Reactor 3 were distilled at reduced pressure. Isopropyl alcohol (IP A, 23.9 kg) was charged to Reactor 3 and then the batch was distilled down. IPA (23.2 kg) was again added to Reactor 3. The charge/distillation/charge cycle was repeated. The batch temperature in Reactor 3 was adjusted to 70+15 °C. Reactor 3 was then charged with DI water (1.8 L). Concentrated HC1 (1015 mL) was added to Reactor 3 over at least 15 min at 70+15 °C.

A seed of the Compound A-HCl was prepared by combining a seed and IPA in a separate container. The Compound A-HCl seed was added to Reactor 3 as a slurry. The batch in Reactor 3 was aged at 70+15 °C for at least 15 min to ensure that the seed held. The batch in Reactor 3 was cooled to 20+5 °C over at least 1 h. Heptane (24.5 kg) was added to Reactor 3 at 20+5 °C over at least 1 h. The batch was aged at 20+5 °C for at least 15 min. The contents of Reactor 3 were filtered through an Aurora filter fitted with a <25 μιη PTFE or PP filter cloth. The mother liquor was used to rinse Reactor 3.

A 50% v/v IP A/heptane solution was prepared, in advance, in a separate container by adding the IPA and heptane to the container and shaking. The filter cake from Reactor 3 was washed with the 50% IP A/heptane solution. If needed, the IP A/heptane mixture, or heptane alone, can be added to Reactor 3 prior to filtering the contents through the Aurora filter. The cake was washed with heptane. The cake was dried under nitrogen and vacuum until there was about < 0.5 wt% heptane by GC analysis. The product was analyzed for purity and wt% assay by achiral HPLC, for wt% by QNMR, for water content by KF, for form by XRD, for chiral purity by chiral HPLC, and for K and P content by ICP elemental analysis.

Compound A-HCl had a purity of 99.56 area% and 88.3 wt% assay by achiral HPLC, and 89.9 wt% by QNMR. The water content was 0.99 wt% as determined by KF. The chiral purity was 99.9%ee as determined by chiral HPLC. The P and K content was found to be 171 ppm and 1356 ppm, respectively, as determined by ICP elemental analysis.

Thus, Example 4 shows the synthesis of Compound A-HCl according to the disclosure.

EXAMPLE 5

SYNTHESIS OF (R)-6-(l-(8-FLUORO-6-(l-METHYL-lH-PYRAZOL-4-YL)- [l,2,4]TRIAZOLO[4,3-A]PYRIDIN-3-YL)ETHYL)-3-(2-METHOXYETHOXY)-l,6- NAPHTHYRIDIN-5(6H)-ONE (COMPOUND A) – ROUTE 3

Scheme 5: Route 3 – Synthesis of (R)-6-(l-(8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)- l,6-naphthyridin-5(6H)-one (compound A)

(R)-6-(l-(8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)- l,6-naphthyridin-5(6H)-one was synthesized according to Scheme 5, Route 3, by the following procedure. 0.760 g (1.6 mmol) N’-iS-fluoro-S-il-methyl-lH-pyrazol-4-yl)pyridin-2-yl)-2-(3-(2-methoxyethoxy)-5-oxo- l,6-naphthyridin-6(5H)-yl)propanehydrazide (HYDZ) and 0.62 g (2.4 mmol) triphenylphosphine were taken up in 16 mL THF. 0.31 mL (2.4 mmol) trimethylsilyl (TMS)-azide was added, followed by addition of 0.37 mL (2.4 mmol) DEAD, maintaining the reaction temperature below 33 °C. The reaction was stirred at room temperature for 50 minutes. The reaction mixture was concentrated in vacuo.

The crude material was taken up in dichloromethane and loaded onto silica gel. The crude material was purified via medium pressure liquid chromatography using a 90: 10: 1 DCM : MeOH : NH4OH solvent system. 350 mg, (48% yield) of (R)-6-(l-(8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one was collected as a tan solid. The (S) isomer was also collected. The product had a purity of 97% by HPLC.

Thus, Example 5 shows the synthesis of enantiomerically pure Compound A according to the disclosure.

EXAMPLE 6

SYNTHESIS OF (R)-6-(l-(8-FLUORO-6-(l-METHYL-lH-PYRAZOL-4-YL)- [l,2,4]TRIAZOLO[4,3-A]PYRIDIN-3-YL)ETHYL)-3-(2-METHOXYETHOXY)-l,6-NAPHTHYRIDIN-5(6H)-ONE (COMPOUND A) AND THE HYDROCHLORIDE SALT- ROUTE 3

Scheme 6: Route 3 – Synthesis of (R)-6-(l-(8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one (compound A) and the hydrochloride salt

(R)-6-(l-(8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one was synthesized according to Scheme 6, Route 3, by the following procedure. Benzothiazyl disulfide (3.31 g, 9.97 mmol), HYDZ (4.0 g, 8.31 mmol), and a stir bar were added to a 50 mL 3-neck flask fitted with a reflux condenser topped with a nitrogen inlet, a thermocouple and a septum. The flask headspace was purged with nitrogen, and the solids were suspended in MeCN (20.00 mL, 5 mL/g) at ambient conditions. The flask contents were heated to 50 °C on a heating mantle. Finally,

trimethylphosphine, solution in THF (9.97 ml, 9.97 mmol) was added dropwise by syringe pump with stirring over 1 h. An ice pack was affixed to the side of the flask in lieu of a reflux condenser. After about 0.5 h from addition, the resulting suspension was sampled and analyzed by, showing about 99% conversion of penultimate, and about 94% Compound A vs.

benzothiazole-2-thiol (“BtSH”) adduct selectivity.

After about 0.75 h from addition, the yellow reaction mixture was cooled to 0 °C in an ice bath, and 30% hydrogen peroxide in water (2.037 mL, 19.94 mmol) was added dropwise over 2 hours. The reaction solution was allowed to warm to room temperature overnight.

The suspension was heated to 30 °C, held at that temperature for 3 h and then cooled to room temperature. After cooling was complete, an aliquot was filtered and the filtrate was analyzed by liquid chromatography, showing 99% Compound A vs. BtSH adduct (91% purity for Compound A overall).

A Celite filtration pad about 0.5″ thick was set up on a 50 mL disposable filter frit and wetted with toluene (32.0 mL, 8 mL/g). The reaction suspension was transferred to the Celite pad and filtered to remove BtSH-related byproducts, washing with MeCN (2.000 mL, 0.5 mL/g). The filtrate was transferred to a 100 mL round bottom flask, and treated with 30 mL (7.5 Vol) of an aqueous quench solution consisting of sodium bicarbonate (7.5 ml, 8.93 mmol) and sodium thiosulfate (3.75 ml, 4.74 mmol) at overall about 5 wt% salt. The suspension was stirred for about 15 min and then the layers were allowed to separate. Once the layers were cut, the aqueous waste stream was analyzed by LC, showing 8% loss. The organic stream was similarly analyzed, showing 71% assay yield, implying about 20% loss to waste cake.

The organic cut was transferred to a 3-neck 50 mL round bottom flask with magnetic stir bar, thermocouple, and a shortpath distillation head with an ice-cooled receiving flask. The boiling flask contents were distilled at 55 °C and 300 torr pressure. The volume was reduced to 17 mL. The distillation was continued at constant volume with concomitant infusion of IPA (about 75 mL). The resulting thin suspension was filtered into a warm flask and water (0.8 mL) was added. The solution was heated to 80 °C. After this temperature had been reached, hydrochloric acid, 37% concentrated (0.512 ml, 6.23 mmol) was added, and the solution was seeded with about 30 mg (about 1 wt%) Compound A-HC1 salt. The seed held for 15 min. Next the suspension was cooled to 20 °C over 2 h. Finally heptane (17 mL, 6 Vol) was added over 2 h by syringe pump. The suspension was allowed to stir under ambient conditions overnight.

The yellow-green solid was filtered on an M-porosity glass filter frit. The wet cake was washed with 1: 1 heptane/IPA (2 Vol, 5.5 mL) and then with 2 Vol additional heptane (5.5 mL). The cake was dried by passage of air. The dried cake (3.06 g , 78.5 wt%, 94 LC area% Compound A, 62% yield) was analyzed by chiral LC showing optical purity of 99.6% ee.

Thus, Example 6 shows the synthesis of enantiomerically pure Compound A and the hydrochloric salt thereof, according to the disclosure.

EXAMPLE 7

RE-CRYSTALLIZATION OF COMPOUND A

A-HCI A monohydrate

Scheme 7: Re-crystallization of Compound A

Compound A-HCI was recrystallized to Compound A. A (60 L) jacketed reactor, Reactor 1, with a jacket temperature of 20 °C was charged with 5291 g, 1.0 equivalent of Compound A-HCI. 2 Vol (10.6 L) of IPA and 1 Vol (5.3 L) of water were added to Reactor 1 and agitation of Reactor 1 was initiated.

An aqueous NaHC03 solution was prepared in advance by charging NaHC03 (1112 g) and water (15.87 L, 3 Vol) into an appropriate container and shaking well until all solids were dissolved. The prepared NaHC03 solution was added to Reactor 1 over at least 30 min, maintaining the batch temperature below 30 °C. The batch temperature was then adjusted to about 60 °C. The reaction solution was filtered by transferring the contents of Reactor 1 through an in-line filter to a second reactor, Reactor 2, having a jacket temperature of 60+5 °C. Reactor 2 was charged with water (21.16 L) over at least 30 min through an in-line filter, maintaining the batch temperature at approximately 60 °C. After the addition, the batch temperature was adjusted to approximately 60 °C.

A seed was prepared by combining Compound A seed (0.01 equivalents) and IP A/water (20:80) in an appropriate container, in an amount sufficient to obtain a suspension. The seed preparation step was performed in advance. Reactor 2 was charged with the seed slurry. The batch was aged at 55-60 °C for at least 15 min. The batch was cooled to 20+5 °C over at least 1 h. The batch from Reactor 2 was recirculated through a wet mill for at least 1 h, for example, using 1 fine rotor stator at 60 Hz, having a flow rate of 4 L/min, for about 150 min.

The reaction mixture was sampled for particle size distribution during the milling operation. The solids were analyzed by Malvern particle size distribution (PSD) and

microscopic imaging. At the end of the milling operation a sample of the reaction mixture was again analyzed. The supernatant concentration was analyzed by HPLC, and the solids were analyzed by Malvern PSD and microscopic imaging to visualize the resulting crystals.

The batch temperature was adjusted to 35+5 °C and the batch was aged for at least 1 h. The batch was cooled to 20+5 °C over at least 2 h. The reaction mixture was sampled to determine the amount of product remaining in the supernatant. The supernatant concentration was analyzed by HPLC for target of <5 mg/mL Compound A in the supernatant. The contents of Reactor 2 were filtered through an Aurora filter fitted with a <25 μιη PTFE or PP filter cloth.

A 20% v/v IP A/water solution was prepared and the filter cake from Reactor 2 was washed with the 20% IP A/water solution. The cake was then washed with water. If needed, the IP A/water solution, or water alone, can be added to Reactor 2 prior to filtering to rinse the contents of the reactor. The cake was dried under moist nitrogen and vacuum until target residual water and IPA levels were reached. The product had 3.2-4.2% water by KF analysis. The product was analyzed by GC for residual IPA (an acceptable about less than or equal to about 5000 ppm). The yield and purity were determined to be 100% and 99.69% (by HPLC), respectively.

Thus, Example 6 shows the recrystallization of Compound A from the HC1 salt, Compound A-HC1, according to the disclosure.

EXAMPLE 8

SYNTHESIS OF (R)-6-(l-(8-FLUORO-6-(l-METHYL-lH-PYRAZOL-4-YL)- [l,2,4]TRIAZOLO[4,3-A]PYRIDIN-3-YL)ETHYL)-3-(2-METHOXYETHOXY)-l,6- NAPHTHYRIDIN-5(6H)-ONE (COMPOUND A)

HYDZ A

Scheme 8 Synthesis of (R)-6-(l-(8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one

(R)-6-(l-(8-fluoro-6-(l-methyl-lH-pyrazol-4-yl)-[l,2,4]triazolo[4,3-a]pyridin-3-yl)ethyl)-3-(2-methoxyethoxy)-l,6-naphthyridin-5(6H)-one was synthesized according to Scheme 8 by the following procedure. A clean and dry 60 L reactor was fitted with a reflux condenser, nitrogen inlet, and vented to a scrubber (Reactor 1). The jacket temperature of Reactor 1 was set to 20 °C. A scrubber was set up to the vent of Reactor 1, and aqueous bleach solution was charged to the scrubber. The circulating pump (commercial 5.25% NaOCl) was initiated. The scrubber pump was turned on and N2 sweep on Reactor 1 was started. Reactor 1 was charged with 1599.5 g (0.52 equivalents) of Lawesson’s reagent. Reactor 1 was then charged with 24.4 L acetonitrile (MeCN). Agitation of Reactor 1 was initiated. 3664.7 g (1.0 equivalent) of HYDZ was added to the reactor in portions over 1+0.5 h, using acetonitrile (5 L) as rinse. The reactor was heated to 50+5 °C and aged until an LC assay shows consumption of HYDZ (> 99% conversion).

The reactor was cooled to 20 °C and the reaction was assayed by HPLC for

Compound A. The assay showed a 99% crude yield of Compound A.

The contents of Reactor 1 were transferred to second reactor, Reactor 2, through a 1 micron inline filter. Reactor 2 was charged with 2 L of water. Reactor 2 was connected to a batch concentrator and vacuum distilled until a final volume of about 10 L. The jacket temperature was 50 °C during distillation and the pot temperature was maintained below 50 °C. The batch was then cooled to 20 °C.

In a separate container a solution of 10% K2CO3 was prepared by adding 1160 g K2CO3 and 10450 mL water to the container and shaking the container well until all the solid was dissolved. The K2CO3 solution was added to Reactor 2 through an in-line filter (5 μηι). 13 kg of purified water was added to the reactor through the in-line filter (5 μηι).

A Compound A seed was added to the reactor through an addition port. The resulting slurry was aged for one hour during which crystallization was observed. The reactor was placed under vacuum and charged with 16 L of water. The resulting slurry was aged at 20 °C overnight. The product slurry was filtered through a 25 μιη filter cloth and washed with 10 L of a 10% MeCN in water solution, followed by 12 L of water. The product was dried on a frit under a stream of ambient humidity filtered air.

Compound A was isolated as a monohydrate crystalline solid which reversibly dehydrates at < 11% RH. After drying, there was 3.9 wt.% water present in constant weight solid as determined by KF. 3.317 kg, 89% yield, of Compound A was isolated as a pale yellow solid. The product had a purity of 99.4 wt.% as determined by LCAP.

EXAMPLE 9

SYNTHESIS OF NAPH – ROUTE 1

CuBr (5-10%)

ethyl 5-bromo-2- Bromonaphthyridinone Naphthyridinone ether methylnicotinate

Scheme 9: Synthesis of NAPH – Route 1

The NAPH starting material for the synthesis of Compound A was synthesized according to Scheme 9, Route 1 by the following procedure. The jacket temperature of a 6 L jacketed reactor, Reactor 1, was set to 22 °C. 2409 g (1.0 equiv) of ethyl 5-bromo-2-methylnicotinate, 824 g (1.0 equivalent) of triazine, and 3.6 L dimethyl sulfoxide (DMSO) were added to the reactor. The jacket temperature was adjusted to 45 °C. The reactor was agitated until a homogenous solution resulted. Once complete dissolution has occurred (visually) the jacket of Reactor 1 was cooled to 22 °C.

A second, 60 mL reactor, Reactor 2, was prepared. 8.0 L of water was charged to a scrubber. 4.0 L of 10 N sodium hydroxide was added to the scrubber and the scrubber was connected to Reactor 2. The cooling condenser was started. 6411.2 g of cesium carbonate and 12.0 L of DMSO were added to Reactor 2. Agitation of Reactor 2 was initiated. The batch temperature of Reactor 2 was adjusted to 80 °C. The solution from Reactor 1 was added slowly over 1 h at 80 °C, while monitoring the internal temperature. 1.2 L of DMSO was added to Reactor 1 as a rinse. The DMSO rinse was transferred from Reactor 1 to Reactor 2 over 6 min. Reactor 2 was agitated for more than 1 h and the conversion to 3-bromo-l,6-naphthyridin-5(6H)-one was monitored by HPLC until there was < 1.0% ethyl 5-bromo-2-methylnicotinate remaining. When the reaction was complete the batch temperature was adjusted to 60 °C. 24.0 L (10V) of water was added to Reactor 2 over 2 h, maintaining a reaction temperature of 60+5 °C, using a peristaltic pump at 192 mL/min. Reactor 2 was cooled to 22 °C over 1 h 10 min. Stirring was continued at 22+5 °C until the supernatant assays for less than 3mg/mL of 3-bromo-l,6-naphthyridin-5(6H)-one (analyzed by HPLC). The crystallized product was filtered through an Aurora filter fitted with 25 μιη polypropylene filter cloth. The reactor and filter cake were washed with a 75 wt% H20-DMSO solution (3 Vol made from 1.6 L DMSO and 5.6 L water), followed by water (7.2 L, 3 Vol), and finally toluene (7.2 L, 3 Vol). The product cake was dried on the aurora filter under vacuum with a nitrogen stream at ambient temperature. The product was determined to be dry when the KF was < 2.0 wt% water. 2194 g of 3-bromo-l,6-naphthyridin-5(6H)-one was isolated as a beige solid. The chemical purity was 99.73%. The adjusted yield was 2031.6 g (91.9%).

