New Drug Approvals

Home » Uncategorized (Page 9)

Category Archives: Uncategorized

DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE

Blog Stats

  • 4,798,787 hits

Flag and hits

Flag Counter

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 37.9K other subscribers
Follow New Drug Approvals on WordPress.com

Archives

Categories

Recent Posts

Flag Counter

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 37.9K other subscribers
DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his Ph.D from ICT, 1991,Matunga, Mumbai, India, in Organic Chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with AFRICURE PHARMA, ROW2TECH, NIPER-G, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India as ADVISOR, earlier assignment was with GLENMARK LIFE SCIENCES LTD, as CONSUlTANT, Retired from GLENMARK in Jan2022 Research Centre as Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Total Industry exp 32 plus yrs, Prior to joining Glenmark, he has worked with major multinationals like Hoechst Marion Roussel, now Sanofi, Searle India Ltd, now RPG lifesciences, etc. He has worked with notable scientists like Dr K Nagarajan, Dr Ralph Stapel, Prof S Seshadri, etc, He did custom synthesis for major multinationals in his career like BASF, Novartis, Sanofi, etc., He has worked in Discovery, Natural products, Bulk drugs, Generics, Intermediates, Fine chemicals, Neutraceuticals, GMP, Scaleups, etc, he is now helping millions, has 9 million plus hits on Google on all Organic chemistry websites. His friends call him Open superstar worlddrugtracker. His New Drug Approvals, Green Chemistry International, All about drugs, Eurekamoments, Organic spectroscopy international, etc in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 32 PLUS year tenure till date Feb 2023, Around 35 plus products in his career. He has good knowledge of IPM, GMP, Regulatory aspects, he has several International patents published worldwide . He has good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, Polymorphism etc., He suffered a paralytic stroke/ Acute Transverse mylitis in Dec 2007 and is 90 %Paralysed, He is bound to a wheelchair, this seems to have injected feul in him to help chemists all around the world, he is more active than before and is pushing boundaries, He has 100 million plus hits on Google, 2.5 lakh plus connections on all networking sites, 100 Lakh plus views on dozen plus blogs, 227 countries, 7 continents, He makes himself available to all, contact him on +91 9323115463, email amcrasto@gmail.com, Twitter, @amcrasto , He lives and will die for his family, 90% paralysis cannot kill his soul., Notably he has 38 lakh plus views on New Drug Approvals Blog in 227 countries......https://newdrugapprovals.wordpress.com/ , He appreciates the help he gets from one and all, Friends, Family, Glenmark, Readers, Wellwishers, Doctors, Drug authorities, His Contacts, Physiotherapist, etc He has total of 32 International and Indian awards

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

Tegeprotafib


Tegeprotafib

CAS 2407610-46-0

Molecular Weight326.30
FormulaC13H11FN2O5S

PTPN2/1-IN-1, YGY4WEM0NZ

5-(1-fluoro-3-hydroxy-7-methoxynaphthalen-2-yl)-1,1-dioxo-1,2,5-thiadiazolidin-3-one

Tegeprotafib (PTPN2/1-IN-1) (Compound 124) is an orally active PTPN1 and PTPN2 inhibitor with IC50s of 4.4 nM and 1-10 nM against PTPN2 and PTP1B, respectively.