The jacket temperature of a 100 L reactor, Reactor 3, was set to 15+5 °C. 6.45 L of 2-methoxyethanol was added to the reactor and agitation was initiated. (8107 g) lithium tert-butoxide was added portion- wise to the reactor, maintaining the reactor temperature in a range of 15 °C to 24 °C. 3795 g of 3-bromo-l,6-naphthyridin-5(6H)-one was added to the reactor. 4 mL of 2-methoxyethanol was added to rinse the solids on the wall of the reactor. The reactor contents were stirred for at least 5 min. The reaction mixture was heated to distillation to remove i-BuOH and water, under 1 atm of nitrogen (jacket temperature 145 °C). Distillation continued until the pot temperature reached 122+3 °C. The reactor contents were sampled and analyzed for water content by KF. The reaction mixture was cooled to less than 35 °C. 243 g CuBr was added to the reactor. The reaction mixture was de-gassed by applying vacuum to 50 torr and backfilling with nitrogen three times. The batch was heated to 120+5 °C while maintaining the jacket temperature below 150 °C. The batch was agitated (174 RPM) for 15.5 h. A sample of the reaction was taken and the reaction progress was monitored by HPLC. When the remaining 3-bromo-l,6-maphthyridin-5(6H)-one was less than 1%, the jacket temperature was cooled down to 25 °C.

An Aurora filter was equipped with a 25 μιη PTFE cloth and charged with Celite®. The reactor content was transferred onto the filter cloth and the filtrate was collected in the reactor. 800 mL of 2-methoxyethanol was added to the reactor and agitated. The reactor contents were transferred onto the filter and the filtrate was collected in the reactor. 5.6 L of acetic acid was added to the reactor to adjust the pH to 6.5, while maintaining the temperature at less than 32 °C. The batch was then heated to 80 °C. The reaction mixture was concentrated to 3.0+5 Vol (about 12 L) at 80+5 °C via distillation under vacuum.

In a separate container labeled as HEDTA Solution, 589.9 g of N-(2-hydroxyethyl)ethylenediaminetriacetic acid trisodium salt hydrate and 7660 mL water were mixed to prepare a clear solution. The HEDTA solution was slowly added to the reactor while maintaining the temperature of the batch at about 80-82 °C. The batch was then cooled to 72 °C.

An aqueous seed slurry of NAPH (31.3g) in 200 mL of water was added to the reactor. The slurry was aged for 30+10 min. 20 L of water was slowly added to the reactor to maintain the temperature at 65+5 °C. The batch was aged at 65+5 °C for 30 min. The batch was cooled to 20 °C over 1 h. The reactor contents were purged with compressed air for 1 h, and then the batch was further cooled to – 15 °C and aged for 12.5 h. The batch was filtered through a centrifuge fitted with 25 μιη PTFE filter cloth. 5.31 Kg of wet cake was collected (60-62 wt ). The wet cake was reslurried in 6V HEDTA solution and filtered through the centrifuge. The collected wet cake was dried in the centrifuge, and transferred to an Aurora filter for continued drying.

2.82 kg (76% isolated yield) of NAPH was collected having a 2.7% water content by KF.

Thus, Example 8 shows the synthesis of NAPH according to the examples.

EXAMPLE 10

SYNTHESIS OF NAPH – ROUTE 2

Scheme 10: Synthesis of NAPH via Route 2

The NAPH starting material for the synthesis of Compound A was synthesized according to Scheme 10, Route 2, by the following procedure.

Preparation of protected 2-methoxy-pyridin-4ylamine. A 1600 L reactor was flushed with nitrogen and charged with 120 L of N,N-dimethylacetamide, 100.0 kg 2-methoxy-pyridin-4-ylamine, and 89.6 kg triethylamine, maintaining the temperature of the reactor at less than 20 °C. In a separate container, 103.0 kg pivaloyl chloride was dissolved in 15.0 L of N,N-dimethylacetamide and cooled to less than 10 °C. The pivaloyl chloride solution was added to the reactor using an addition funnel over 3.2 hours while maintaining the reactor temperature between 5 °C and 25 °C. The addition funnel was washed with 15.0 L of N,N-dimethylacetamide, which was added to the reactor. The reaction was stirred for 2.3 hours at 20-25 °C. A sample of the reaction was taken and analyzed for 2-methoxy-pyridin-4ylamine by TLC. No 2-methoxy-pyridin-4ylamine remained in the solution and the reaction was aged at 20-25 °C under nitrogen over night. 1200 L of deionized water was added to the reaction over 2

hours at while the reaction was maintained at 5-15 °C. The resulting mixture was stirred at 15 °C for 2 hours and then cooled to 5 °C. The reaction was centrifugated at 700-900 rpm in 3 batches. Each batch was washed 3 times with deionized water (3x 167 L) at 800 rpm. The wet solids obtained were dried under vacuum at 55 °C for 18 hours in 2 batches, sieved and dried again under vacuum at 55 °C for 21 hours until the water content was < 0.2% as determined by KF. 80.4 kg (89.7% yield) of the protected 2-methoxy-pyridin-4ylamine was collected as a white solid.

Preparation of protected 3-formyl-4-amino-2-methoxypyridine. A 1600 L reactor was flushed with nitrogen and charged with 1000 L of THF and 70.5 kg of the protected 2-methoxy-pyridin-4ylamine. The reaction was stirred for 10 min at 15-25°C. The reaction was cooled to -5 °C and 236.5 kg of w-hexyllithium (solution in hexane) was added over 11.5 hours while maintaining the temperature of the reaction at <-4°C. The reaction was maintained at <-4°C for 2 hours. A sample of the reaction was quenched with D20 and the extent of the ortho-lithiation was determined by 1H NMR (98.2% conversion). 61.9 kg dimethylforaiamide (DMF) was added at <-4°C over 3.2 h. After stirring 7.5 hours at <-4°C, a sample of the reaction was assayed for conversion by HPLC (98.5% conversion).

A 1600 L reactor, Reactor 2, was flushed with nitrogen and charged with 145 L THF and 203.4 kg of acetic acid. The resulting solution was cooled to -5 °C. The content of the first reactor was transferred to Reactor 2 over 2.5 hours at 0 °C. The first reactor was washed with 50 L THF and the washing was transferred into Reactor 2. 353 L deionized water was added to Reactor 2 while maintaining the temperature at less than 5 °C. After 15 min of decantation, the aqueous layer was removed and the organic layer was concentrated at atmospheric pressure over 5 hours until the volume was 337 L. Isopropanol (350 L + 355 L) was added and the reaction was again concentrated at atmospheric pressure until the volume was 337 L. Distillation was stopped and 90 L of isopropanol was added to the reactor at 75-94 °C. 350 L of deionized water was added to the reactor at 60-80 °C over 1 h (the temperature was about 60-65 °C at the end of the addition). The reaction was cooled to 0-5 °C. After 1 hour, the resulting suspension was filtered. Reactor 2 was washed twice with deionized water (2x 140L). The washings were used to rinse the solid on the filter. The wet solid was dried under vacuum at 50 °C for 15 h. 71.0 kg (80% yield) of the protected 3-formyl-4-amino-2-methoxypyridine was produced. The purity of the formyl substituted pyridine was found to be 92.7% by LCAP.

A 1600 L reactor, Reactor 3, was flushed with nitrogen and successively charged with 190 L ethanol, 128.7 kg of protected 3-formyl-4-amino-2-methoxypyridine, 144 L of deionized water and 278.2 kg of sodium hydroxide. The batch was heated to 60-65°C and 329.8 kg of the bisulfite adduct was added over 1 h. After lh of stirring, a sample was taken for HPLC analysis which showed 100% conversion. The batch was aged 2 hours at 60-65 °C, then was allowed to slowly cool down to 20-25 °C. The batch was aged 12 h at 20-25 °C. The batch was filtered and the reactor was washed with water (2x 125 L). The washings were used to rinse the solid on the filter. The wet solid was transferred to the reactor with 500 L deionized water and heated to 45-50 °C for 1 h. The batch was allowed to return to 20-25 °C (24 h). The solid was filtered and the reactor was washed with deionized water (2x 250 L). The washings were used to rinse the solid on the filter. 112.5 kg of wet white solid was obtained (containing 85.1 Kg (dry) of the naphthyridine, 72.3% yield, greater than 97% purity as determined by HPLC). The wet product was used directly in the next step, without drying.

A 1600 L reactor was flushed with nitrogen and charged with 417 L of deionized water and 112.5 kg of the wet napthyridine. The scrubber was filled with 700 L of water and 92.2 kg monoethanolamine. A solution of hydrochloric acid (46.6 kg diluted in 34 L of deionized water) was added to the reactor at 15-20 °C over 10 minutes. The batch was heated to 60-65 °C for 3 h. A sample of the batch was taken and contained no remaining starting material as determined by TLC. A solution of concentrated sodium hydroxide (58.2 kg in 31 L of deionized water) was added to the reactor at 60-65 °C. 65% of the solution was added over 15 min and then the batch was seeded with crystallized NAPH. Crystallization was observed after 2.5 h and then the remaining35% of the sodium hydroxide solution was added (pH – 11.1). The batch was cooled to 25-30 °C and a solution of sodium phosphate monobasic (1.8 kg in 2.9 L of deionized water) was added over 25 min at 25-30 °C) (pH = 6.75). The batch was stirred at 15-20 °C for 12 hours and filtered. The reactor was washed twice with deionized water (2x 176 L). The washings were used to rinse the solid on the filter. The wet solid was dried under vacuum at 50 °C until the water content was < 5% (by KF), to give 78.1 kg (73.8% yield, > 95%)) of NAPH as a beige powder.

Thus, Example 9 shows the synthesis of NAPH according to the disclosure.

EXAMPLE 11

SYNTHESIS OF (R)-2-(3-(2-METHOXYETHOXY)-5-OXO-l,6-NAPHTHYRIDIN-6(5H)- YL)PROPANOIC ACID NAPHTHALENE-2-SULFONATE (NAPA)

6N HCI/ THF 80C

Scheme 11: Synthesis of NAPA, Route 3

NAPA was synthesized according to Scheme 11, Route 3 by the following procedure. 4.75 g of 3-(2-Methoxyethoxy)-l,6-naphthyridin-5(6H)-one was suspended in 45 mL of DMF. 2.58 mL (s)-methyl lactate and 9.05 g triphenylphosphine were added to the suspension. The reaction mixture was cooled to 0 °C. 5.12 mL diethyl azodicarboxylate (DEAD) was added dropwise via syringe. The mixture was stirred at 0 °C for 1 h. A sample of the reaction was taken and the reaction was determined to be complete by LCMS. The reaction mixture was concentrated under vacuum to give crude material as a yellow oil.

1 g of the crude material was loaded in dichloromethane onto a silia gel pre-column. The sample was purified using the Isco Combi-Flash System; column 40 g, solvent system hexane/ethyl acetate, gradient 0-100% ethyl acetate over 15 minutes. Product eluted at 100% ethyl acetate. The product fractions were combined and concentrated under vacuum. 256 mg of (R)-methyl 2-(3-(2-methoxyethoxy)-5-oxo-l,6-naphthyridin-6(5H)-yl)propanoate was collected as a pale yellow oil.

The remaining residue was partitioned between benzene and 6N aq hydrochloric acid (35.9 mL). The acidic layer was extracted with benzene (3x), diethyl ether (2x), ethyl acetate (2x) and dichloromethane (lx). The dichloromethane layer was back extracted with 6N aq. Hydrochloric acid (2x). The aqueous layer was diluted with THF (80 mL). The mixture was heated at 80 °C for 3 h. The reaction mixture was concentrated to remove the THF. The remaining acidic water layer was extracted with ethyl acetate and dichloromethane. The aqueous layer was concentrated under vacuum. The remaining solid was triturated with methanol. The mixture was filtered to remove the solid (naphthyridone). The methanol layer was concentrated under vacuum. The remaining solid was dried overnight on a freeze drier. 10.2 g of material was collected as a yellow solid. NAPA made up 72% of the material as determined by HPLC.

1.0 g of the crude material was dissolved in minimal hot iPrOH then filtered and cooled to RT. Crystallization didn’t occur; therefore the solution was cooled in the freezer overnight. A yellow precipitate formed. The solid was collected on a glass frit and was washed with minimal iPrOH. 171 mg of yellow solid was collected, which was NAPA with a small amount of naphthyridone by LC-MS and 1H NMR.

Acid-base extraction. About 1 g of the crude material was dissolved in saturated aqueous sodium bicarbonate. The crude material was extracted with dichloromethane. The pH of the aqueous layer was adjusted to 6-7 with acetic acid then extracted with dichloromethane. 11 mg of the product was isolated; the majority of the product remained in the aqueous layer. The pH was reduced to approximately 4-5 with additional acetic acid. The aqueous layer was extracted with dichloromethane, ethyl acetate, and 15% methanol/dichloromethane. The organic layers were concentrated under vacuum to yield 260 mg of NAPA as the free base, as determined by LC-MS.

Thus, Example 10 shows the synthesis of NAPA according to the disclosure.

EXAMPLE 12

SYNTHESIS OF BISULFITE ADDUCT

DMSO

(COCI)2

MeCX ,ΟΗ Et3N

O

aqueous solution

Scheme 12: Synthesis of bisulfite adduct

Method 1

The bisulfite adduct was synthesize according to Method 1 of Scheme 12 by the following procedure. A 2L round-bottom flask (RBF) was purged with nitrogen and charged with 73.1 mL of reagent grade oxalyl chloride and 693 mL methylene chloride. The batch was cooled to less than -40 °C. 88 mL of dimethyl sulfoxide was added to the flask via an addition funnel at less than -40 °C. After the addition, the batch was stirred for 10 in at -60 °C. 97 mL diethylene glycol monomethyl ether was added to the flask at less than -50 °C over 10 min. The resulting white slurry was stirred at -60 °C for 30 min. 229 mL triethylamine was added to the flask via an addition funnel at less than -30 °C over 1 h. The batch was warmed to RT. 300 mL MTBE was added to the flask and the batch was stirred for 15 min. The slurry was filtered through a fritted funnel and the cake was washed with 300 mL MTBE. The filtrate was concentrated to 350-400g and then filtered again to remove triethylamine-HCl salt, and the solid was rinsed with MTBE, resulting in 357.7 g of a slightly yellow filtrate solution. The solution was assayed by QNMR and comprised 19 wt (68 g) of the desired aldehyde (70% crude yield). The solution was concentrated to 150.2 g.

A 500 mL RBF was charged with 60.0 g sodium bisulfite and 150 mL of water to give a clear solution. The concentrated aldehyde solution was added to the aqueous bisulfite solution over 5 min. An exothermic temperature rising was observed up to 60 °C from 18 °C. The solution was rinsed with 15 mL water. The resulting yellow solution was cooled to RT and was stirred under a sweep of nitrogen overnight.. A QNMR of the solution was taken. The solution contained 43 wt.% of the bisulfite adduct (300 g, 70% yield).

Method 2

The bisulfite adduct was synthesized according to Method 2 of Scheme 12 by the following procedure. A 2500 L reactor was flushed with nitrogen and charged with 657.5 L of 2-methoxyethanol. 62.6 kg of lithium hydroxide monohydrate was added to the reactor while maintaining the temperature at less than 30 °C. The reactor was heated to 113+7 °C. 270 L of solvent were distilled over 1 h and then the reactor temperature was adjusted to 110 °C. 269.4 kg of bromoacetaldehyde diethyl acetal was added over 16 minutes, maintaining the temperature between 110 and 120 °C. The reaction was heated to reflux (115-127°C) for 13 hours. A sample of the reaction was assayed and conversion to 2-(2-methoxyethoxy)acetaldehyde was found to be 98.3%. The reaction was cooled to 15-20°C and 1305 L of methyl ie/t-butyl ether (MTBE) and 132 L of deionized water was added to the reactor. The reaction was stirred for 20 min and then was decanted. The aqueous layer was transferred into a 1600 L reactor and the organic layer was kept in the first reactor. The aqueous layer was extracted with 260 L of MTBE for 10 min. After 10 min decantation, the aqueous layer was removed and the organic layer was transferred to the first reactor. The mixed organic layers were washed twice, 15 min each, with a mixture of concentrated sodium hydroxide solution (2x 17.3 kg) diluted in deionized water (2x 120 L). The aqueous layers were removed, and the organic layer was concentrated at atmospheric pressure at 60-65 °C until the volume was 540 L. The organic layer was cooled down to 15-20 °C to give 2-(2-methoxyethoxy)acetaldehyde as an orange liquid solution (417.4 kg) containing 215.2 kg of pure product (87.3% yield) as determined by 1H NMR and HPLC assay.

A 1600 L reactor, Reactor 3, was flushed with nitrogen and charged with 595 L deionized water followed by 37.8 kg sulfuric acid over 25 minutes via addition funnel, while maintaining the temperature below 25 °C. The addition funnel was washed with 124 L of deionized water and the washing was added to Reactor 3.

A 2500 L reactor, Reactor 4, was flushed with nitrogen and charged with 417.4 kg of the solution of the 2-(2-methoxyethoxy)acetaldehyde. The content of Reactor 3 was transferred into Reactor 4 over 25 min while maintaining the temperature of Reactor 4 below 35 °C. The batch was aged at 30-35 °C for 3 hours. A sample of the batch was taken and assayed for 2-(2- methoxyethoxy)acetaldehyde. No 2-(2-methoxyethoxy)acetaldehyde remained. The batch was aged 5 h then cooled to 15-20 °C.

A solution of sodium carbonate (39.2 kg) in deionized water (196 L) was prepared in Reactor 3. The sodium carbonate solution was transferred to Reactor 4 over 25 min while maintaining the temperature of Reactor 4 below 30 °C. The pH of the resulting mixture was pH 5-6. 1.0 kg sodium carbonate was added by portion until the pH was about 7-8. A solution of sodium bisulfite (116.5 kg) in deionized water (218 L) was prepared in Reactor 3. The sodium bisulfite solution was transferred to Reactor 4 over 20 min while maintaining the temperature of Reactor 4 below 30 °C. Reactor 3 was washed with deionized water (15 L) and the washing was added to Reactor 4. The batch was stirred for 1.2 hours. 23.3 kg sodium bisulfite was added to Reactor 4 and the batch was aged overnight. The batch was concentrated under vacuum at 30-50 °C over 6.5 hours until precipitation was observed. The batch was cooled to 0-10°C at atmospheric pressure. After 30 min at 0-10 °C, the suspension was filtered on 2 filters. Reactor 4 was washed with deionized water (2x 23 L). The first washing was used to rinse the solid on the first filter and the second washing was used to rinse the solid on the second filter. Filtrates were joined to give 473.9 kg of an aqueous solution of the bisulfite adduct (202.5 kg of pure product, 76.3% yield) as a yellow liquid.

Thus, Example 11 shows the synthesis of the bisulfite adduct according to the invention.

EXAMPLE 13

SYNTHESIS OF 2,3-DIFLUORO-5-(l-METHYL-lH-PYRAZOL-4-YL)PYRIDINE

Scheme 13: Synthesis of 2,3-difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine, precursor to PYRH

2,3-Difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine was synthesized according to Scheme 13 by the following procedure. A boronic-ate complex slurry was prepared in a first 3-neck-2-L round-bottom flask (RBF #1). RBF #1 was charged with 141 g (66.4 wt%, 0.9 equivalents based on boronic ester) of lithium 2-hydroxy-4,4,5,5-tetramethyl-2-(l-methyl-lH-pyrazol-4-yl)-l,3,2-dioxaborolan-2-uide. 120 mL (1.6 Vol relative to 5-chloro-2,3-difluoropyridine) of nitrogen- sparged (2 h) 2-BuOH and 120 mL (1.6 Vol) nitrogen-sparged (2 h) water were added to RBF #1. Agitation and N2 sweep were initiated. The reaction was aged at 20 °C for at least 30 min (reactions aged to 24 h were also successful).

] A second 3-neck-2-L round-bottom flask (RBF #2) was charged with 1.48 g (0.004 equivalents) of Xphos-palladacycle and 450 mL (6 Vol relative to 5-chloro-2,3-difluoropyridine) of nitrogen- sparged (2 h) 2-BuOH. Vacuum/N2 flush was cycled through RBF #2 three times to inert the RBF with N2. The batch in RBF #2 was heated to 80 °C. 75 g (1.0 equivalents) of 5-chloro-2,3-difluoropyridine was added to RBF #2.

The slurry of boronic-ate complex was transferred from RBF #1 to a 500 mL dropping funnel. RBF #1 was rinsed with 30 mL (0.4 Vol) 2-BuOH. Using the dropping funnel, the slurry of boronic-ate complex was added over 1 h to the hot solution mixture in RBF #2. After 1 h, 95% conversion was observed. If greater than 90% conversion was not observed, additional boronic-ate complex slurry was added (0.1 equivalents at a time with 1.6 Vol of 1: 1 2-BuOH/water relative to boronic-ate complex). After the conversion was complete, the batch was cooled to 50 °C. While cooling, 600 mL (8 Vol) of toluene was added to RBF #2. 300 mL (4 Vol) of 20% w/v NaHS03 in water was added to RBF #2 and the batch was stirred at 50 °C for at least 1 h. The batch was polish filtered using a 5 micron Whatman filter at 50 °C, into a 2-L Atlas reactor. RBF #2 was rinsed with 30 mL (4.0 Vol) of a 1: 1 2-BuOH:toluene solution. The temperature of the batch was adjusted to 50 °C in the Atlas reactor while stirring. The stirring was stopped and the phases were allowed to settle for at least 15 min while maintaining the batch at 50 °C. The bottom, aqueous layer was separated from the batch. The Atlas reactor was charged with 300 mL (4 Vol) of a 20% w/v NaHS03 solution and the batch was stirred at 50°C for 1 h. The agitation was stopped and the phases were allowed to settle for at least 15 min at 50 °C. The bottom, aqueous layer was removed. Agitation was initiated and the Atlas reactor was charged with 200 mL (4 Vol) of 0.5 M KF while keeping the batch at 50 °C for at least 30 min. The agitation was stopped and the phases were allowed to settle for at least 15 min at 50 °C. The bottom, aqueous layer was removed. Agitation was initiated and the reactor was charged with 300 mL (4 Vol) of water. The batch was aged at 50 °C for at least 30 min. Agitation was stopped and the phases were allowed to settle for at least 15 min at 50 °C. The bottom, aqueous later was removed.

The organic phase was concentrated by distillation under reduced pressure (180 torr, jacket temp 70°C, internal temp about 50 °C) to a minimal stir volume (about 225 mL). 525 mL (7 Vol) of 2-BuOH was added to the Atlas reactor. The organic batch was again concentrated using reduced pressure (85-95 torr, jacket temp 75 °C, internal temp about 55 °C) to a minimal stir volume (about 125 mL). The total volume of the batch was adjusted to 250 mL with 2-BuOH.

525 mL (7 Vol) heptane was added to the slurry mixture in the Atlas reactor. The jacket temperature was adjusted to 100 °C and the batch was aged for more than 15 min, until the batch became homogeneous. The batch was cooled to 20 °C over at least 3 h. A sample of the mixture was taken and the supernatant assayed for 2,3-difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine. If the concentration was greater than 10 mg/mL, the aging was continued for at least 1 h until the supernatant concentration was less than 10 mg/mL. The batch was filtered using a medium frit. The filter cake was washed with 150 mL (2 Vol) 30% 2-BuOH/heptane solution followed by 150 mL (2 Vol) heptane. The filter cake was dried under N2/vacuum. 76.64 g of 2,3-difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine was isolated as a white solid (87% yield).

A 60 L jacketed reactor was fitted with a reflux condenser. The condenser cooling was initiated at 0+5 °C. The reactor was charged with 2612 g (1 equivalent) of 2,3-difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine and placed under an atmosphere of nitrogen. 31.7 L (12.2 Vol) water was added to the reactor and the resulting slurry was nitrogen sparged for 1 h with agitation. 7221 mL (6 equivalents) of hydrazine (35 wt% in water) was added to the reactor under a nitrogen atmosphere. The reactor was heated to 100 °C for 2+2 h until reaction was complete by HPLC analysis. The reactor was cooled to 20 °C over 2+1 h at a rate of 40°C/h. The reactor contents were stirred for 10+9 hours until the desired supernatant assay (< 2mg/mL PYRH in mother liquor). The reactor contents were filtered through an Aurora filter fitted with 25 μιη polypropylene filter cloth. The collected filter cake was washed with 12.0 L (4.6 V) of water in three portions. The filter cake was dried on the Aurora filter for 4-24 h at 22+5 °C, or until the product contained less than 0.5% water as determined by KF. The dry product was collected. 2.69 kg (97% yield) 2,3-Difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine was collected as a white crystalline solid. The solid had a water content of 12 ppm as determined by KF.

Thus, Example 12 shows the synthesis of 2,3-Difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine, a precursor to PYRH, according to the disclosure.

EXAMPLE 14

SYNTHESIS OF PYRH – ROUTE 2

Scheme 14: Synthesis of 3-fluoro-2-hydrazinyl-5-(l-methyl-lH-pyrazol-4-yl)-pyridine (PYRH)

3-fluoro-2-hydrazinyl-5-(l-methyl-lH-pyrazol-4-yl)-pyridine was synthesized according to Scheme 14 by the following procedure. A 60 L jacketed reactor was fitted with a 5 L addition funnel and the jacket temperature was set to 20+5 °C. 36.0 L (15 Vol) of 2-methyltetrahydrofuran was added to the reactor via a 20 μιη inline filter with vacuum using polypropylene transfer lines. The solution was sparged by bubbling nitrogen through a dipstick in the solution for 1+0.5 h with agitation. After 1 h the dipstick was removed but the nitrogen sweep continued. 1.55 kg of sparged 2-MeTHF was removed to be used as rinse volumes. 36.7 g of Pd2dba3, 75.6 g X-Phos, 259 g of tetrabutylammonium bromide, and 7397 g of potassium phosphate tribasic were added to the reactor. The manhole was rinsed with 0.125 kg of sparged 2-MeTHF. The reactor was agitated and the nitrogen sweep continued for 1+0.5 h. Then the nitrogen sweep was stopped and the reaction left under a positive pressure of nitrogen.

3.6 L (1.5 Vol) of sparged water was prepared in advance by bubbling nitrogen through a 4 L bottle of water for 1+0.5 h. The nitrogen sparged water was transferred to the 5 L addition funnel via a 20 μηι inline filter with vacuum using polypropylene transfer lines, then slowly added to the reaction while maintaining the internal temperature at 20+5 °C. The 5 L addition funnel was replaced with a 2 L addition funnel. 2412 g of 5-chloro-2,3-difluoropyridine was added to the 2 L addition funnel. The 5-chloro-2,3-difluoropyridine was then added to the reaction through the 2 L addition funnel. The 2L addition funnel was rinsed with 0.060 kg of sparged 2-MeTHF. 83.8 g (1.15 equivalents) of l-methylpyrazole-4-boronic acid, pinacol ester was added to reactor, the reactor was swept with nitrogen for 1+0.5 h, then left under a positive pressure of nitrogen. The internal temperature of the reactor was adjusted to 70+5 °C. The batch was agitated at 70+5 °C for at least 4 hours after the final reagent was added. A sample was taken from the reaction and the reaction progress assayed for conversion. The progress of the reaction was checked every 2 hours until the reaction was completed (e.g., greater than 99% conversion). The batch was cooled to 20+5 °C.

A 20% w/v sodium bisulfite solution (12.0 L, 5 Vol) was prepared by charging 12.0 L of water then 2411 g sodium bisulfite to an appropriate container and agitating until

homogeneous. The 20% sodium bisulfite solution was transferred into the reactor and agitated for 30 minutes. The agitation was stopped, the phases allowed to settle, and the aqueous phase was removed. A 0.5 M potassium fluoride solution (12.0 L, 5 Vol) was prepared by charging 12.0 L of water and 348 g of potassium fluoride to an appropriate container and agitating until homogenous. The 0.5 M potassium fluoride solution was transferred into the reactor and agitated for 30 min. The agitation was stopped, the phases were allowed to settle, and the aqueous phase was removed. A 25% w/v sodium chloride solution (12.0 L, 5 Vol) was prepared by charging an appropriate container with 12.0 L of water and 2999 g of sodium chloride and agitating until homogeneous. The 25% sodium chloride solution was transferred into the reactor and agitated for 30 min. The agitation was stopped, the phases were allowed to settle, and the aqueous phase was removed from the reactor.

The organic phase was distilled at constant volume (36 L, 15 Vol) while maintaining the internal temperature of the reactor at 50+5 °C by adjusting the vacuum pressure until no more than 0.3% of water remained. 2-Methyltetrahydrofuran was added to the reactor as needed to

maintain constant volume. The batch was cooled to 20 °C and transferred into drums. The batch was transferred using a polish filter (using a 5 μιη inline filter) into a 60 L jacketed reactor with a batched concentrator attached. 1.2 L of 2-MeTHF was used to rinse the drums. The batch was concentrated to about 9 Vol while maintaining the internal temperature of the vessel at 50+5 °C by adjusting the vacuum pressure. The batch was then distilled at constant volume (22.0 L, 9Vol) while maintaining the internal temperature of the vessel at 50+5 °C by adjusting the vacuum pressure. Heptane was added with residual vacuum until a 15% 2-MeTHF:heptane supernatant mixture was obtained. The pressure was brought to atmospheric pressure under nitrogen. The reactor was cooled to 20+5 °C over 2+2 h. The batch was agitated at 20+5 °C until an assay of the supernatant indicated that the amount of product was 7 mg/mL 2,3-difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine.

A 10% 2-MeTHF:heptane (7.2 L, 3 Vol) wash solution was prepared by mixing 720 mL of 2-MeTHF and 6.5 L of heptane. The batch slurry was filtered through an Aurora filter fitted with a 25 μιη polypropylene filter cloth, resulting in heavy crystals that required pumping with a diaphragm pump using polypropylene transfer lines through the top of the reactor while stirring. The mother liquor was recycled to complete the transfer. The reactor and filter cake were washed with two portions of the 10% 2-MeTHF:heptane wash solution (3.6 L each). The product cake was dried on a frit under a nitrogen stream at ambient temperature. The 2,3-difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine was determined to be dry when the 1H NMR assay was < 0.05+0.05. 2.635 kg was isolated as an off white crystalline solid (85% yield).

A 60 L jacketed reactor was fitted with a reflux condenser. The condenser cooling was initiated at 0+5 °C. The reactor was charged with 2612 g (1 equivalent) of 2,3-difluoro-5-(l-methyl-lH-pyrazol-4-yl)pyridine and placed under an atmosphere of nitrogen. 31.7 L (12.2 Vol) water was added to the reactor and the resulting slurry was nitrogen sparged for 1 h with agitation. 7221 mL (6 equivalents) of hydrazine (35 wt% in water) was added to the reactor under a nitrogen atmosphere. The reactor was heated to 100 °C for 2+2 h until reaction was complete by HPLC analysis. The reactor was cooled to 20 °C over 2+1 h at a rate of 40°C/h. The reactor contents were stirred for 10+9 hours until the desired supernatant assay was reached (< 2mg/mL PYRH in mother liquor). The reactor contents were filtered through an Aurora filter fitted with 25 μιη polypropylene filter cloth. The collected filter cake was washed with 12.0 L

(4.6 V) of water in three portions. The filter cake was dried on the Aurora filter for 4-24 h at 22+5 °C, or until the product contained less than 0.5% water as determined by KF. The dry product was collected. 2.69 kg was isolated as a white crystalline solid (97% yield). The water content was determined to be 12 ppm by KF.

 

WO2007075567A1 * Dec 18, 2006 Jul 5, 2007 Janssen Pharmaceutica, N.V. Triazolopyridazines as tyrosine kinase modulators
WO2007138472A2 * May 18, 2007 Dec 6, 2007 Pfizer Products Inc. Triazolopyridazine derivatives
WO2008008539A2 * Jul 13, 2007 Jan 17, 2008 Amgen Inc. Fused heterocyclic derivatives useful as inhibitors of the hepatocyte growth factor receptor
WO2008051805A2 * Oct 18, 2007 May 2, 2008 Sgx Pharmaceuticals, Inc. Triazolo-pyridazine protein kinase modulators
WO2008155378A1 * Jun 19, 2008 Dec 24, 2008 Janssen Pharmaceutica Nv Polymorphic and hydrate forms, salts and process for preparing 6-{difluoro[6-(1-methyl-1h-pyrazol-4-yl)[1,2,4]triazolo[4,3-b]pyridazin-3-yl]methyl}quinoline

References:
1. Hughes, P. E.; et. al. Abstract 728: AMG 337, a novel, potent and selective MET kinase inhibitor, has robust growth inhibitory activity in MET-dependent cancer models. Cancer Res 2014, 74, 728.
2. Boezio, A. A.; et. al. Discovery and optimization of potent and selective triazolopyridazine series of c-Met inhibitors. Bioorg Med Chem Lett 2009, 19(22), 6307-6312.
3. ClinicalTrials.gov Phase 2 Study of AMG 337 in MET Amplified Gastric/Esophageal Adenocarcinoma or Other Solid Tumors. NCT02016534 (retrieved 10-06-2015)
4. ClinicalTrials.gov A Study of AMG 337 in Subjects With Advanced Solid Tumors. NCT01253707 (retrieved 10-06-2015)

/////////// AMG-337,  AMG337,  AMG 337,  1173699-31-4, AMGEN, ESOPHAGUS

O=C1C2=C(N=CC(OCCOC)=C2)C=CN1[C@@H](C3=NN=C4C(F)=CC(C5=CN(C)N=C5)=CN43)C

Share

Asvasiran sodium (ALN-RSV01)

 phase 2  Comments Off on Asvasiran sodium (ALN-RSV01)
Apr 132016
 

RNA, (C-U-U-G-A-C-U-U-U-G-C-U-A-A-G-A-G-C-C-DT-DT), COMPLEX WITH RNA (G-G-C-U-C-U-U-A-G-C-A-A-A-G-U-C-A-A-G-DT-DT)

Duplex of guanylyl-(3′->5′)-guanylyl-(3′->5′)-cytidylyl-(3′->5′)-uridylyl-(3′->5′)-cytidylyl-(3′->5′)-uridylyl-(3′->5′)-uridylyl-(3′->5′)-adenylyl-(3′->5′)-guanylyl-(3′->5′)-cytidylyl-(3′->5′)-adenylyl-(3′->5′)-adenylyl-(3′->5′)-adenylyl-(3′->5′)-guanylyl-(3′->5′)-uridylyl-(3′->5′)-cytidylyl-(3′->5′)-adenylyl-(3′->5′)-adenylyl-(3′->5′)-guanylyl-(3′->5′)-thymidylyl-(3′->5′)-thymidine and thymidylyl-(5′->3′)-thymidylyl-(5′->3′)-cytidylyl-(5′->3′)-cytidylyl-(5′->3′)-guanylyl-(5′->3′)-adenylyl-(5′->3′)-guanylyl-(5′->3′)-adenylyl-(5′->3′)-adenylyl-(5′->3′)-uridylyl-(5′->3′)-cytidylyl-(5′->3′)-guanylyl-(5′->3′)-uridylyl-(5′->3′)-uridylyl-(5′->3′)-uridylyl-(5′->3′)-cytidylyl-(5′->3′)-adenylyl-(5′->3′)-guanylyl-(5′->3′)-uridylyl-(5′->3′)-uridylyl-(5′->3′)-cytidine

Asvasiran sodium (ALN-RSV01),

C401H500N150O290P40,

CAS 1386946-83-3, 870094-26-1

Alnylam Pharmaceuticals

  • Originator Alnylam Pharmaceuticals
  • Class Antivirals; Small interfering RNA
  • Mechanism of Action Nucleocapsid protein modulators; RNA interference

Treatment of Human Respiratory Syncytial Virus (RSV) Infection

Nucleocapsid protein modulators, RNA interference

  • 05 Nov 2014 Alnylam receives patent allowance for RNAi technology in USA
  • 20 Feb 2014 Suspended – Phase-II for Respiratory syncytial virus infections in USA (Intranasal) (Alnylam Form 10-K filed in February 2014)
  • 20 Feb 2014 Suspended – Phase-I for Respiratory syncytial virus infections in Europe (Intranasal) (Alnylam Form 10-K filed in February 2014)

 

Aerosolised ALN-RSV01 – Alnylam; ALN RSV01; Intranasal ALN-RSV01 – Alnylam

Alnylam, under license from the University of South Alabama, and with Asian licensee Kyowa Hakko Kirin (formerly Kyowa Hakko Kogyo), is developing a nasally administered formulation of asvasiran sodium (ALN-RSV01), an siRNA that targets the respiratory syncytial virus (RSV) N gene and inhibits viral replication, for the potential treatment or prevention of RSV infection.