Cancer immunotherapy regimens targeting immune evasion mechanisms including checkpoint blockade (e.g., PD-1/PD-L1 and CTLA-4 blocking antibodies) have been shown to be effective in treating in a variety of cancers, dramatically improving outcomes in some populations refractory to conventional therapies. However, incomplete clinical responses and the development of intrinsic or acquired resistance will continue to limit the patient populations who could benefit from checkpoint blockade.
      Protein tyrosine phosphatase non-receptor type 2 (PTPN2), also known as T cell protein tyrosine phosphatase (TC-PTP), is an intracellular member of the class 1 subfamily of phospho-tyrosine specific phosphatases that control multiple cellular regulatory processes by removing phosphate groups from tyrosine substrates. PTPN2 is ubiquitously expressed, but expression is highest in hematopoietic and placental cells (Mosinger, B. Jr. et al., Proc NatlAcad Sci USA 89:499-503; 1992). In humans, PTPN2 expression is controlled post-transcriptionally by the existence of two splice variants: a 45 kDa form that contains a nuclear localization signal at the C-terminus upstream of the splice junction, and a 48 kDa canonical form which has a C-terminal ER retention motif (Tillmann U. et al., Mol Cell Biol 14:3030-3040; 1994). The 45 kDa isoform can passively transfuse into the cytosol under certain cellular stress conditions. Both isoforms share an N-terminal phospho-tyrosine phosphatase catalytic domain. PTPN2 negatively regulates signaling of non-receptor tyrosine kinases (e.g., JAK1, JAK3), receptor tyrosine kinases (e.g., INSR, EGFR, CSF1R, PDGFR), transcription factors (e.g., STAT1, STAT3, STAT5a/b), and Src family kinases (e.g., Fyn, Lck). As a critical negative regulator of the JAK-STAT pathway, PTPN2 functions to directly regulate signaling through cytokine receptors, including IFNγ. The PTPN2 catalytic domain shares 74% sequence homology with PTPN1 (also called PTP1B), and shares similar enzymatic kinetics (Romsicki Y. et al., Arch Biochem Biophys 414:40-50; 2003).
      Data from a loss of function in vivo genetic screen using CRISPR/Cas9 genome editing in a mouse B16F10 transplantable tumor model show that deletion of Ptpn2 gene in tumor cells improved response to the immunotherapy regimen of a GM-CSF secreting vaccine (GVAX) plus PD-1 checkpoint blockade (Manguso R. T. et al., Nature 547:413-418; 2017). Loss of Ptpn2 sensitized tumors to immunotherapy by enhancing IFNγ-mediated effects on antigen presentation and growth suppression. The same screen also revealed that genes known to be involved in immune evasion, including PD-L1 and CD47, were also depleted under immunotherapy selective pressure, while genes involved in the IFNγ signaling pathway, including IFNGR, JAK1, and STAT1, were enriched. These observations point to a putative role for therapeutic strategies that enhance IFNγ sensing and signaling in enhancing the efficacy of cancer immunotherapy regimens.
      Protein tyrosine phosphatase non-receptor type 1 (PTPN1), also known as protein tyrosine phosphatase-1B (PTPiB), has been shown to play a key role in insulin and leptin signaling and is a primary mechanism for down-regulating both the insulin and leptin receptor signaling pathways (Kenner K. A. et al., J Biol Chem 271: 19810-19816, 1996). Animals deficient in PTP1B have improved glucose regulation and lipid profiles and are resistant to weight gain when treated with a high fat diet (Elchebly M. et al., Science 283: 1544-1548, 1999). Thus, PTP1B inhibitors are expected to be useful for the treatment of type 2 diabetes, obesity, and metabolic syndrome.

SCHEME

PATENT

Calico Life Sciences LLC; AbbVie Inc. , WO2021127499

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021127499&_cid=P21-M9UYU6-17583-1

Example 25: 5-(1-fluoro-3-hydroxy-7-methoxynaphthalen-2-yl)-1λ6,2,5-thiadiazolidine-1,1,3-trione (Compound 124)

Example 25A: benzyl 3-(benzyloxy)-7-methoxynaphthalene-2-carboxylate

A mixture of 3-hydroxy-7-methoxy-2-naphthoic acid (75 g, 344 mmol) and cesium carbonate (336 g, 1031 mmol) in N,N-dimethylformamide (687 mL) was rapidly stirred for 5 minutes at 23 °C. Thereafter, benzyl bromide (84 mL, 705 mmol) was added. After 90 minutes, the mixture was poured into H2O (1 L) and extracted with ethyl acetate (4 × 300 mL). The combined organic layers were washed with saturated aqueous ammonium chloride (3 × 100 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to afford a brown solid. The crude solid was collected by filtration, slurried with tert-butyl methyl ether/heptanes (1:2, 3 × 100 mL), then dried in vacuo (12 mbar) at 40 °C to afford the title compound (122.5 g, 307 mmol, 89% yield) as a beige solid. MS (APCI+) m/z 399 [M+H]+.