.In June 2007, a phase II trial was initiated; in January 2008, top-line data were reported . In March 2013, development was ongoing . In August 2008, Kyowa planned to file the drug for marketing approval in 2014. In March 2013, Alnylam was planning on seeking to outlicense the program to continue to advance the program in other regions .

Alnylam is also developing second-generation agents.

Ex-Asian licensee, Cubist Pharmaceuticals, in collaboration with Alnylam, was previously developing the program for the potential treatment or prevention of RSV infection . However, in February 2013, the deal was terminated . Alnylam was also developing an inhaled formulation of asvasiran sodium; however, in February 2014, the drug was no longer listed on the company’s development pipeline.

WO-2006074346
WO-2009076679
WO-2006062596
WO-2010048590

WO 2016022464

WO 2015173701

WO 2015026792

WO 2014209983

WO 2014031784

US 20130273037

Nucleic Acids Research (2012), 40(21), 10585-10595

WO 2011163518

Drugs of the Future (2009), 34(10), 781-783

Current Opinion in Infectious Diseases (2008), 21(6), 639-643

Antiviral Research (2008), 77(3), 225-231

 

 

John Maraganore, president and chief executive officer of Alnylam Pharmaceuticals,

 

Delivering Value with Integrated Communications led by Cynthia Clayton, Vice President, Investor Relations and Corporate Communications at Alnylam Pharmaceuticals

From the left, Alnylam COO Barry Greene, Adrian Dede, Lauren Virnoche, CEO

Dr. Rachel Meyers, Senior Vice President, Research at Alnylam Pharmaceuticals

 

 

Dr. Dinah Sah, Vice President of Research and the head of the Alnylam HD team

//////Asvasiran sodium, ALN-RSV01, PHASE 2, Alnylam

SOME OTHER CHEMISTRY

Figure 6: GalNAc–siRNA conjugates.

From Delivery materials for siRNA therapeutics

Nature Materials12,967–977doi:10.1038/nmat3765

http://www.nature.com/nmat/journal/v12/n11/fig_tab/nmat3765_F6.html

 

 

\

 

http://www.google.com/patents/EP2836595A2?cl=en

Share

Zamicastat

 Uncategorized  Comments Off on Zamicastat
Apr 122016
 

2D chemical structure of 1080028-80-3

 

 CAS 1080028-80-3 BASE
1383828-47-4 OF HCL SALT
C21 H21 F2 N3 O S BASE
2H-​Imidazole-​2-​thione, 1-​[(3R)​-​6,​8-​difluoro-​3,​4-​dihydro-​2H-​1-​benzopyran-​3-​yl]​-​1,​3-​dihydro-​5-​[2-​[(phenylmethyl)​amino]​ethyl]​-(R)-5-(2-(Benzylamino)ethyl)-1-(6,8-difluorochroman-3-yl)-1H-imidazole-2(3H)-thione
(R)-5-(2-(Benzylamino)ethyl)-1-(6,8-difluorochroman-3-yl)-1H-imidazole-2(3H)-thione
Molecular Weight, 401.47 BASE
img
BIAL – PORTELA & CA., S.A. [PT/PT]; À Avenida da Siderurgia Nacional P-4745-457 S. Mamede do Coronado (PT)

 

Zamicastat is a dopamine beta-monooxygenase inhibitor in phase I clinical studies at BIAL for the treatment of hypertension and heart failure.
Zamicastat is a potent and selective dopamine β-mono-oxygenase inhibitor. Zamicastat Prevents the Deterioration of Cardiometabolic and Inflammatory Biomarkers in a Genetic Model of Salt-sensitive Hypertension. Chronic high salt intake deteriorates several cardiometabolic and inflammatory biomarkers in Dahl/SS rats, which can be prevented by dopamine β-hydroxylase inhibition with zamicastat.
crystalline forms of l-[(3R)-6,8-difluoro- 3,4-dihydro-2H-l-benzopyran-3-yl]-l,3-dihydro-5-[2-[(phenylmethyl)amino]ethyl]-2H- imidazole-2-thione, i.e. the Renantiomer of

Figure imgf000002_0001

and processes for preparing the same. Background and prior art:Interest in the development of inhibitors of dopamines-hydroxylase (ϋβΗ) has centred on the hypothesis that inhibition of this enzyme may provide significant clinical improvements in patients suffering from cardiovascular disorders such as hypertension or chronic heart failure. The rationale for the use of ϋβΗ inhibitors is based on their capacity to inhibit the biosynthesis of noradrenaline, which is achieved via enzymatic hydroxylation of dopamine. Activation of neurohumoral systems, chiefly the sympathetic nervous system, is the principal clinical manifestation of congestive heart failure (Parmley, W.W., Clinical Cardiology, 18: 440-445, 1995). Congestive heart failure patients have elevated concentrations of plasma noradrenaline (Levine, T.B. et al., Am. J. Cardiol., 49: 1659-1666, 1982), increased central sympathetic outflow (Leimbach, W.N. et al., Circulation, 73: 913- 919, 1986) and augmented cardiorenal noradrenaline spillover (Hasking, G.J. et al., Circulation, 73:615-621, 1966). Prolonged and excessive exposure of the myocardium to noradrenaline may lead to down-regulation of cardiac β] -adrenoceptors, remodelling of the left ventricle, arrhythmias and necrosis, all of which can diminish the functional integrity of the heart. Congestive heart failure patients who have high plasma concentrations of noradrenaline also have the most unfavourable long-term prognosis (Cohn, J.N. et al., N. Engl. J. Med., 311 :819-823, 1984). Of greater significance is the observation that plasma noradrenaline concentrations are already elevated in asymptomatic patients with no overt heart failure and can predict ensuing mortality and morbidity (Benedict, C.R. et al., Circulation, 94:690-697, 1996). An activated sympathetic drive is not therefore merely a clinical marker of congestive heart failure, but may contribute to progressive worsening of the disease.

Potent dopamines-hydroxylase inhibitors having high potency and significantly reduced brain access are disclosed in WO 2008/136695. WO 2008/136695 describes compounds of formula I:

Figure imgf000003_0001

I where Rls R2 and R3 are the same or different and signify hydrogens, halogens, alkyl, nitro, amino, alkylcarbonylamino, alkylamino or dialkylamino group; R4 signifies -alkylaryl or – alkylheteroaryl; X signifies CH2, oxygen atom or sulphur atom; n is 2 or 3; including the individual (R)- and (S)-enantiomers or mixtures of enantiomers thereof; and including pharmaceutically acceptable salts and esters thereof, wherein the term alkyl means hydrocarbon chains, straight or branched, containing from one to six carbon atoms, optionally substituted by aryl, alkoxy, halogen, alkoxycarbonyl or hydroxycarbonyl groups; the term aryl means a phenyl or naphthyl group, optionally substituted by alkyl, alkyloxy, halogen or nitro group; the term halogen means fluorine, chlorine, bromine or iodine; the term heteroaryl means heteroaromatic group. In particular, WO 2008/136695 describes l-[(3R)-6,8-difluoro-3,4-dihydro-2H-l-benzopyran-3-yl]-l,3-dihydro-5-[2- [(phenylmethyl)amino]ethyl]-2H-Imidazole-2-thione.

Processes for the preparation of compounds of formula I, and in particular l-[(3R)-6,8- difluoro-3,4-dihydro-2H-l-benzopyran-3-yl]-l,3-dihydro-5-[2-[(phenylmethyl)amino] ethyl] -2H-Imidazole-2-thione, are described in WO 2008/136695 and are incorporated by reference herein. It is known that polymorphic forms of the same drug may have substantially different pharmaceutically important properties such as dissolution characteristics and bioavailability as well as stability of the drug. Furthermore, different forms may have different particle size, hardness and glass transition temperature. Thus, one form may provide significant advantages over other forms of the same drug in solid dosage form manufacture processes, such as accurate measurement of the active ingredients, easier filtration, or improved stability during granulation or storage. Furthermore, a particular process suitable for one form may also provide drug manufacturers several advantages such as economically or environmentally suitable solvents or processes, or higher purity or yield of the desired product.

2D chemical structure of 1080028-80-3

PATENT

http://www.google.com/patents/WO2012087174A2?cl=en

Preparation of compound 2

[0090] Six lots of compound 2 (designated as lots 1, 2, 3, 4, 5 and 6) were prepared. The starting materials were prepared according to the following experimental protocols.

Lot 1 (Form A)

To a suspension of (R)-5-(2-aminoethyl)-l-(6,8-difluorochroman-3-yl)-lH- imidazole-2(3H)-thione (6.23 g, 20 mmol) in a mixture of Dichloromethane (DCM – 40 ml) and Methanol (40.0 ml) was added BENZALDEHYDE (2.230 ml, 22.00 mmol). To the resulting clear solution SODIUM CYANOBOROHYDRIDE (1.9 g, 28.7 mmol) was added in portions at 20-25°C to avoid intensive foaming and the solution was stirred at 20- 25°C for 40 h. The solution was quenched at 20-25°C with IN HC1 (35 ml), neutralised with 3N NaOH (35 ml), the mixture was extracted with DCM (200 ml). The organic phase was washed with brine, dried (MgS04), evaporated to dryness. The oily residue crystallised from 2-propanol (40 ml) at 20-25°C over a week-end. The crystals were collected, washed with 2-propanol, dried to give 5.2 g of the crude product. Re- crystallisation from 2-propanol-DCM hasn’t removed all impurities. Everything collected, evaporated with silica, applied on a column, eluted with Ethyl Acetate (EA)->EA-MeOH 9:1->4: 1, fractions 8-25 collected to give 3.8 g. Re-crystallised from 2-propanol (45 ml) and DCM (120 ml, removed on a rotavap) to give 2.77 g => initial lot (a) (HPLC 98.3% area) and 0.3 g of undissolved filtered off, by TLC right product. Initial lot (a) re- crystallised from 2-propanol (35 ml) and DCM (95 ml, removed on a rotavap) to give 2.51 g => initial lot (b) (HPLC 98.3% area). Combined with the above undissolved, re- crystallised from acetonitrile (200 ml, reflux to ice bath) to give 2.57 g => initial lot (c) (HPLC 98.8% area). Re-crystallised from acetonitrile (180 ml, reflux to 15°C) to give 2.25 g => Lot 1 (HPLC 99.2% area), mp 190-92°C. Lot 2 (Form A)

[0092] (R)-5-(2-(benzylamino)ethyl)-l-(6,8-difluorochroman-3-yl)-lH-imidazole- 2(3H)-thione (12 g, 29.9 mmol) was dissolved with heating to reflux in Tetrahydrofuran (300 ml), the solution was cooled to 5-10°C, Water (510 ml) was added slowly (approx 10 min) with stirring. The mixture was stirred for 1 h, solid was collected, washed with water, dried to give 11.73 g of product, by HPLC 1% of (R)-5-(2-Aminoethyl)-l-(6,8- difluorochroman-3-yl)-l,3-dihydroimidazole-2-thione hydrochloride and 1% of less polar impurity. The product was dissolved in Tetrahydrofuran (300 ml) with heating to reflux, 2- Propanol (150 ml) was added, the solution was concentrated to approx 100 ml (crystallisation occured), stirred in ice for 1.5 h. Solid was collected, washed with 2- propanol, dried to give 11.2 g of product, by HPLC 0.8% of (R)-5-(2-aminoethyl)-l-(6,8- difluorochroman-3-yl)-lH-imidazole-2(3H)-thione hydrochloride and 0.5% of less polar impurity. The product was dissolved in Tetrahydrofuran (300 ml) with heating to reflux, 2- Propanol (150 ml) was added, the solution was concentrated to approx 100 ml (crystallisation occured), stirred at 20-25°C for 1 h. Solid was collected, washed with 2- propanol, dried to give (R)-5-(2-(benzylamino)ethyl)-l-(6,8-difluorochroman-3-yl)-lH- imidazole-2(3H)-thione (10.22 g, 25.5 mmol, 85 % yield).,

Lot 3 (form B)

To (R)-5-(2-aminoethyl)-l-(6,8-difluorochroman-3-yl)-lH-imidazole-2(3H)- thione (2.36 g, 7.58 mmol) in a mixture of Methanol (15.00 ml) and Dichloromethane (15 ml) was added BENZALDEHYDE (0.845 ml, 8.34 mmol). To the resulting clear solution SODIUM CYANOBOROHYDRIDE (0.702 g, 10.61 mmol) was added in portions at 20- 25°C to avoid intensive foaming and the solution was stirred at 20-25°C for 40 h. The solution was quenched at 20-25°C with IN HC1 (12 ml), neutralised with 3N NaOH (12 ml), the mixture was extracted with DCM (100 ml). The organic phase was washed with brine, dried (MgS04), evaporated to dryness. The residue was purified on a column with EA-MeOH 9: 1 as eluent, fractions collected, concentrated to approx 20 ml, cooled in ice. The precipitate collected, washed with Ethyl Acetate-Petroleum Ether 1 : 1, dried on air to give (R)-5-(2-(benzylamino)ethyl)-l-(6,8-difluorochroman-3-yl)-lH-imidazole-2(3H)- thione (1.55 g, 3.86 mmol, 50.9 % yield). Lot 4 (Form A)

To a 500 mL flask set up for atmospheric distillation was added (R)-5-(2- (benzylamino)ethyl)-l-(6,8-difluorochroman-3-yl)-lH-imidazole-2(3H)-thione (20 g, 49,8 mmol) and Tetrahydrofuran (400 ml) to afford a suspension. The suspension was heated until full dissolution was achieved (61°C) whereupon it was filtered. The resulting solution was then heated to 66°C in order to commence the distillation. A mixture of Water (125 ml) & 2-Propanol (125 ml) was added at the same rate as the distillate was collected. The distillation was continued until 400 mL of distillate was collected. Crystallisation commenced after ~320 mL of distillate was collected. The suspension was cooled to 20°C and aged for 45 min. before filtering and washing with additional 2- propanol (80 mL) and then dried under vacuum at 50°C overnight to give (R)-5-(2- (benzylamino)ethyl)-l-(6,8-difluorochroman-3-yl)-lH-imidazole-2(3H)-thione (18.79 g, 94%). Lot 5 (Form A)

To a mixture of Methanol (66 L) and Water (10 L) at 20°C was added purified (R)-5-(2-(benzylamino)ethyl)-l-(6,8-difluorochroman-3-yl)-lH-imidazole-2(3H)-thione hydrochloride (4.37 kg, 9.98 mol) to afford a suspension. The reaction mixture was then heated to 67°C to affect complete dissolution, whereupon IN Sodium hydroxide (10.48 Ls 10.48 mol, 1.05 eq) was added in a single portion. The reaction mixture was adjusted back to 67°C and held at 67°C for 30 min. The reaction mixture was then cooled to 20°C and aged at 20°C for at least 30 min. The reaction was then filtered and the filter cake washed with aqueous Methanol (1 : 1 v/v, 20 L), sucked down for 15 min. and then dried at 45°C under vacuum, to afford (R)-5-(2-(benzylamino)ethyl)-l-(6,8-difluorochroman-3-yl)-lH- imidazole-2(3H)-thione (3.855 kg, 96%) as a pale tan crystalline solid.

PATENT

WO 2015038022

http://www.google.com/patents/WO2015038022A1?cl=en

 

processes .

(J?) -5- (2-Aminoethyl) -1- (6, 8-difluorochroman-3-yl) -1, 3-dihydroimidazole-2 -thione hydrochloride (the compound of formula 1, below) is a potent, non-toxic and peripherally selective inhibitor of ϋβΗ, which can be used for treatment of certain cardiovascular disorders. Compound 1 is disclosed in WO2004/033447 , along with processes for its preparation.

1

The process disclosed in WO2004/033447 involves the reaction of ( R) – 6 , 8 -difluorochroman-3 -ylamine hydrochloride (the structure of ( R) -6, 8-difluorochroman-3 -ylamine is shown below as compound QA) , [4 – ( tert-butyldimethylsilanyloxy) -3 -oxobutyl] carbamic acid tert-butyl ester and potassium thiocyanate .

QA

(R) -6 , 8-difluorochroman- 3 -ylamine (compound QA) is a key intermediate in the synthesis of compound 1. The stereochemistry at the carbon atom to which the amine is attached gives rise to the stereochemistry of compound 1, so it is advantageous that compound QA is present in as pure enantiomeric form as possible. In other words, the (R) -enantiomer of compound QA should be in predominance, with little or no (S) enantiomer present. Thus, the process for preparing compound QA will advantageously produce compound QA with as high enantiomeric excess (ee) as possible.

Advantageous processes for preparing, for example, the compound of formula QA have now been found. In one aspect, the processes involve a biotransformation step. In another aspect, the processes involve chemical transformation. The processes may also be employed in the preparation of similar precursors useful in the production of other peripherally-selective inhibitors of dopamine -β -hydroxylase .

WO2008/136695 discloses a compound of formula YA, its (R) or (S) enantiomer, a mixture of its (R) and (S) enantiomers, or pharmaceutically acceptable salts thereof.

YA

The (R) -enantiomer of the compound of formula YA has been found to be a potent dopamines-hydroxylase inhibitor having high potency and significantly reduced brain access.

As disclosed in WO2008/136695 , the compound of formula YA may be prepared by reacting the compound of formula 1 with benzaldehyde under reductive alkylation conditions. In particular, (R) -5- (2 -aminoethyl ) -1- (6 , 8-difluorochroman-3 -yl) – 1 , 3 -dihydroimidazole-2 -thione and benzaldehyde may be reacted in the presence of a solvent or mixture of solvents, and a reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride .

process comprises the following steps:

The route from 2 , 4-difluorophenol may be as described 9/064210.

Preferably, the reagents and conditions are:

(i) H2S04, acetic acid

(ii) NaOCl, MeOH/water

(iii) Ru-based catalyst, H2, 30 bars, MeOH

(iv) aqueous KOH, MeOH, L-tartaric acid

(v) KSCN, AcOH/lPA

(vi) NaBH4, BF3.THF complex, THF then IPA

n one aspect, the process comprises the following steps

i. KOH, Thioglycolic acid or cysteine

ii. MEK

According to an aspect of the present invention, there is provided the following 2 -part synthetic route from the starting material 2 , 4 -difluorophenol to (R) -5- (2 -aminoethyl ) -1- (6 , 8-difluorochroman-3 -yl) -1 , 3 -dihydroimidazole-2 – thione

hydrochloride :

Part (1)

Preferred reagents and conditions:

a) HMTA, CF3COOH, 115°C, 18 hours

b) CH2CHCN, DABCO, DMF, water, 70°C, 16 hours

c) H2S04, AcOH, 100°C, 1 hour

d) NaClO, NaOH, MeOH, 25°C, 24 hours

e) (R) -C3 -TunePhosRu (acac) 2 S/C 3000, 30 bar H2, MeOH, 80°C, 20 hours

f) Water, 2-propanol, reflux to 20°C

g) 40% KOH, MeOH, reflux, 24 hours

h) L-tartaric acid, ethanol, water, RT, 1 hour

Part (2)

Preferred reagents and conditions

a’) methyl vinyl ketone, t-BuONa, EtOAc, EtOH, 40-50°C, 2-3 hours

Br2, MeOH, 20-25°C, 5 hours

water, reflux, 1 hour

KOH, AcOH, reflux, 1 hour

HCl, water, 2-propanol, 75 °C, 4 hours

KSCN, AcOH, 100°C, 2-4 hours

NaHC03, water, EtOH

NaBH4, 2-propanol, THF, water, 20-25°C, 16 hours

HCl, 2-propanol, water, reflux, 1-2 hours

The ( R ) -5- (2-Aminoethyl) -1- (6, 8-difluorochroman-3 -yl) -1,3-dihydroimidazole-2 – thione hydrochloride may then be used to

prepare (R) -5- (2- (benzylamino) ethyl) -1- (6, 8-difluorochroman-3 -yl) -lH-imidazole-2 (3H) -thione as follows.

Preferred reaction conditions/reagents:

q) NaBH(OAc)3, PhCHO, IPA;

t) NaOH, MeOH , H20

Either r) and s) :

r) HCI aq;

s) MeOH/Toluene;

Or n) , o) and p) :

n) HCI aq;

o) MeOH, toluene;

p) IPA.

EXAMPLES

Example 1

Nitro chromene synthesis

To 3 , 5-difluoro-2-hydroxybenzaldehyde (lOg, 63mmol, leq) , di-n-butylamine (4.1g, 32mmol, 0.5eq) , phtalic anhydride (18.7g, 126mmol, 2eq) in toluene (500mL) was added nitroethanol (5.75g, 63mmol, leq) . The round bottomed flask fitted with a dean stark apparatus was refluxed for 18h. The mixture was cooled and nitroethanol (5.75g, 63mmol, leq) was added. The resulting reaction mixture was then reflux for 12h. After cooling, the solution was evaporated down to approximately 150mL and purified over silica gel (eluent ethyl acetate : hexane 1:1) this gave several fractions that contained only the product by TLC, these was evaporated under reduced pressure to yield 1.8g which was 100% pure by HPLC aera. Several more fractions were collected containing a mixture of product and starting material. These were combined and washed with 2% NaOH solution (2x50mL) to remove starting material. The organic layer was washed with water (50mL) , dried over sodium sulfate and evaporated under reduced pressure to give 2.49g of brown solid ( 100% pure by HPLC aera) . More fractions were collected. These were combined, washed with 2% NaOH solution (3xl00mL) , water (lOOmL) and dried over sodium sulfate. This was then filtered and evaporated down in vacuum to yield 6.14g of a brown solid which was 91.3% pure by HPLC aera. 6 , 8 -difluoro-3 -nitro-2H-chromene (9.90g, 73.4%) was obtained as a brown solid.

Example 2

Nitro chromene synthesis with column purification

To a solution of isobenzofuran-1 , 3 -dione (4,68 g, 31,6 mmol) , 3 , 5-difluoro-2 -hydroxybenzaldehyde (2,5 g, 15,81 mmol) in Toluene (25 ml) was added 2 -nitroethanol (2,88 g, 31,6 mmol). The resulting mixture was heated to reflux overnight (Dean stark) .

The reaction conversion was checked by TLC (eluent PE/EtOAc 9:1) . A yellow spot was observed and corresponds to the expected product .

Reaction was cooled to room temperature and a plug of silica gel was performed. A pale brown solid (3.9g) was obtained. “””H-NMR showed presence of product and starting material. The solid was dissolved in diethylether and the organic layer was washed with aqueous sodium carbonate, dried over Na2S04, filtered and concentrated under reduced pressure. A pale brown solid (1.7g,) was obtained. The 1H-NMR was indicated no starting material but still polymer from nitroethanol and residue of phtalic anhydride. A second silica plug (eluent: PE/EtOAc 95:5) was done. A pale yellow solid (1.5g) was obtained. 1H-NMR of solid showed only product and polymer. The solid was recrystallized from methanol/water . A pale yellow solid (1.05g, 31.2%) was obtained.

Example 3

Nitro chromene synthesis without column purification

To a solution of isobenzofuran- 1 , 3 -dione (18,74 g, 127 mmol) , 3 , 5-difluoro-2 -hydroxybenzaldehyde (10 g, 63,3 mmol) in Toluene (100 ml) was added 2 -nitroethanol (6,86 ml, 95 mmol) . The resulting mixture was heated to reflux for 24h (Dean stark) .

The reaction conversion was checked by HPLC and by 1H-NMR. Only 50% conversion was obtained.

The reaction mixture was cooled to room temperature and diluted with DCM (lOOmL) and 1M NaOH solution (200mL) .

The biphasic system was stirred for 30 minutes and then separated (very difficult to see phase separation) . The aqueous layer was washed with DCM (50mL) and the combined organic layers were washed twice with water (2x50ml) , dried over sodium sulfate. The filtered organic layer was concentrated under reduced pressure. To the residue was added methanol (50mL) . The methanol was then removed by distillation under reduced pressure. A brown solution precipitated when most of the methanol was removed. More methanol was added and more solid crushed out then few drops of water was added to increase the product precipitation. The brown slurry was stirred for 30 minutes and filtered. The brown solid was washed with methanol/water (1:9, 5mL) and dried in a vacuum oven at 40°C for 12h.6, 8-difluoro-3 -nitro-2H-chroraene (4,9 g, 22,99 mmol,) was obtained as brown solid in 36.3% yield.

HPLC showed a purity of 98% and 1H-NMR confirmed the structure and purity around 95%

Example 4

Reduction of nitro chromene to nitro-alkane (racemic mixture)

To a suspension of 6 , 8 -difluoro-3 -nitro-2H-chromene (213mg, 0,999 mmol) and silica (0,8 g, 0,999 mmol) in a mixture of CHC13 (10 ml) and IPA (3,4 ml) at 0°C was added portion wise sodium borohydride (95 mg, 2,498 mmol). The resulting mixture was stirred at 0°C for 45 minutes. Reaction conversion was checked by HPLC. 1 mL of acetic acid was added at 0°C and the resulting mixture was stirred for 30 minutes at room temperature. The slurry was filtered and the silica was washed with DCM. The filtrate was diluted with ethyl acetate and water and the biphasic system was separated. The aqueous layer was back extracted with ethyl acetate. The combined organic layers were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure.

6 , 8-difluoro-3 -nitrochroman (196mg, 0,911 mmol, 91 % yield) was obtained as a pale yellow oil.

Example 5

Preparation of 6 , 8 -difluorochroman-3 -one from nitro chromene

A solution of 6, 8-difluoro-3 -nitro-2H-chromene (lOOmg, 0,469 mmol) in acetic acid (0.5 ml) is added slowly to a stirred slurry of iron (262 mg, 4,69 mmol) in acetic acid (1 ml) at 60.deg. C. The reaction mixture is stirred at 60. °C for 2 hour then allowed to cool to room temperature and stirred overnight. The reaction mixture is poured onto ice-water (30 ml) and filtered through Celite. The solid was wash with dichloromethane (DCM) (50 ml) . The organic portion is separated and washed with water (2 x 30 ml) and brine (30 ml) , dried over MgS04, filtered and concentrated in vacuo to give a brown oil. 6,8-difluorochroman-3 -one (75 mg, 0,407 mmol, 87 % yield) was obtained as a brown oil.

Example 6

Preparation of 6 , 8-difluorochroman-3 -one from methyl 6,8-difluoro-2H-chromen-3 -yl-carbamate

Methanol (1000m ml) was added to a slurry of methyl fluoro-2H-chromen-3 -yl -carbamate (250 g, 1.037 mol) hydrogen chloride 6N (2000 ml, 12 mol) at room temperature. The resulting mixture was reflux and stirred for 2 hours. Reaction monitored by HPLC.

Reaction was not complete but was stopped in order to avoid degradation of the product. The yellow solution was cooled to room temperature. A slurry (two type of solid) was observed and diluted with diethyl ether (300mL) . The resulting slurry was stirred at 5°C for 1 hour then filtered. The yellow solid was washed with water. The resulting wet yellow solid was suspended in diethylether (400mL) and petroleum ether (PE) (400mL) was added. Slight yellow solid was stirred at room temperature overnight, filtered and washed with PE (300mL) , dried in a vacuum oven at 30 °C for 4h. The wet sample was checked by NMR. No starting material was detected. A pale yellow solid (72.5g, solid 1) was obtained. The mother liquors were concentrated to dryness. A yellow solid was obtained, suspended in diethyl ether and PE. The slurry was then stirred for 4 hours, filtered, washed with PE . A dark yellow solid (4.5g, solid 2) was obtained. Solid 1 (2g) was diluted in DCM and washed with water (pH =6). The organic layer was then dried over Na2S04, filtered, concentrated to dryness. A crystalline pale yellow solid (1.9g, solid 3) was obtained. NMR showed the same purity for solid 3 as for solid 1. The remaining part of solid 1 was then diluted in DCM. The resulting organic layer was washed with water, dried over Na2S04, filtered and then concentrated to dryness. Slight yellow crystalline solid (68.5g, solid 4) was obtained. NMR confirmed high quality material.

Loss on Drying (LOD) : 1.03% .

Example 7

Biotransformation: Transaminases

Codexis transaminases ATA-025, ATA-251 and ATA-P2-A07 recognized 6 , 8 -difluorochroman-3 -one as the substrate and produced the corresponding 6 , 8 -difluorochroman-3 -amine .

 

 

PATENT

WO 2014077715

WO 2013002660

WO 2008136695

REFERNCES

International Journal of Pharmaceutics (Amsterdam, Netherlands) (2016), 501(1-2), 102-111.

WO2012087174A2 Dec 21, 2011 Jun 28, 2012 BIAL – PORTELA & Cª., S.A. Crystalline forms and processes for their preparation
WO2012087174A3 * Dec 21, 2011 May 10, 2013 BIAL – PORTELA & Cª., S.A. Crystalline forms and processes for their preparation
WO2013002660A2 Jun 29, 2012 Jan 3, 2013 BIAL – PORTELA & Cª, S.A. Process
WO2014077715A1 * Nov 14, 2013 May 22, 2014 BIAL – PORTELA & Cª, S.A. 1,3-dihydroimidazole-2-thione derivatives for use in the treatment of pulmonary arterial hypertension and lung injury
US8481582 May 6, 2008 Jul 9, 2013 Bial-Portela & Ca, S.A. 1,3-dihydroimidazole-2-thione derivatives as inhibitors of dopamine-beta-hydroxylase
US8865913 Jun 19, 2013 Oct 21, 2014 Bial-Portela & Ca, S.A. Crystalline forms and processes for their preparation
WO1995007284A1 * Aug 29, 1994 Mar 16, 1995 Smithkline Beecham Plc Phosphinic acid derivatives with anti-hyper glycemic and/or anti-obesity activity
WO2006044293A2 * Oct 11, 2005 Apr 27, 2006 Pharmacopeia Drug Discovery, Inc. Bicyclic compounds as selective melanin concentrating hormone receptor antagonists for the treatment of obesity and related disorders
WO2012007548A1 * Jul 14, 2011 Jan 19, 2012 Dsm Ip Assets B.V. (r)-selective amination
WO2013002660A2 * Jun 29, 2012 Jan 3, 2013 BIAL – PORTELA & Cª, S.A. Process
GR1005093B * Title not available

///////Zamicastat, BIA-5-1058, dopamine beta-monooxygenase inhibitor, phase I,  clinical studies, BIAL,  treatment of hypertension , heart failure.

S=C4NC=C(CCNCc1ccccc1)N4[C@@H]2Cc3cc(F)cc(F)c3OC2

Share

Etamicastat

 phase 2, Uncategorized  Comments Off on Etamicastat
Apr 122016
 

img

Etamicastat HCl salt
CAS: 677773-32-9 (HCl salt)

CAS 760173-05-5 (free base).
Chemical Formula: C14H16ClF2N3OS
Molecular Weight: 347.8088

Synonym: BIA 5-453; BIA5-453; BIA-5-453; Etamicastat

IUPAC/Chemical Name: (R)-5-(2-aminoethyl)-1-(6,8-difluorochroman-3-yl)-1,3-dihydro-2H-imidazole-2-thione hydrochloride

5-(2-Aminoethyl)-1-((3R)-6,8-difluoro-3,4-dihydro-2H-chromen-3-yl)-1,3-dihydro-2h-imidazole-2-thione

R)-5-(2-aminoethyl)-1-(6,8-difluorochroman-3-yl)-1,3-dihydroimidazole-2-thione hydrochloride,

PHASE 2, Treatment of Heart Failure Therapy, Hypertension

Bial-Portela and Ca, S.A

is a novel peripherally selective dopamine β-hydroxylase (DBH) inhibitor being developed by Bial-Portela and Ca, S.A. for treatment of hypertension and congestive heart failure.(1) The compound was shown to be well tolerated in healthy volunteers.

Etamicastat, also known as BIA 5-453, is a potent, reversible, peripherally selective dopamine β-hydroxylase inhibitor (DBH inhibitor). Chronic dopamine ß-hydroxylase inhibition with etamicastat effectively decreases blood pressure, although does not prevent the development of hypertension in the spontaneously hypertensive rat.

Figure

aReagents and conditions: a) Boc2O, EtOH, rt, 2 h; b) TBDMS-Cl, Et3N, DMAP, DCM, rt, 18 h; c) Dess–Martin periodinane, DCM, rt, 1 h; d) 2, KSCN, AcOH, EtOAc, reflux, 7 h; e) 2 N HCl, EtOAc, rt, 2 h.

 

 

Paper

Development of the Asymmetric Hydrogenation Step for Multikilogram Production of Etamicastat

Laboratory of Chemistry, Department of Research & Development, BIAL, 4745-457 S. Mamede do Coronado, Portugal
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00041
Publication Date (Web): March 21, 2016
Copyright © 2016 American Chemical Society
*Tel: 351-22-9866100. Fax: 351-22-9866192. E-mail: alexander.beliaev@bial.com.
Abstract Image

The asymmetric hydrogenation of methyl (6,8-difluoro-2H-chromen-3-yl)carbamate is a key step in the manufacturing route to etamicastat. A development of this step including the ruthenium or rhodium catalyst screening and the influence of the catalyst preparation (isolated, preformed in solution or in situ), solvent, temperature, pressure, additive, and concentration on the performance of the given ligand was discussed. Scale-up experiments for the best catalysts under optimized conditions were described.

 2D chemical structure of 760173-05-5

 

 PAPER

Synthesis and biological evaluation of novel, peripherally selective chromanyl imidazolethione-based inhibitors of dopamine beta-hydroxylase
J Med Chem 2006, 49(3): 1191
PATENT

in the processes .

(J?) -5- (2-Aminoethyl) -1- (6, 8-difluorochroman-3-yl) -1, 3-dihydroimidazole-2 -thione hydrochloride (the compound of formula 1, below) is a potent, non-toxic and peripherally selective inhibitor of ϋβΗ, which can be used for treatment of certain cardiovascular disorders. Compound 1 is disclosed in WO2004/033447 , along with processes for its preparation.

1

The process disclosed in WO2004/033447 involves the reaction of ( R) – 6 , 8 -difluorochroman-3 -ylamine hydrochloride (the structure of ( R) -6, 8-difluorochroman-3 -ylamine is shown below as compound QA) , [4 – ( tert-butyldimethylsilanyloxy) -3 -oxobutyl] carbamic acid tert-butyl ester and potassium thiocyanate .

QA

(R) -6 , 8-difluorochroman- 3 -ylamine (compound QA) is a key intermediate in the synthesis of compound 1. The stereochemistry at the carbon atom to which the amine is attached gives rise to the stereochemistry of compound 1, so it is advantageous that compound QA is present in as pure enantiomeric form as possible. In other words, the (R) -enantiomer of compound QA should be in predominance, with little or no (S) enantiomer present. Thus, the process for preparing compound QA will advantageously produce compound QA with as high enantiomeric excess (ee) as possible.

Advantageous processes for preparing, for example, the compound of formula QA have now been found. In one aspect, the processes involve a biotransformation step. In another aspect, the processes involve chemical transformation. The processes may also be employed in the preparation of similar precursors useful in the production of other peripherally-selective inhibitors of dopamine -β -hydroxylase .

WO2008/136695 discloses a compound of formula YA, its (R) or (S) enantiomer, a mixture of its (R) and (S) enantiomers, or pharmaceutically acceptable salts thereof.

YA

The (R) -enantiomer of the compound of formula YA has been found to be a potent dopamines-hydroxylase inhibitor having high potency and significantly reduced brain access.

As disclosed in WO2008/136695 , the compound of formula YA may be prepared by reacting the compound of formula 1 with benzaldehyde under reductive alkylation conditions. In particular, (R) -5- (2 -aminoethyl ) -1- (6 , 8-difluorochroman-3 -yl) – 1 , 3 -dihydroimidazole-2 -thione and benzaldehyde may be reacted in the presence of a solvent or mixture of solvents, and a reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride .

The compound of formula W may be prepared using a process as disclosed herein from the nitro chromene compound M.

The compound of formula WA may also be prepared using a process comprising bromination of 2 , 4 -difluorophenol to give bromophenol, alkylation of bromophenol with 4 -chloro-3 -oxo butanoate to give ketone followed by cyclization and decarboxylation to produce compound WA.

WA

According to an aspect of the present invention, there is provided the following 2 -part synthetic route from the starting material 2 , 4 -difluorophenol to (R) -5- (2 -aminoethyl ) -1- (6 , 8-difluorochroman-3 -yl) -1 , 3 -dihydroimidazole-2 – thione

hydrochloride :

Part (1)

Preferred reagents and conditions:

a) HMTA, CF3COOH, 115°C, 18 hours

b) CH2CHCN, DABCO, DMF, water, 70°C, 16 hours

c) H2S04, AcOH, 100°C, 1 hour

d) NaClO, NaOH, MeOH, 25°C, 24 hours

e) (R) -C3 -TunePhosRu (acac) 2 S/C 3000, 30 bar H2, MeOH, 80°C, 20 hours

f) Water, 2-propanol, reflux to 20°C

g) 40% KOH, MeOH, reflux, 24 hours

h) L-tartaric acid, ethanol, water, RT, 1 hour

Part (2)

Preferred reagents and conditions

a’) methyl vinyl ketone, t-BuONa, EtOAc, EtOH, 40-50°C, 2-3 hours

Br2, MeOH, 20-25°C, 5 hours

water, reflux, 1 hour

KOH, AcOH, reflux, 1 hour

HCl, water, 2-propanol, 75 °C, 4 hours

KSCN, AcOH, 100°C, 2-4 hours

NaHC03, water, EtOH

NaBH4, 2-propanol, THF, water, 20-25°C, 16 hours

HCl, 2-propanol, water, reflux, 1-2 hours

The ( R ) -5- (2-Aminoethyl) -1- (6, 8-difluorochroman-3 -yl) -1,3-dihydroimidazole-2 – thione hydrochloride

EXAMPLES

Example 1

Nitro chromene synthesis

To 3 , 5-difluoro-2-hydroxybenzaldehyde (lOg, 63mmol, leq) , di-n-butylamine (4.1g, 32mmol, 0.5eq) , phtalic anhydride (18.7g, 126mmol, 2eq) in toluene (500mL) was added nitroethanol (5.75g, 63mmol, leq) . The round bottomed flask fitted with a dean stark apparatus was refluxed for 18h. The mixture was cooled and nitroethanol (5.75g, 63mmol, leq) was added. The resulting reaction mixture was then reflux for 12h. After cooling, the solution was evaporated down to approximately 150mL and purified over silica gel (eluent ethyl acetate : hexane 1:1) this gave several fractions that contained only the product by TLC, these was evaporated under reduced pressure to yield 1.8g which was 100% pure by HPLC aera. Several more fractions were collected containing a mixture of product and starting material. These were combined and washed with 2% NaOH solution (2x50mL) to remove starting material. The organic layer was washed with water (50mL) , dried over sodium sulfate and evaporated under reduced pressure to give 2.49g of brown solid ( 100% pure by HPLC aera) . More fractions were collected. These were combined, washed with 2% NaOH solution (3xl00mL) , water (lOOmL) and dried over sodium sulfate. This was then filtered and evaporated down in vacuum to yield 6.14g of a brown solid which was 91.3% pure by HPLC aera. 6 , 8 -difluoro-3 -nitro-2H-chromene (9.90g, 73.4%) was obtained as a brown solid.

Example 2

Nitro chromene synthesis with column purification

To a solution of isobenzofuran-1 , 3 -dione (4,68 g, 31,6 mmol) , 3 , 5-difluoro-2 -hydroxybenzaldehyde (2,5 g, 15,81 mmol) in Toluene (25 ml) was added 2 -nitroethanol (2,88 g, 31,6 mmol). The resulting mixture was heated to reflux overnight (Dean stark) .

The reaction conversion was checked by TLC (eluent PE/EtOAc 9:1) . A yellow spot was observed and corresponds to the expected product .

Reaction was cooled to room temperature and a plug of silica gel was performed. A pale brown solid (3.9g) was obtained. “””H-NMR showed presence of product and starting material. The solid was dissolved in diethylether and the organic layer was washed with aqueous sodium carbonate, dried over Na2S04, filtered and concentrated under reduced pressure. A pale brown solid (1.7g,) was obtained. The 1H-NMR was indicated no starting material but still polymer from nitroethanol and residue of phtalic anhydride. A second silica plug (eluent: PE/EtOAc 95:5) was done. A pale yellow solid (1.5g) was obtained. 1H-NMR of solid showed only product and polymer. The solid was recrystallized from methanol/water . A pale yellow solid (1.05g, 31.2%) was obtained.

Example 3

Nitro chromene synthesis without column purification

To a solution of isobenzofuran- 1 , 3 -dione (18,74 g, 127 mmol) , 3 , 5-difluoro-2 -hydroxybenzaldehyde (10 g, 63,3 mmol) in Toluene (100 ml) was added 2 -nitroethanol (6,86 ml, 95 mmol) . The resulting mixture was heated to reflux for 24h (Dean stark) .

The reaction conversion was checked by HPLC and by 1H-NMR. Only 50% conversion was obtained.

The reaction mixture was cooled to room temperature and diluted with DCM (lOOmL) and 1M NaOH solution (200mL) .

The biphasic system was stirred for 30 minutes and then separated (very difficult to see phase separation) . The aqueous layer was washed with DCM (50mL) and the combined organic layers were washed twice with water (2x50ml) , dried over sodium sulfate. The filtered organic layer was concentrated under reduced pressure. To the residue was added methanol (50mL) . The methanol was then removed by distillation under reduced pressure. A brown solution precipitated when most of the methanol was removed. More methanol was added and more solid crushed out then few drops of water was added to increase the product precipitation. The brown slurry was stirred for 30 minutes and filtered. The brown solid was washed with methanol/water (1:9, 5mL) and dried in a vacuum oven at 40°C for 12h.6, 8-difluoro-3 -nitro-2H-chroraene (4,9 g, 22,99 mmol,) was obtained as brown solid in 36.3% yield.

HPLC showed a purity of 98% and 1H-NMR confirmed the structure and purity around 95%

Example 4

Reduction of nitro chromene to nitro-alkane (racemic mixture)

To a suspension of 6 , 8 -difluoro-3 -nitro-2H-chromene (213mg, 0,999 mmol) and silica (0,8 g, 0,999 mmol) in a mixture of CHC13 (10 ml) and IPA (3,4 ml) at 0°C was added portion wise sodium borohydride (95 mg, 2,498 mmol). The resulting mixture was stirred at 0°C for 45 minutes. Reaction conversion was checked by HPLC. 1 mL of acetic acid was added at 0°C and the resulting mixture was stirred for 30 minutes at room temperature. The slurry was filtered and the silica was washed with DCM. The filtrate was diluted with ethyl acetate and water and the biphasic system was separated. The aqueous layer was back extracted with ethyl acetate. The combined organic layers were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure.

6 , 8-difluoro-3 -nitrochroman (196mg, 0,911 mmol, 91 % yield) was obtained as a pale yellow oil.

Example 5

Preparation of 6 , 8 -difluorochroman-3 -one from nitro chromene

A solution of 6, 8-difluoro-3 -nitro-2H-chromene (lOOmg, 0,469 mmol) in acetic acid (0.5 ml) is added slowly to a stirred slurry of iron (262 mg, 4,69 mmol) in acetic acid (1 ml) at 60.deg. C. The reaction mixture is stirred at 60. °C for 2 hour then allowed to cool to room temperature and stirred overnight. The reaction mixture is poured onto ice-water (30 ml) and filtered through Celite. The solid was wash with dichloromethane (DCM) (50 ml) . The organic portion is separated and washed with water (2 x 30 ml) and brine (30 ml) , dried over MgS04, filtered and concentrated in vacuo to give a brown oil. 6,8-difluorochroman-3 -one (75 mg, 0,407 mmol, 87 % yield) was obtained as a brown oil.

Example 6

Preparation of 6 , 8-difluorochroman-3 -one from methyl 6,8-difluoro-2H-chromen-3 -yl-carbamate

Methanol (1000m ml) was added to a slurry of methyl fluoro-2H-chromen-3 -yl -carbamate (250 g, 1.037 mol) hydrogen chloride 6N (2000 ml, 12 mol) at room temperature. The resulting mixture was reflux and stirred for 2 hours. Reaction monitored by HPLC.

Reaction was not complete but was stopped in order to avoid degradation of the product. The yellow solution was cooled to room temperature. A slurry (two type of solid) was observed and diluted with diethyl ether (300mL) . The resulting slurry was stirred at 5°C for 1 hour then filtered. The yellow solid was washed with water. The resulting wet yellow solid was suspended in diethylether (400mL) and petroleum ether (PE) (400mL) was added. Slight yellow solid was stirred at room temperature overnight, filtered and washed with PE (300mL) , dried in a vacuum oven at 30 °C for 4h. The wet sample was checked by NMR. No starting material was detected. A pale yellow solid (72.5g, solid 1) was obtained. The mother liquors were concentrated to dryness. A yellow solid was obtained, suspended in diethyl ether and PE. The slurry was then stirred for 4 hours, filtered, washed with PE . A dark yellow solid (4.5g, solid 2) was obtained. Solid 1 (2g) was diluted in DCM and washed with water (pH =6). The organic layer was then dried over Na2S04, filtered, concentrated to dryness. A crystalline pale yellow solid (1.9g, solid 3) was obtained. NMR showed the same purity for solid 3 as for solid 1. The remaining part of solid 1 was then diluted in DCM. The resulting organic layer was washed with water, dried over Na2S04, filtered and then concentrated to dryness. Slight yellow crystalline solid (68.5g, solid 4) was obtained. NMR confirmed high quality material.

Loss on Drying (LOD) : 1.03% .

Example 7

Biotransformation: Transaminases

Codexis transaminases ATA-025, ATA-251 and ATA-P2-A07 recognized 6 , 8 -difluorochroman-3 -one as the substrate and produced the corresponding 6 , 8 -difluorochroman-3 -amine .

PATENT
WO 2004033447
WO 2008094056
WO 2008143540
WO 2009064210

References

1: Igreja B, Wright LC, Soares-da-Silva P. Sustained high blood pressure reduction with etamicastat, a peripheral selective dopamine β-hydroxylase inhibitor. J Am Soc Hypertens. 2015 Dec 19. pii: S1933-1711(15)00838-4. doi: 10.1016/j.jash.2015.12.011. [Epub ahead of print] PubMed PMID: 26803288.

2: Loureiro AI, Bonifácio MJ, Fernandes-Lopes C, Pires N, Igreja B, Wright LC, Soares-da-Silva P. Role of P-glycoprotein and permeability upon the brain distribution and pharmacodynamics of etamicastat: a comparison with nepicastat. Xenobiotica. 2015;45(9):828-39. doi: 10.3109/00498254.2015.1018985. Epub 2015 Jun 10. PubMed PMID: 25915108.

3: Loureiro AI, Soares-da-Silva P. Distribution and pharmacokinetics of etamicastat and its N-acetylated metabolite (BIA 5-961) in dog and monkey. Xenobiotica. 2015;45(10):903-11. doi: 10.3109/00498254.2015.1024780. Epub 2015 Apr 14. PubMed PMID: 25869244.

4: Pires NM, Igreja B, Moura E, Wright LC, Serrão MP, Soares-da-Silva P. Blood pressure decrease in spontaneously hypertensive rats folowing renal denervation or dopamine β-hydroxylase inhibition with etamicastat. Hypertens Res. 2015 Sep;38(9):605-12. doi: 10.1038/hr.2015.50. Epub 2015 Apr 9. PubMed PMID: 25854989.

5: Bonifácio MJ, Sousa F, Neves M, Palma N, Igreja B, Pires NM, Wright LC, Soares-da-Silva P. Characterization of the interaction of the novel antihypertensive etamicastat with human dopamine-β-hydroxylase: comparison with nepicastat. Eur J Pharmacol. 2015 Mar 15;751:50-8. doi: 10.1016/j.ejphar.2015.01.034. Epub 2015 Jan 29. PubMed PMID: 25641750.

6: Pires NM, Loureiro AI, Igreja B, Lacroix P, Soares-da-Silva P. Cardiovascular safety pharmacology profile of etamicastat, a novel peripheral selective dopamine-β-hydroxylase inhibitor. Eur J Pharmacol. 2015 Mar 5;750:98-107. doi: 10.1016/j.ejphar.2015.01.035. Epub 2015 Jan 30. PubMed PMID: 25641747.

7: Igreja B, Pires NM, Bonifácio MJ, Loureiro AI, Fernandes-Lopes C, Wright LC, Soares-da-Silva P. Blood pressure-decreasing effect of etamicastat alone and in combination with antihypertensive drugs in the spontaneously hypertensive rat. Hypertens Res. 2015 Jan;38(1):30-8. doi: 10.1038/hr.2014.143. Epub 2014 Oct 9. PubMed PMID: 25298210.

8: Loureiro AI, Bonifácio MJ, Fernandes-Lopes C, Igreja B, Wright LC, Soares-da-Silva P. Etamicastat, a new dopamine-ß-hydroxylase inhibitor, pharmacodynamics and metabolism in rat. Eur J Pharmacol. 2014 Oct 5;740:285-94. doi: 10.1016/j.ejphar.2014.07.027. Epub 2014 Jul 21. PubMed PMID: 25058908.

9: Almeida L, Nunes T, Costa R, Rocha JF, Vaz-da-Silva M, Soares-da-Silva P. Etamicastat, a novel dopamine β-hydroxylase inhibitor: tolerability, pharmacokinetics, and pharmacodynamics in patients with hypertension. Clin Ther. 2013 Dec;35(12):1983-96. doi: 10.1016/j.clinthera.2013.10.012. Epub 2013 Dec 2. PubMed PMID: 24296323.

10: Loureiro AI, Rocha JF, Fernandes-Lopes C, Nunes T, Wright LC, Almeida L, Soares-da-Silva P. Human disposition, metabolism and excretion of etamicastat, a reversible, peripherally selective dopamine β-hydroxylase inhibitor. Br J Clin Pharmacol. 2014 Jun;77(6):1017-26. doi: 10.1111/bcp.12274. PubMed PMID: 24168152; PubMed Central PMCID: PMC4093927.

11: Loureiro AI, Fernandes-Lopes C, Bonifácio MJ, Wright LC, Soares-da-Silva P. N-acetylation of etamicastat, a reversible dopamine-β-hydroxylase inhibitor. Drug Metab Dispos. 2013 Dec;41(12):2081-6. doi: 10.1124/dmd.113.053736. Epub 2013 Sep 6. PubMed PMID: 24013186.

12: Nunes T, Rocha JF, Vaz-da-Silva M, Falcão A, Almeida L, Soares-da-Silva P. Pharmacokinetics and tolerability of etamicastat following single and repeated administration in elderly versus young healthy male subjects: an open-label, single-center, parallel-group study. Clin Ther. 2011 Jun;33(6):776-91. doi: 10.1016/j.clinthera.2011.05.048. PubMed PMID: 21704242.

13: Vaz-da-Silva M, Nunes T, Rocha JF, Falcão A, Almeida L, Soares-da-Silva P. Effect of food on the pharmacokinetic profile of etamicastat (BIA 5-453). Drugs R D. 2011;11(2):127-36. doi: 10.2165/11587080-000000000-00000. PubMed PMID: 21548660; PubMed Central PMCID: PMC3585837.

14: Rocha JF, Vaz-Da-Silva M, Nunes T, Igreja B, Loureiro AI, Bonifácio MJ, Wright LC, Falcão A, Almeida L, Soares-Da-Silva P. Single-dose tolerability, pharmacokinetics, and pharmacodynamics of etamicastat (BIA 5-453), a new dopamine β-hydroxylase inhibitor, in healthy subjects. J Clin Pharmacol. 2012 Feb;52(2):156-70. doi: 10.1177/0091270010390805. PubMed PMID: 21343348.

15: Nunes T, Rocha JF, Vaz-da-Silva M, Igreja B, Wright LC, Falcão A, Almeida L, Soares-da-Silva P. Safety, tolerability, and pharmacokinetics of etamicastat, a novel dopamine-β-hydroxylase inhibitor, in a rising multiple-dose study in young healthy subjects. Drugs R D. 2010;10(4):225-42. doi: 10.2165/11586310-000000000-00000. PubMed PMID: 21171669; PubMed Central PMCID: PMC3585840.

16: Beliaev A, Learmonth DA, Soares-da-Silva P. Synthesis and biological evaluation of novel, peripherally selective chromanyl imidazolethione-based inhibitors of dopamine beta-hydroxylase. J Med Chem. 2006 Feb 9;49(3):1191-7. PubMed PMID: 16451083.