Example 25B: 3-(benzyloxy)-7-methoxynaphthalene-2-carboxylic acid

To a suspension of the product of Example 25A (122.5 g, 307 mmol) in methanol (780 mL) was added 6 M aqueous sodium hydroxide (154 mL, 922 mmol). The heterogeneous, brown slurry was agitated with an overhead mechanical stirrer and heated to an internal temperature of 68 °C. After 15 minutes, the mixture was cooled to room temperature in an ice bath, and 6 M HCl (250 mL) was added over 5 minutes. The off-white solid was collected by filtration, washed with H2O (3 × 500 mL), and dried to constant weight in vacuo at 65 °C to afford the title compound (84.1 g, 273 mmol, 89% yield) as a white solid. MS (APCI+) m/z 309 [M+H]+.

Example 25C: 3-(benzyloxy)-7-methoxynaphthalen-2-amine

To a suspension of the product of Example 25B (84.1 g, 273 mmol), in toluene (766 mL) and tert-butanol (766 mL) was added triethylamine (40.3 mL, 289 mmol). The homogeneous black solution was heated to an internal temperature of 80 °C under nitrogen, and diphenyl phosphorazidate (62.2 mL, 289 mmol) was added dropwise over 90 minutes with the entire

reaction behind a blast shield. After 5 hours, the reaction was cooled to room temperature, diluted with H2O (1.5 L), and extracted with ethyl acetate (3 × 150 mL). The combined organic layers were washed with brine (2 × 100 mL), dried over sodium sulfate, filtered and concentrated to give 180.1 g of a dark brown solid. The solid was carried forward to hydrolysis without further purification.

To the crude intermediate was added diethylenetriamine (475 mL, 4.40 mol). The heterogeneous suspension was heated to an internal temperature of 130 °C under nitrogen, at which time a homogeneous dark orange solution formed. After 16 hours, the mixture was cooled to room temperature in an ice bath, and H2O (1.5 L) was added slowly over 3 minutes, resulting in precipitation of a yellow solid and a concomitant exotherm to an internal temperature of 62 °C. Once the heterogeneous suspension had cooled to room temperature, the crude solid was dissolved in CH2Cl2 (1.5 L), and the layers were separated. The aqueous layer was back-extracted with CH2Cl2 (3 × 150 mL), and the combined organic layers were washed with brine (3 × 100 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to afford 78.8 g of an orange solid. The solid was slurried with isopropanol (50 mL), collected via filtration, re-slurried with isopropanol (1 × 50 mL), and dried in vacuo (15 mbar) at 35 °C to afford the title compound (60.12 g, 215 mmol, 79% yield over two steps) as a yellow solid. MS (APCI+) m/z 280 [M+H]+.

Example 25D: methyl {[3-(benzyloxy)-7-methoxynaphthalen-2-yl]amino}acetate

To a mixture of the product of Example 25C (59.2 g, 212 mmol) and potassium carbonate (58.6 g, 424 mmol) in dimethylformamide (363 mL) and H2O (1.91 mL, 106 mmol) was added methyl 2-bromoacetate (30.1 mL, 318 mmol). The suspension was vigorously stirred at room temperature for 5 minutes and then heated to an internal temperature of 60 °C. After 70 minutes, the suspension was cooled to room temperature and diluted with H2O (600 mL) and ethyl acetate (500 mL). The aqueous layer was extracted with ethyl acetate (2 × 300 mL), and the combined organic layers were washed with saturated aqueous ammonium chloride (3 × 60 mL), dried over sodium sulfate, filtered, and concentrated to afford 104.3 g of a pale beige solid. The solid was triturated with heptanes (200 mL). The resulting beige solid was collected via filtration, washed with additional heptanes (2 × 30 mL), and dried in vacuo (15 mbar) at 35 °C to afford the title compound (72.27 g, 206 mmol, 97% yield) as an off-white solid. MS (APCI+) m/z 352 [M+H]+.

Example 25E: methyl {[3-(benzyloxy)-1-fluoro-7-methoxynaphthalen-2-yl]amino}acetate To a mixture of the product of Example 25D (30.0 g, 85 mmol) and N-fluorobenzenesulfonimide (26.9 g, 85 mmol) was added tetrahydrofuran (THF) (854 mL), and

the resulting homogeneous yellow solution was stirred at room temperature. After 90 minutes, residual oxidant was quenched by adding a solution of sodium thiosulfate pentahydrate (10.59 g, 42.7 mmol) in water (150 mL), and the mixture was stirred at room temperature for 30 minutes. Thereafter, ethyl acetate (600 mL) was added, the aqueous layer was separated, and the organic layer was washed with a solution of sodium carbonate (18.10 g, 171 mmol) in water (30 mL), followed by water:brine (1:1, 1 × 20 mL). The organic fraction was dried over sodium sulfate, filtered, and the concentrated in vacuo to afford a bright yellow/orange solid. The solids were triturated with tert-butyl methyl ether (300 mL), collected via filtration, and the filter cake (N-(phenylsulfonyl)benzenesulfonamide) was washed with tert-butyl methyl ether (2 × 100 mL). The filtrate was concentrated to afford 34.6 g of a dark red oil that was purified by flash chromatography (750 g SiO2, heptanes to 20% ethyl acetate/heptanes) to afford the title compound (16.07 g, 43.5 mmol, 51% yield) as a yellow solid. MS (APCI+) m/z 370 [M+H]+. Example 25F: methyl {[3-(benzyloxy)-1-fluoro-7-methoxynaphthalen-2-yl](sulfamoyl)amino}acetate

To a solution of chlorosulfonyl isocyanate (5.13 mL, 59.1 mmol) in dichloromethane (83 mL) at 0 °C was added tert-butanol (5.65 mL, 59.1 mmol) slowly so that the internal temperature remained less than 10 °C. After stirring for 30 minutes at 0 °C, a preformed solution of the product of Example 25E (14.55 g, 39.4 mmol) and triethylamine (10.98 mL, 79 mmol) in dichloromethane (68.9 mL) was added slowly via addition funnel so that the internal temperature remained below 10 °C. Upon complete addition, the addition funnel was rinsed with dichloromethane (23 mL). The resulting solution was stirred for 30 minutes at 0 °C, and then the reaction mixture was quenched with H2O (20 mL). The layers were separated, and the aqueous layer was extracted with dichloromethane (2 × 30 mL). The combined organic layers were washed with brine (1 × 30 mL), dried over sodium sulfate, filtered and concentrated in vacuo to give an orange oil. The residue was dissolved in ethyl acetate (200 mL) and washed with water:brine (1:1, 2 × 50 mL) to remove residual triethylamine hydrochloride. The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo to give methyl {[3-(benzyloxy)-1-fluoro-7-methoxynaphthalen-2-yl][(tert-butoxycarbonyl)sulfamoyl]amino}acetate which was used without purification.

To a solution of methyl {[3-(benzyloxy)-1-fluoro-7-methoxynaphthalen-2-yl][(tert-butoxycarbonyl)sulfamoyl]amino}acetate in dichloromethane (98 mL) was added trifluoroacetic acid (45.5 mL, 591 mmol), and the resulting dark solution was stirred at room temperature. After 20 minutes, the reaction was quenched by slow addition of saturated aqueous sodium bicarbonate (691 mL) via an addition funnel. The layers were separated, and the aqueous layer was extracted with dichloromethane (2 × 50 mL). The combined organic layers were concentrated to give a dark red oil; upon addition of tert-butyl methyl ether (60 mL), a yellow solid precipitated that was collected via filtration, washed with tert-butyl methyl ether (2 × 30 mL) and dried in vacuo (15 mbar) at 35 °C to give the title compound (13.23 g, 29.5 mmol, 75% yield over two steps) as a light yellow solid. MS (ESI+) m/z 449 [M+H]+.