PATENT CITATIONS
Cited Patent Filing date Publication date Applicant Title
WO1995007284A1 * Aug 29, 1994 Mar 16, 1995 Smithkline Beecham Plc Phosphinic acid derivatives with anti-hyper glycemic and/or anti-obesity activity
WO2006044293A2 * Oct 11, 2005 Apr 27, 2006 Pharmacopeia Drug Discovery, Inc. Bicyclic compounds as selective melanin concentrating hormone receptor antagonists for the treatment of obesity and related disorders
WO2012007548A1 * Jul 14, 2011 Jan 19, 2012 Dsm Ip Assets B.V. (r)-selective amination
WO2013002660A2 * Jun 29, 2012 Jan 3, 2013 BIAL – PORTELA & Cª, S.A. Process
GR1005093B * Title not available
Reference
1 * AL NEIRABEYEH M. ET AL.: “Methoxy and hydroxy derivatives of 3,4-dihydro-3-(di-n-propylamino)-2H-1-benzopyrans: new synthesis and dopaminergic activity“, EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 26, no. 5, 1991, EDITIONS SCIENTIFIQUE ELSEVIER, PARIS; FR, pages 497 – 504, XP023870436, ISSN: 0223-5234, DOI: 10.1016/0223-5234(91)90145-D
2 * BELIAEV, A. ET AL.: “Process Research for Multikilogram Production of Etamicastat: A Novel Dopamine ß-Hydroxylase Inhibitor“, ORGANIC PROCESS RESEARCH & DEVELOPMENT, no. 16, 2012, American Chemical Society, Washington; US, pages 704 – 709, XP002731798, DOI: 10.1021/op300012d
3 * BOYE, S. ET AL.: “N,N-Disubstituted aminomethyl benzofuran derivatives: synthesis and preliminary binding evaluation“, BIOORGANIC & MEDICINAL CHEMISTRY, no. 7, 1999, ELSEVIER SCIENCE LTD; GB, pages 335 – 341, XP002731795, ISSN: 0968-0896, DOI: 10.1016/S0968-0896(98)00239-9
4 * COMOY, C. ET AL.: “3-Amino-3,4-dihydro-2H-1-benzopyran Derivatives as 5-HT1A Receptor Ligandsand Potential Anxiolytic Agents. 2. Synthesis and QuantitativeStructure-Activity Relationship Studies of Spiro[pyrrolidine- andpiperidine-2,3′(2’H)-benzopyrans]“, JOURNAL OF MEDICINAL CHEMISTRY., vol. 39, no. 21, 1996, AMERICAN CHEMICAL SOCIETY. WASHINGTON; US, pages 4285 – 4298, XP002731797, ISSN: 0022-2623, DOI: 10.1021/JM950861W
5 * SHIN, C. ET AL.: “Total Synthesis of Bistratamide G, a Metabolite of the PhilippinesAscidian Lissoclinum bistratum, from Dehydrotripeptides“, CHEMISTRY LETTERS, vol. 33, no. 6, 2004, Chemical Society of Japan, Tokyo; JP, pages 664 – 665, XP002731799, ISSN: 0366-7022, DOI: 10.1246/cl.2004.664
6 * VASSE, J. L. ET AL.: “New efficient conditions for the reduction with NADH models“, SYNLETT, October 1998 (1998-10-01), THIEME INTERNATIONAL, STUTTGART; DE, pages 1144 – 1146, XP002731796, ISSN: 0936-5214, DOI: 10.1055/s-1998-1876
7 * XIAO, G.-Q. ET AL.: “3-Nitro-2H-chromenes as a New Class of Inhibitors against Thioredoxin Reductase and Proliferation of Cancer Cells“, ARCHIV DER PHARMAZIE, no. 345, 2012, VCH VERLAGSGESELLSCHAFT MBH, WEINHEIM; DE, pages 767 – 770, XP002731794, ISSN: 0365-6233, DOI: 10.1002/ardp.201200121

////////Etamicastat, BIA-5-453 , PHASE 2, Treatment, Heart Failure Therapy, Hypertension, Bial-Portela and Ca, S.A

SMILES Code: FC1=CC(F)=C(OC[C@H](N2C(CCN)=CNC2=S)C3)C3=C1.[H]Cl

c1c(cc(c2c1C[C@H](CO2)n3c(c[nH]c3=S)CCN)F)F

Share

FDA approves new drug Venclexta (venetoclax) for chronic lymphocytic leukemia in patients with a specific chromosomal abnormality

 Uncategorized  Comments Off on FDA approves new drug Venclexta (venetoclax) for chronic lymphocytic leukemia in patients with a specific chromosomal abnormality
Apr 122016
 
Venetoclax.svg
Venclexta (venetoclax)
04/11/2016 12:12 PM EDT
The U.S. Food and Drug Administration today approved Venclexta (venetoclax) for the treatment of patients with chronic lymphocytic leukemia (CLL) who have a chromosomal abnormality called 17p deletion and who have been treated with a least one prior therapy. Venclexta is the first FDA-approved treatment that targets the B-cell lymphoma 2 (BCL-2) protein, which supports cancer cell growth and is overexpressed in many patients with CLL.

April 11, 2016

Release

The U.S. Food and Drug Administration today approved Venclexta (venetoclax) for the treatment of patients with chronic lymphocytic leukemia (CLL) who have a chromosomal abnormality called 17p deletion and who have been treated with at least one prior therapy. Venclexta is the first FDA-approved treatment that targets the B-cell lymphoma 2 (BCL-2) protein, which supports cancer cell growth and is overexpressed in many patients with CLL.

According to the National Cancer Institute, CLL is one of the most common types of leukemia in adults, with approximately 15,000 new cases diagnosed each year. CLL is characterized by the progressive accumulation of abnormal lymphocytes, a type of white blood cell. Patients with CLL who have a 17p deletion lack a portion of the chromosome that acts to suppress cancer growth. This chromosomal abnormality occurs in approximately 10 percent of patients with untreated CLL and in approximately 20 percent of patients with relapsed CLL.

“These patients now have a new, targeted therapy that inhibits a protein involved in keeping tumor cells alive,” said Richard Pazdur, director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research. “For certain patients with CLL who have not had favorable outcomes with other therapies, Venclexta may provide a new option for their specific condition.”

The efficacy of Venclexta was tested in a single-arm clinical trial of 106 patients with CLL who have a 17p deletion and who had received at least one prior therapy. Trial participants took Venclexta orally every day, beginning with 20 mg and increasing over a five-week period to 400 mg. Results showed that 80 percent of trial participants experienced a complete or partial remission of their cancer.

Venclexta is indicated for daily use after detection of 17p deletion is confirmed through the use of the FDA-approved companion diagnostic Vysis CLL FISH probe kit.

The most common side effects of Venclexta include low white blood cell count (neutropenia), diarrhea, nausea, anemia, upper respiratory tract infection, low platelet count (thrombocytopenia) and fatigue. Serious complications can include pneumonia, neutropenia with fever, fever, autoimmune hemolytic anemia, anemia and metabolic abnormalities known as tumor lysis syndrome. Live attenuated vaccines should not be given to patients taking Venclexta.

The FDA granted the Venclexta application breakthrough therapy designation, priority review status, and accelerated approval for this indication. These are distinct programs intended to facilitate and expedite the development and review of certain new drugs in light of their potential to benefit patients with serious or life-threatening conditions. Venclexta also received orphan drug designation, which provides incentives such as tax credits, user fee waivers and eligibility for exclusivity to assist and encourage the development of drugs for rare diseases.

Venclexta is manufactured by AbbVie Inc. of North Chicago, Illinois, and marketed by AbbVie and Genentech USA Inc. of South San Francisco, California. The Vysis CLL FISH probe kit is manufactured by Abbott Molecular of Des Plaines, Illinois.

///FDA,  approves,  new drug, chronic lymphocytic leukemia,  specific chromosomal abnormality, Venclexta, venetoclax,  fda 2016
Share

PF-05387552

 Uncategorized  Comments Off on PF-05387552
Apr 112016
 

str1

str1

STR3

CID 50992153.png

PF-05387552

IRAK4

CAS 1604034-71-0
C25 H27 N5 O2
11H-​Indolo[3,​2-​c]​quinoline-​9-​carbonitrile, 2-​methoxy-​3-​[3-​(4-​methyl-​1-​piperazinyl)​propoxy]​-
2-methoxy-3-[3-(4-methylpiperazin-1-yl)propoxy]-11H-indolo[3,2-c]quinoline-9-carbonitrile
Molecular Weight429.51
Molecular Formula: C25H27N5O2
Molecular Weight: 429.51418 g/mol

Synthesis

 

str1

PAPER

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

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

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

http://www.sciencedirect.com/science/article/pii/S0960894X14002832?np=y

Image for unlabelled figure

IRAK4 is responsible for initiating signaling from Toll-like receptors (TLRs) and members of the IL-1/18 receptor family. Kinase-inactive knock-ins and targeted deletions of IRAK4 in mice cause reductions in TLR induced pro-inflammatory cytokines and these mice are resistant to various models of arthritis.

Herein we report the identification and optimization of a series of potent IRAK4 inhibitors. Representative examples from this series showed excellent selectivity over a panel of kinases, including the kinases known to play a role in TLR-mediated signaling. The compounds exhibited low nM potency in LPS- and R848-induced cytokine assays indicating that they are blocking the TLR signaling pathway.

A key compound (26) from this series was profiled in more detail and found to have an excellent pharmaceutical profile as measured by predictive assays such as microsomal stability, TPSA, solubility, and c log P. However, this compound was found to afford poor exposure in mouse upon IP or IV administration. We found that removal of the ionizable solubilizing group (32) led to increased exposure, presumably due to increased permeability. Compounds 26 and 32, when dosed to plasma levels corresponding to ex vivo whole blood potency, were shown to inhibit LPS-induced TNFα in an in vivo murine model.

To our knowledge, this is the first published in vivo demonstration that inhibition of the IRAK4 pathway by a small molecule can recapitulate the phenotype of IRAK4 knockout mice.

L. Nathan TurneyL. Nathan Tumey, Ph.D., Principal Research Scientist, Pfizer Global R&D

 

REFERENCES

STR3

///////////TLR signaling, Indoloquinoline, IRAK4, Kinase inhibitor, Inflammation, PF-05387552, PF 05387552,  1604034-71-0

N#Cc3ccc4c5cnc2cc(OCCCN1CCN(C)CC1)c(OC)cc2c5nc4c3

 

 

सुकून उतना ही देना प्रभू, जितने से जिंदगी चल जाये। औकात बस इतनी देना, कि औरों का भला हो जाये।
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO …..FOR BLOG HOME CLICK HERE

Join me on Linkedin

View Anthony Melvin Crasto Ph.D's profile on LinkedIn

Join me on Facebook FACEBOOK

Join me on twitterFollow amcrasto on Twitter
Join me on google plus Googleplus
Join me on Researchgate

Anthony Melvin Crasto Dr.

 amcrasto@gmail.com

 

09b37-misc2b027LIONEL MY SON

He was only in first standard in school when I was hit by a deadly one in a million spine stroke called acute transverse mylitis, it made me 90% paralysed and bound to a wheel chair, Now I keep him as my source of inspiration and helping millions, thanks to millions of my readers who keep me going and help me to keep my son happy

सुकून उतना ही देना प्रभू, जितने से
जिंदगी चल जाये।
औकात बस इतनी देना,
कि औरों का भला हो जाये।
Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL  

Share

Debiopharm and Aurigene dual c-src / jak inhibitors

 Uncategorized  Comments Off on Debiopharm and Aurigene dual c-src / jak inhibitors
Apr 112016
 

SCHEMBL2237115.png

STR3str4

Debio 1142

Jak2 tyrosine kinase inhibitor; Src tyrosine kinase inhibitor

N-[4-methyl-3-[2-[4-(4-methylpiperazin-1-yl)anilino]-5-oxo-7,8-dihydropyrido[4,3-d]pyrimidin-6-yl]phenyl]-3-(trifluoromethyl)benzamide

Molecular Formula: C33H32F3N7O2
Molecular Weight: 615.64809 g/mol
1332328-01-4
Benzamide, N-​[3-​[7,​8-​dihydro-​2-​[[4-​(4-​methyl-​1-​piperazinyl)​phenyl]​amino]​-​5-​oxopyrido[4,​3-​d]​pyrimidin-​6(5H)​-​yl]​-​4-​methylphenyl]​-​3-​(trifluoromethyl)​-

Debiopharm S.A., Aurigene Discovery Technologies Ltd.

ALLISTER Andrès MC, Maximilien Murone,Saumitra Sengupta, Shankar Jayaram Shetty

https://www.google.co.in/patents/WO2011101806A1?cl=en

Bicyclic compounds and their uses as dual c-src / jak inhibitors

STR3

Apr. 14 /PR Newswire/ –Debiopharm and Aurigene Sign Agreement for the Development and Commercialisation of Debio 1142, a Novel Inhibitor of an Undisclosed Oncology Pathway

LAUSANNE, Switzerland and BANGALORE, India, April 14, 2011 /PRNewswire/ — Debiopharm Group(TM) (Debiopharm), a global biopharmaceutical development specialist that focuses on serious medical conditions and particularly oncology, and Aurigene Discovery Technologies Ltd (Aurigene), a Bangalore-based drug discovery company, signed on March 23, 2011 an option and exclusive worldwide license agreement concerning the development and commercialisation of Debio 1142, a novel inhibitor of an undisclosed oncology pathway.

“We are very excited about this new collaboration with Aurigene. Their business model offers a one stop solution for structure guided drug design, lead optimisation and preclinical work. The Debio 1142 project aims at developing inhibitors targeting a key oncology pathway, which plays essential roles in various solid tumours, including resistance to chemotherapy” said Dr Rolland-Yves Mauvernay, president and founder of Debiopharm S.A.

“Coming as it does after a successful collaboration programme we already had with Debiopharm, and as a continuation of our close to 5 year association, the relationship between Debiopharm and Aurigene demonstrates the strategic fit between organisations with complimentary scientific skills. We are happy that we have the opportunity to continue to work with Debiopharm, in a unique business model that has been tailor-made to meet each partners’ needs” added CSN Murthy, CEO of Aurigene.

About Debiopharm Group

Debiopharm Group(TM) (Debiopharm) is a Swiss-based global biopharmaceutical group of companies with a focus on the development of prescription drugs that target unmet medical needs. The group in-licenses, develops and/or co-develops promising biological and small molecule drug candidates having reached clinical development phases I, II or III as well as earlier stage candidates. It develops its products for global registration and maximum commercial potential. The products are out-licensed to pharmaceutical partners for sales and marketing. Debiopharm Group is also active in the field of companion diagnostics with a view to progressing in the area of personalised medicine. Debiopharm independently funds the worldwide development of all of its products while providing expertise in pre-clinical and clinical trials, manufacturing, drug delivery and formulation, and regulatory affairs. For more information on Debiopharm Group(TM), please visit: http://www.debiopharm.com.

About Aurigene

Aurigene Discovery Technologies Limited is a Bangalore-based biotech focused on collaborative drug discovery with pharmaceutical and biotech companies on a risk-sharing basis. Aurigene has fully integrated drug discovery infrastructure, from Target to IND, along with strong in house structural biology and fragment based drug design capabilities. The company is engaged in over 20 discovery collaborations with US and European large and mid-pharma companies in Oncology, Inflammatory disorders and anti-infectives. For more information on Aurigene, please visit: http://www.aurigene.com.

PATENT

WO 2011101806

http://www.google.co.in/patents/WO2011101806A1?cl=en

PAPER

Journal of Chemical and Pharmaceutical Research (2014), 6(4), 1146-1152

http://jocpr.com/vol6-iss4-2014/JCPR-2014-6-4-1146-1152.pdf

REFERENCES

INDIAN PATENTS

7554/CHENP/2012

415/CHE/2010

https://www.debiopharm.com/our-business/pipeline.html

http://www.giiresearch.com/report/labd315710-debiopharm-international-sa-product-pipeline.html

Patent ID Date Patent Title
US2013143895 2013-06-06 BICYCLIC COMPOUNDS AND THEIR USES AS DUAL C-SRC / JAK INHIBITORS
US8440679 2013-05-14 Bicyclic compounds and their uses as dual c-SRC / JAK inhibitors

///////////Debio 1142, Jak2 tyrosine kinase inhibitor,  Src tyrosine kinase inhibitor, Debio-1142, Debiopharm S.A.Aurigene Discovery Technologies Ltd, 1332328-01-4

c21cnc(nc1CCN(C2=O)c3c(ccc(c3)NC(=O)c4cc(ccc4)C(F)(F)F)C)Nc5ccc(cc5)N6CCN(CC6)C

Share

CN-128 for the treatment of thelassemia and iron overload

 Uncategorized  Comments Off on CN-128 for the treatment of thelassemia and iron overload
Apr 102016
 

Figure imgf000011_0002

STR3

CN-128

(R)-3-Hydroxy-1-(1-hydroxy-3-benzyl propyl-2-)2-methyl pyridine-4(1H)-one

IND Filing

CN-128 is potentially for the treatment of thelassemia and iron overload.

Zhejiang University, 浙江大学

CAS No. 1335282-04-6

C15 H17 N O3, 4(1H)​-​Pyridinone, 3-​hydroxy-​1-​[(1R)​-​1-​(hydroxymethyl)​-​2-​phenylethyl]​-​2-​methyl-
Molecular Weight, 259.30

Many diseases in humans and animals are caused by excessive accumulated metals, such as iron. Among such diseases, excess iron is accumulated in various tissues, which is called iron overload disorders, formerly known as siderosis Haemorrhagic. Excess iron has the following sources: 1) long-term blood transfusion; 2) the gastrointestinal system absorbing excess iron, because stimulated by diseases such as anemia. It is necessary to repeat transfusion for some patients with severe anemia, for example, β-thalassemia, as well as other anemia requiring transfusion therapy. Excessive iron absorption from the gastrointestinal tract usually occurs in hemochromatosis patients and in anemia patients who do not require blood transfusion, such as thalassemia intermedia. If iron overload disease is not treated, it will result in severe tissue damage, especially the liver, heart and endocrine organs, and ultimately lead to death. Iron chelators can remove and clear excess iron from such organs, relieve symptoms and reduce the corresponding mortality.

Desferrioxamine (DFO) is an effective iron chelator for a long time. However, in the treatment of the diseases mentioned above, the biggest disadvantage regarding DFO and its salts is its poor oral absorption capability. So, administration is achieved with a slow injection method (8∼12h/day), patients need to wear a portable drug delivery device during treatment, such as mounting the syringe on a mechanical pressing device. This method is inconvenient, and also expensive, which largely limits the utilization of DFO, especially for thalassemia-prone areas, such as Mediterranean, Middle East, and India &South East Asia, it plays no role in treatment of malaria in world-wide and sickle cell anemia in some African countries, which is a very serious problem to the populations there. Image loading...