Example 25G: 5-(1-fluoro-3-hydroxy-7-methoxynaphthalen-2-yl)-1λ6,2,5-thiadiazolidine-1,1,3-trione

To a solution of the product of Example 25F (13.23 g, 29.5 mmol) in tetrahydrofuran (THF) (355 mL) at room temperature was added solid potassium tert-butoxide (3.31 g, 29.5 mmol), and the resulting solution was stirred at room temperature. After 10 minutes, the reaction was quenched with 1 M hydrochloric acid (90 mL) and diluted with ethyl acetate (400 mL). The layers were separated, and the aqueous layer was extracted with ethyl acetate (2 × 120 mL). The combined organic layers were washed with brine (3 × 50 mL), then dried over sodium sulfate, filtered and concentrated. The crude 5-[3-(benzyloxy)-1-fluoro-7-methoxynaphthalen-2-yl]-1λ6,2,5-thiadiazolidine-1,1,3-trione was used in the subsequent reaction without further purification.

A mixture of crude intermediate, 5-[3-(benzyloxy)-1-fluoro-7-methoxynaphthalen-2-yl]-1λ6,2,5-thiadiazolidine-1,1,3-trione (12.28 g, 29.5 mmol) and pentamethylbenzene (13.11 g, 88 mmol) in dichloromethane (147 mL) was cooled to an internal temperature of –76 °C under an atmosphere of dry nitrogen. Subsequently, a 1 M solution of boron trichloride (59.0 mL, 59.0 mmol) in CH2Cl2 was added dropwise over 15 minutes, so as not to raise the internal temperature past –72 °C. Over the course of the addition, the reaction turned dark brown and became homogeneous. Incomplete conversion was observed, and additional boron trichloride (2 × 5.90 mL, 2 × 5.90 mmol) was added, resulting in full conversion. The reaction was quenched at –75 °C with CH2Cl2/methanol (10:1, 140 mL) via cannula transfer under nitrogen over 15 minutes, then slowly warmed to room temperature over 20 minutes under nitrogen. The volatiles were removed in vacuo to afford a brown/tan solid, which was collected by filtration, and slurried with heptanes (5 × 40 mL) and CH2Cl2 (3 × 40 mL). The crude solid was suspended in isopropanol (75 mL), warmed until the material dissolved, then allowed to cool slowly to room temperature over 1 hour. The solid was collected by filtration, washed with heptanes (2 × 30 mL), and dried in vacuo (15 mbar) at 60 °C to afford 5.11 g of a white solid. The mother liquor was concentrated, and the process was repeated to give an additional 1.96 g of a white solid. The batches were combined to obtain the title compound (7.07 g, 21.67 mmol, 73.5% yield over two steps). 1H NMR (CD3OD) δ ppm 7.60 (dd, J = 9.1, 1.5 Hz, 1H), 7.25 (d, J = 2.6, 1H), 7.16 (dd, J = 9.1, 2.6 Hz, 1H), 7.04 (s, 1 H), 4.56 (s, 2H), 3.89 (s, 3 H); MS (ESI) m/z 325 [M–H].

PATENT

WO2020186199 

WO2019246513 

PATENT

compound 124 [US20230019236A1]

https://patentscope.wipo.int/search/en/detail.jsf?docId=US389737555&_cid=P21-M9UYQD-14144-1

[1]. Elliot FARNEY, et al. Protein tyrosine phosphatase inhibitors and methods of use thereof. Patent WO2019246513A1.

///////Tegeprotafib, PTPN2/1-IN-1, YGY4WEM0NZ

Probenecid


Probenecid

  • 57-66-9
  • 4-(Dipropylsulfamoyl)benzoic acid
  • Probenecid acid
  • Benemid

4-(dipropylsulfamoyl)benzoic acid

C13H19NO4S, 285.359


  • HC 5006
  • NSC-18786

FDA APPROVED, 10/25/2024, sulopenem etzadroxil, probenecid, Orlynvah, To treat uncomplicated urinary tract infections (uUTI)
Drug Trial Snapshot

Probenecid, also sold under the brand name Probalan, is a medication that increases uric acid excretion in the urine. It is primarily used in treating gout and hyperuricemia.

Probenecid was developed as an alternative to caronamide[1] to competitively inhibit renal excretion of some drugs, thereby increasing their plasma concentration and prolonging their effects.

Experimental Properties

PropertyValueSource
melting point (°C)195 °CPhysProp
water solubility27.1 mg/LNot Available
logP3.21HANSCH,C ET AL. (1995)
pKa3.4SANGSTER (1994)
Patent NumberPediatric ExtensionApprovedExpires (estimated)
US12109197No2024-10-082039-04-01US flag
US11554112No2023-01-172039-04-01US flag
US11478428No2022-10-252039-12-23US flag
US7795243No2010-09-142029-06-03US flag

PATENT

https://patents.google.com/patent/CN103613521A/en

At present, the production technique of probenecid mainly contains two kinds:

(1) p-methyl benzenesulfonic acid-dipropyl amine method

Take p-methyl benzenesulfonic acid as raw material, through potassium bichromate or potassium permanganate oxidation, then react generation with chlorsulfonic acid generation sulfonating chlorinating to carboxyl benzene sulfonyl chloride, amidate action occurs then in organic solvent and obtain the finished product probenecid.Reaction process route is as follows:

Figure 642971DEST_PATH_IMAGE001

This technique in a large number with an organic solvent, seriously polluted; Heavy metal recovery and treatment cost are high; Chlorsulfonic acid transportation, storage and use are dangerous large, and acid mist is obvious.Along with the increasing of environmental protection pressure, people increase severely day by day to the concern of environment, and this route is substantially in end-of-life state.

(2) to methyl benzenesulfonamide-Halopropane method

To methyl benzenesulfonamide, through potassium bichromate or potassium permanganate oxidation, be P―Carboxybenzenesulfonamide, under the effect of alkali, with Halopropane generation alkylated reaction, after acidifying, obtain probenecid.Reaction process route is as follows:

Figure 201310587646X100002DEST_PATH_IMAGE003

This process using sodium dichromate 99 or potassium permanganate oxidation are to methyl benzenesulfonamide, and yield is on the low side (lower than 50%).In addition, the waste water that contains chromium or manganese is difficult to dispose, and these have all seriously restricted further developing of this technique.

Reaction scheme of the present invention is as follows:

Figure 201310587646X100002DEST_PATH_IMAGE004

embodiment 1

(1) diazotization reaction

Get 68.6g para-amino benzoic acid (0.5mol), 250g water and 127.4ml hydrochloric acid (31%, 1.25mol) join in 2000ml there-necked flask, in ice-water bath, stir, be cooled to 0-5 ℃, drip sodium nitrite solution (34.5g Sodium Nitrite, 0.5mol, be dissolved in 190g water), control temperature at 10-20 ℃, it is 4 hours that time for adding is controlled, after dropping finishes, at this temperature, continue reaction 1 hour, obtain diazotization reaction liquid.

(2) sulfonating chlorinating reaction

In 5000ml there-necked flask, add 250g water, 765ml hydrochloric acid (31%, 7.5mol), in ice-water bath, stir, be cooled to-5 ℃, start to pass into liquid sulfur dioxide, control temperature at-3–1 ℃, when passing into 64g sulfurous gas (1mol), sulfurous gas absorbs complete, obtains sulfonating chlorinating reagent.

In sulfonating chlorinating reagent, add diazotization reaction liquid, adding the time control of diazotization reaction liquid is 5 hours, is warming up to gradually 5-10 ℃, continues reaction 8 hours at this temperature; Filtration obtains 121g to carboxyl benzene sulfonyl chloride.

(3) synthetic probenecid reaction

In 1000ml there-necked flask, add 350g water, 152g dipropyl amine (1.5mol), open and stir, when temperature is greater than 15 ℃, start to divide gradually 40 batches add step (2) gained to carboxyl benzene sulfonyl chloride, temperature control 40-50 ℃, adds and at this temperature, stirs 3 hours continuing after carboxyl benzene sulfonyl chloride.Drip hydrochloric acid (31%), regulate pH value to 2-3, continue to stir 1 hour.Filter, obtain 135g probenecid crude product, put in 500ml pure water, agitator treating 1 hour, heavy 122.8g after filtering, being dried, yield 86.2%(is in para-amino benzoic acid), purity 98.2%.

embodiment 2

(1) diazotization reaction

Get 68.6g para-amino benzoic acid (0.5mol), 250g water and 152.9ml hydrochloric acid (31%, 1.5mol) join in 2000ml there-necked flask, in ice-water bath, stir, be cooled to 0-5 ℃, drip sodium nitrite solution (36.0g Sodium Nitrite, 0.52mol, be dissolved in 190g water), control temperature at 0-10 ℃, it is 3 hours that time for adding is controlled, after dropping finishes, at this temperature, continue reaction 1 hour, obtain diazotization reaction liquid.

(2) sulfonating chlorinating reaction

In 5000ml there-necked flask, add 250g water, 887ml hydrochloric acid (31%, 8.7mol), in ice-water bath, stir, be cooled to-5 ℃, start to pass into liquid sulfur dioxide, control temperature at 0-5 ℃, when passing into 112g sulfurous gas (1.75mol), sulfurous gas absorbs complete, obtains sulfonating chlorinating reagent.

In sulfonating chlorinating reagent, add diazotization reaction liquid, adding the time control of diazotization reaction liquid is 4 hours, is warming up to gradually 5-15 ℃, continues reaction 5 hours at this temperature; Filtration obtains 150g to carboxyl benzene sulfonyl chloride.

(3) synthetic probenecid reaction

In 1000ml there-necked flask, add 350g water, 192g dipropyl amine (1.9mol), open and stir, when temperature is greater than 15 ℃, start to divide gradually 35 batches add step (2) gained to carboxyl benzene sulfonyl chloride, temperature control 40-50 ℃, adds and at this temperature, stirs 2 hours continuing after carboxyl benzene sulfonyl chloride.Drip hydrochloric acid (31%), regulate pH value to 2-3, continue to stir 1 hour.Filter, obtain 155.4g probenecid crude product, put in 500ml pure water, agitator treating 1 hour, heavy 129.5g after filtering, being dried, yield 90.9%(is in para-amino benzoic acid), purity 98.7%.

embodiment 3

(1) diazotization reaction

Get 68.6g para-amino benzoic acid (0.5mol), 250g water and 203.9ml hydrochloric acid (31%, 2mol) join in 2000ml there-necked flask, in ice-water bath, stir, be cooled to-10–5 ℃, drip sodium nitrite solution (38.0g Sodium Nitrite, 0.55mol, be dissolved in 190g water), control temperature at 0-10 ℃, it is 5 hours that time for adding is controlled, after dropping finishes, at this temperature, continue reaction 1 hour, obtain diazotization reaction liquid.

(2) sulfonating chlorinating reaction

In 5000ml there-necked flask, add 250g water, 968ml hydrochloric acid (31%, 9.5mol), in ice-water bath, stir, be cooled to-5 ℃, start to pass into liquid sulfur dioxide, control temperature at 5-10 ℃, when passing into 160g sulfurous gas (2.5mol), sulfurous gas absorbs complete, obtains sulfonating chlorinating reagent.

In sulfonating chlorinating reagent, add diazotization reaction liquid, adding the time control of diazotization reaction liquid is 3 hours, is warming up to gradually 10-15 ℃, continues reaction 20 hours at this temperature; Filtration obtains 146.7g to carboxyl benzene sulfonyl chloride, needn’t be dried, and directly enters next step reaction.

(3) synthetic probenecid reaction

In 1000ml there-necked flask, add 350g water, 202g dipropyl amine (2mol), open to stir, when temperature is greater than 30 ℃, start to divide gradually 30 batches add step (2) gained to carboxyl benzene sulfonyl chloride, temperature control 40-50 ℃, adds and at this temperature, stirs 4 hours continuing after carboxyl benzene sulfonyl chloride.Drip hydrochloric acid (31%), regulate pH value to 2-3, continue to stir 1 hour.Filtration obtains 151.7g probenecid crude product, puts in 500ml pure water, and agitator treating 1 hour, heavy 128.5g after filtering, being dried, yield 90.2%(is in para-amino benzoic acid), purity 98.8%.Medical uses

Probenecid is primarily used to treat gout and hyperuricemia.

Probenecid is sometimes used to increase the concentration of some antibiotics and to protect the kidneys when given with cidofovir. Specifically, a small amount of evidence supports the use of intravenous cefazolin once rather than three times a day when it is combined with probenecid.[2]

It has also found use as a masking agent,[3] potentially helping athletes using performance-enhancing substances to avoid detection by drug tests.

Adverse effects

Mild symptoms such as nausea, loss of appetite, dizziness, vomiting, headache, sore gums, or frequent urination are common with this medication. Life-threatening side effects such as thrombocytopeniahemolytic anemialeukemia and encephalopathy are extremely rare.[4] Theoretically probenecid can increase the risk of uric acid kidney stones.

Drug interactions

Some of the important clinical interactions of probenecid include those with captoprilindomethacinketoprofenketorolacnaproxencephalosporinsquinolonespenicillinsmethotrexatezidovudineganciclovirlorazepam, and acyclovir. In all these interactions, the excretion of these drugs is reduced due to probenecid, which in turn can lead to increased concentrations of these.[5]

Pharmacology

Pharmacodynamics

In gout, probenecid competitively inhibits the reabsorption of uric acid through the organic anion transporter (OAT) at the proximal tubules. This leads to preferential reabsorption of probenecid back into plasma and excretion of uric acid in urine,[6] thus reducing blood uric acid levels and reducing its deposition in various tissues.

Probenecid also inhibits pannexin 1.[7] Pannexin 1 is involved in the activation of inflammasomes and subsequent release of interleukin-1β causing inflammation. Inhibition of pannexin 1 thus reduces inflammation, which is the core pathology of gout.[7]

Pharmacokinetics

In the kidneys, probenecid is filtered at the glomerulus, secreted in the proximal tubule and reabsorbed in the distal tubule. Probenicid lowers the concentration of certain drugs in urine drug screens by reducing renal excretion of these drugs.

Historically, probenecid has been used to increase the duration of action of drugs such as penicillin and other beta-lactam antibiotics. Penicillins are excreted in the urine at proximal and distal convoluted tubules through the same organic anion transporter (OAT) as seen in gout. Probenecid competes with penicillin for excretion at the OAT, which in turn increases the plasma concentration of penicillin.[8]

History

During World War II, probenecid was used to extend limited supplies of penicillin. This use exploited probenecid’s interference with drug elimination (via urinary excretion) in the kidneys and allowed lower doses of penicillin to be used.[9]

Probenecid was added to the International Olympic Committee‘s list of banned substances in January 1988, due to its use as a masking agent.[10]

References

  1. ^ Mason RM (June 1954). “Studies on the effect of probenecid (benemid) in gout”Annals of the Rheumatic Diseases13 (2): 120–130. doi:10.1136/ard.13.2.120PMC 1030399PMID 13171805.
  2. ^ Cox VC, Zed PJ (March 2004). “Once-daily cefazolin and probenecid for skin and soft tissue infections”. The Annals of Pharmacotherapy38 (3): 458–463. doi:10.1345/aph.1d251PMID 14970368S2CID 11449580.
  3. ^ Morra V, Davit P, Capra P, Vincenti M, Di Stilo A, Botrè F (December 2006). “Fast gas chromatographic/mass spectrometric determination of diuretics and masking agents in human urine: Development and validation of a productive screening protocol for antidoping analysis”. Journal of Chromatography A1135 (2): 219–229. doi:10.1016/j.chroma.2006.09.034hdl:2318/40201PMID 17027009S2CID 20282106.
  4. ^ Kydd AS, Seth R, Buchbinder R, Edwards CJ, Bombardier C (November 2014). “Uricosuric medications for chronic gout”The Cochrane Database of Systematic Reviews (11): CD010457. doi:10.1002/14651858.CD010457.pub2PMC 11262558PMID 25392987.
  5. ^ Cunningham RF, Israili ZH, Dayton PG (March–April 1981