UK Patent No. 2, 13, 807, US Patent No. 4, 585, 780 and other scientific research have reported the treatment of iron overload symptoms by using 3-hydroxypyridin-4-one derivatives, especially in some pathological symptoms, such as thalassemia, sickle cell anemia, aplastic anemia in children, and idiopathic hemochromatosis, usually, treatment of the first three diseases includes frequent regular blood transfusion. 3-Hydroxypyridin-4-one derivatives, especially CP20 (commercial named Ferriprox) is employed to treat systemic iron overload disorders, and also to treat certain diseases associated with local iron overload distribution, although such patients do not show symptoms of systemic iron overload, i.e. inhibition free radical mediated reactions caused by excess iron ions in certain neurodegenerative diseases and cancer diseases. A serious limitation of CP20 is that the hydroxyl group at 3′ position is vulnerable to glycosylation, which reduces the half-life of this compound (approximately 2∼3 h). So it requires a high dosage, which is associated with obvious side effects. Image loading...

EP0120669 discloses compounds with a 3-hydroxypyrid-4-one in which the H attached to the N atom is substituted by an aliphatic acyl group, or an aliphatic hydrocarbon group, these groups can be further substituted, but not by aromatic groups and their use against illnesses related to iron overload. Molenda et al. disclose in Journal of Medicinal Chemistry 1994, 37, pages 4363-4370 chiral 3-hydroxy-pyridin-4-one compound 6 as enhancing iron excretion.

US Patent No. 6, 465, 604 described a series of 3,5-diphenyl-1,2,4-triazole compounds, wherein including Exjade (commercial name), which has strong affinity to Fe(III), However, its active groups contain two negatively charged oxygen ions and a carboxyl group; it is a tridentate ligand while chelating Fe(III), which forms a Fe-L2 type complex, possessing three unit negative charges itself, that is bad for their discharge from cells/tissues. Moreover, one of the active groups is a nitrogen atom with a lone pair of electrons, Exjade may have a negative effect on the balance of Zn(II) in vivo, at the same time because it has two phenolic hydroxyl groups in different positions (forming intramolecular hydrogen bonds structure similar to cis/trans isomerization), it can be complexed to several zinc ions to form high molecular weight polymers complexes, which is not conducive to its discharge from the cells either. Image loading...

Absorption, distribution, metabolism and excretion of chiral medicines are largely related to the 3D structures of their chiral centers. For drug absorption, chiral compounds entering cells via active transport mechanism are usually carried by special transport proteins, their recognition of enantiomers can be different, resulting in different absorption of enantiomers. For drug distribution, the binding effects of plasma protein and tissues are also somewhat stereoselective, leading to different in vivo distribution of enantiomers; for stereoselective of drug metabolism refers to when the substrate is biotransformated, the pathway and speed of enantiomer metabolism by biological systems can be different. One enantiomer may show ascendant metabolism, and therefore it is of great significance to the indicators including drug transformation and in vivo half-life. Glomerular filtration, tubular secretion and reabsorption of chiral drugs to clear the chiral drugs, having stereoselectivity, while the glomerular filtration rate is closely related to drug’s selectivity to binding plasma protein, so discharge style of enantiomers (urine / feces percentage) and the rate is also different.

Therefore, the qualitative difference of the interactions of a pair of enantiomers with various binding sites may exist or not, and the quantitive difference may exist (strong or weak), which results in the different activities between enantiomers. Thus the selection of optical enantiomers for medical use, requires a comprehensive study of metabolic activity, toxicology and pharmacokinetic properties etc. Thus the chiral nature of the 3-hydroxypyridin-4-one derivatives described in this patent has an important role on in vivo iron chelation.

The effectiveness of many oral 3-hydroxy-4-one derivatives drugs are subject to metabolic reaction of the 3-hydroxy moiety, which may be quickly glycosylated (see Reaction I). The hydroxypyridone after glycosylation loses the ability to chelate Fe(III). We can effectively inhibit glycosylation reaction by introducing hydroxyl groups to alkyl substituted residues on the pyridine ring. In addition, the partition coefficient of 3-hydroxy-pyridin-4-one derivatives has a great impact on the in vivo distribution and toxic effects. We have introduced various alkyl groups to the chiral point of the compound, in order to modify their lipophilicity, i.e. a phenyl group connected to the chiral point in compound IV-b while in IV-a it is a methyl group, and thus compound IV-b is relatively more lipophilic, and easier to penetrate through cell membranes of various tissues and critical barriers such as the blood-brain barrier and the placental barrier, thus affecting its in vivo distribution. Thus increase of hydroxyl groups can affect the intestinal absorption capacity, by introducing a large alkyl group, intestinal absorption of 3-hydroxy-pyridin-4-one derivatives can be enhanced. Image loading...

Reaction I

Image loading...

scheme is as follows: Image loading... Image loading...

Example7. (R)-3-Hydroxy-1-(1-hydroxy-3-benzyl propyl-2-)2-methyl pyridine-4(1H)-one, Number: CN128.

Image loading...

60 g 3-phenyloxy-2-methyl-4H-pyran-one(Example 1) was dissolved in 150 mL n-butanol, then 83.7 g D-phenylalaninol was added in. After thoroughly mixing, the solution was refluxed at 118°C for 36 h. After cooling and filtration, products were purified by silica gel column chromatography with Eluent ethanol: acetic ester=1:40. After Elution, light brown solid was obtained after rotary evaporation, which was then dissolved into 150 mL ethanol and 15 mL water, then it was hydrogenated and debenzylated with 5% Pd/C as catalyst, the solvent was removed under rotary evaporation, the remaining solid was recrystallized with methanol and ether, leading to 25.25 g light yellow solid. The yield was 35.1%. The free alkali’s 1HNMR (DMSO-d6): δ 2.00 (s, 3H), 2.98 (dd, J1=14, J2=5.5, 1H), 3.11 (dd, J1=14, J2=5, 1H), 3.73 (m, 2H), 4.54 (m, 1H), 6.21 (d, J=7, 1H), 7.17 (m, 5H), 7.87 (d, J=7.5, 1H).

 

PATENT

CN 102190644

http://www.google.com/patents/CN102190644B?cl=en

Zhejiang University

\\\\\\\\\\\\CN-128 , ind filed,  thelassemia,  iron overload, zhejiang
c1ccc(cc1)CC(N\2/C=C\C(/C(=C/2C)O)=O)CO
Share

ND 2110

 Uncategorized  Comments Off on ND 2110
Apr 102016
 

STR3

ND -2110

Molecular Formula: C21H28N4O3S
Molecular Weight: 416.53702 g/mol

2-[(3R)-12-{[(1r,4r)-4-(morpholin-4-yl)cyclohexyl]oxy}-7-thia-9,11-diazatricyclo[6.4.0.0²,⁶]dodeca-1(12),2(6),8,10-tetraen-3-yl]acetamide

1388894-17-4

C21 H28 N4 O3 S, 5H-​Cyclopenta[4,​5]​thieno[2,​3-​d]​pyrimidine-​5-​acetamide, 6,​7-​dihydro-​4-​[[trans-​4-​(4-​morpholinyl)​cyclohexyl]​oxy]​-​, (5R)​-
Molecular Weight416.54

ND-2110 is a potent and selective experimental inhibitor of IRAK4 described in patent WO2013106535 [2] and in a poster presented at the American College of Rheumatology meeting in 2012 (Abstract #1062 in Supplement: Abstracts of the American College of Rheumatology & Association of Rheumatology Health Professionals, Annual Scientific Meeting, November 9-4, 2012 Washington DC, Volume 64, Issue S10, Page S1-S1216).

Company Nimbus Therapeutics LLC
Description IL-1 receptor-associated kinase 4 (IRAK4) inhibitor
Molecular Target Interleukin-1 receptor-associated kinase 4 (IRAK4)
Mechanism of Action Interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitor
Therapeutic Modality Small molecule

 

PATENT

WO2013106535

http://www.google.com/patents/WO2013106535A1?cl=en

Example 29: Synthesis of 2-((R)-4-(((lr,4R)-4-morpholinocyclohexyl)oxy)-6,7-

29.3 1-67

Synthesis of compound 29.1. 4-(Morpholin-4-yl)cyclohexan-l-ol (commercially available; 218 mg, 1.2 mmol, 1.50 equiv) was treated with NaH (60% dispersion in mineral oil, 128 mg, 3.2 mmol, 4 equiv) in freshly distilled tetrahydrofuran (15 mL) for 30 min at 0 °C in a water/ice bath under nitrogen. Then a solution of intermediate 25.1 (289 mg, 0.8 mmol, 1.00 equiv) in 5 mL of THF was added via syringe and the resulting solution was allowed to stir for an additional 3 h at 60 °C in an oil bath. The reaction was then quenched with saturated aqueous NH4CI and extracted with 3 x 50 mL of ethyl acetate. The combined organic layers were washed with brine, dried (Na2S04) and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1:5-1:2) and purified to afford compound 29.1 (260 mg, 63%) as a colorless oil.

Synthesis of compound 29.2. To a solution of 29.1 (260 mg, 0.5 mmol, 1.0 equiv) in 10 mL of DCM was added 0.5 mL of concentrated hydrochloric acid in an ice/water bath. The resulting solution was stirred for 2 h and concentrated in vacuo. The residue was neutralized with saturated aqueous Na2C03 and extracted with 3 x 50 mL of ethyl acetate. The organic layers were combined, washed with brine, dried (Na2S04) and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel with DCM MeOH (15:1) to afford the desired alcohol 29.2 (185 mg, 91%) as a colorless oil. [00416] Synthesis of compound 29.3. Alcohol 29.2 (185 mg, 0.46 mmol, 1.00 equiv) was oxidized with dipyridinium dichromate (752 mg, 2.00 mmol, 4.36 equiv) in 50 mL of DMF for 24 h at room temperature. The resulting solution was diluted with water and extracted with 3 x 50 mL of mixed solutions of CHC¾/iso-PrOH. The organic layers were combined, dried (Na2S04) and concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (5:1 to 1:1) and purified to afford 105 mg (55%) of acid 29.3 as a yellow oil.

[00417] Synthesis of Compound 1-67. A 50 mL round-bottom flask containing a solution of acid 29.3 (105 mg, 0.25 mmol, 1.00 equiv), NH4C1 (80 mg, 1.50 mmol, 6.00 equiv), EDCI (57 mg, 0.3 mmol, 1.2 equiv), 4-dimethylaminopyridine (37 mg, 0.3 mmol, 1.2 equiv) and HOBt (40 mg, 0.3 mmol, 1.2 equiv) in 5 mL of anhydrous DMF was stirred for 24 h at room temperature. The resulting solution was diluted with water and extracted with 4 x 50 mL of mixed solution of CHCl3:iso-PrOH. The combined organic layers were concentrated under vacuum. The crude product was purified by preparative HPLC (SHIMADZU) under the following conditions: column: SunFire Prep C18, 19*150mm 5um; mobile phase: water (0.05% NH4CO3) and CH3CN (6.0% CH3CN up to 50.0% in 25 min); UV detection at 254/220 nm. The product-containing fractions were collected and concentrated to give Compound 1-67 (22.5 mg) as a white solid. ¾ NMR (300 MHz, CD3OD) δ 8.43 (s, 1H), 5.27-5.20 (m, 1H), 3.80-3.70 (m, 5H), 3.29-3.27 (m, 1H), 3.12-2.90 (m, 2H), 2.73-2.67 (m, 5H), 2.49-2.42 (m, 1H), 2.32-2.19 (m, 4H), 2.10-2.06 (d, 2H), 1.67-1.46 (m, 4H). MS: m/z 417 (M+H)+.

PATENT

http://www.google.com/patents/WO2012097013A1

Example 29: Synthesis of 2-((R)-4-(((lr,4R)-4-morpholinocyclohexyl)oxy)-6,7- dihydro-5H-cyclopenta[4,5]thieno[2,3-d]pyrimidin-5-yl)acetamide.

Page 280 of 407

2009184-0008

33b

Synthesis of compound 31b. 4-(Morpholin-4-yl)cyclohexan-l-ol (commercially available; 218 mg, 1.2 mmol, 1.50 equiv) was treated with NaH NMR (60% dispersion in mineral oil, 128 mg, 3.2 mmol, 4 equiv) in freshly distilled tetrahydrofuran (15 mL) for 30 min at 0 °C in a water/ice bath under nitrogen. Then a solution of intermediate Hb (289 mg, 0.8 mmol, 1.00 equiv) in 5 mL of THF was added via syringe and the resulting solution was allowed to stir for an additional 3 h at 60 °C in an oil bath. The reaction was then quenched with saturated aqueous NH4CI and extracted with 3 x 50 mL of ethyl acetate. The combined organic layers were washed with brine, dried (Na2S04) and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 :5-1 :2) and purified to afford compound 31b (260 mg, 63%) as a colorless oil.

Synthesis of compound 32b. To a solution of 31b (260 mg, 0.5 mmol, 1.0 equiv) in 10 mL of DCM was added 0.5 mL of concentrated hydrochloric acid in an ice/water bath. The resulting solution was stirred for 2 h and concentrated in vacuo. The residue was neutralized with saturated aqueous Na2C( j and extracted with 3 x 50 mL of ethyl acetate. The organic layers were combined, washed with brine, dried (Na2S04) and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel with DCM/MeOH NMR ( 15: 1 ) to afford the desired alcohol 32b ( 185 mg, 91 %) as a colorless oil.

Synthesis of compound 33b. Alcohol 32b (185 mg, 0.46 mmol, 1.00 equiv) was oxidized with dipyridinium dichromate (752 mg, 2.00 mmol, 4.36 equiv) in 50 mL of DMF for

Page 281 of 407

2009184-0008 24 h at room temperature. The resulting solution was diluted with water and extracted with 3 x 50 mL of mixed solutions of CHCU/iso-PrOH. The organic layers were combined, dried (Na2S04) and concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (5: 1 to 1 : 1 ) and purified to afford 105 mg (55%) of acid 33b as a yellow oil.

Synthesis of Compound. A 50 mL round-bottom flask containing a solution of acid 33b (105 mg, 0.25 mmol, 1.00 equiv), NH4C1 (80 mg, 1.50 mmol, 6.00 equiv), EDCI (57 mg, 0.3 mmol, 1.2 equiv), 4-dimethylaminopyridine (37 mg, 0.3 mmol, 1.2 equiv) and HOBt (40 mg, 0.3 mmol, 1.2 equiv) in 5 mL of anhydrous DMF was stirred for 24 h at room temperature. The resulting solution was diluted with water and extracted with 4 x 50 mL of mixed solution of CHCI3: iso-PrOH. The combined organic layers were concentrated under vacuum. The crude product was purified by preparative HPLC (SHIMADZU) under the following conditions: column: SunFire Prep C I 8, 19* 150mm 5um; mobile phase: water (0.05% Ν¾∞3) and CH3CN (6.0% CH3CN up to 50.0% in 25 min); UV detection at 254/220 nm. The product containing fractions were collected and concentrated to give the product (22.5 mg) as a white solid. Ή MR (300 MHz, CD3OD) δ 8.43 (s, 1H), 5.27-5.20 (m, 1H), 3.80-3.70 (m, 5H), 3.29-3.27 (m, 1 H), 3.12-2.90 (m, 2H), 2.73-2.67 (m, 5H), 2.49-2.42 (m, 1H), 2.32-2.19 (m, 4H), 2.10-2.06 (d, 2H), 1.67- 1.46 (m, 4H). MS: m/z 417 (M+H)+.

Paper

http://pubs.acs.org/doi/abs/10.1021/jm5016044

Recent Advances in the Discovery of Small Molecule Inhibitors of Interleukin-1 Receptor-Associated Kinase 4 (IRAK4) as a Therapeutic Target for Inflammation and Oncology Disorders

Miniperspective

Nimbus Discovery, 25 First Street, Suite 404, Cambridge, Massachusetts 02141, United States
Schrödinger Inc., 120 West Forty-Fifth Street, New York, New York 10036, United States
J. Med. Chem., 2015, 58 (1), pp 96–110
DOI: 10.1021/jm5016044
Abstract Image

IRAK4, a serine/threonine kinase, plays a key role in both inflammation and oncology diseases. Herein, we summarize the compelling biology surrounding the IRAK4 signaling node in disease, review key structural features of IRAK4 including selectivity challenges, and describe efforts to discover clinically viable IRAK4 inhibitors. Finally, a view of knowledge gained and remaining challenges is provided.

 STR3
  1. 78 Romero, D. L.; Robinson, S.; Wessel, M. D.; Greenwood, J. R. IRAK Inhibitors and Uses Thereof. WO201401902, January 16, 2014.

  2. 79.

    Harriman, G. C.; Romero, D. L.; Masse, C. E.; Robinson, S.; Wessel, M. D.; Greenwood, J. R. IRAK Inhibitors and Uses Thereof. WO2014011911A2, January 16, 2014.

  3. 80.

    Harriman, G. C.; Wester, R. T.; Romero, D. L.; Masse, C. E.; Robinson, R.; Greenwood, J. R. IRAK Inhibitors and Uses Thereof. WO2014011906A2, January 16, 2014

STR3

WO 2014194245

WO 2014194201

WO 2014194242

WO 2013106535

WO 2012097013

1. Chaudhary D, Robinson S, Romero DL. (2015)
Recent Advances in the Discovery of Small Molecule Inhibitors of Interleukin-1 Receptor-Associated Kinase 4 (IRAK4) as a Therapeutic Target for Inflammation and Oncology Disorders.
J. Med. Chem.58 (1): 96-110.
2. Harriman GC, Wester RT, Romero DL, Robinson S, Shelley M, Wessel MD, Greenwood JR, Masse CE, Kapeller-Libermann R. (2013)
Irak inhibitors and uses thereof.
Patent number: WO2013106535C1CC(CCC1N2CCOCC2)OC3=C4C5=C(CCC5CC(=O)N)SC4=NC=N3. Assignee: Nimbus Iris, Inc.. Priority date: 18/07/2013. Publication date: 10/01/2012.

http://nimbustx.com/sites/default/files/uploads/posters/irak4_nimbus_acr_poster_2012_small.pdf

///////ND-2110, ND 2110. IRAK4, NIMBUS, GTPL8802

 

NC(=O)CC1CCc2c1c1c(ncnc1s2)OC1CCC(CC1)N1CCOCC1

C1CC(CCC1N2CCOCC2)OC3=C4C5=C(CCC5CC(=O)N)SC4=NC=N3

Share
Follow

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

Join other followers: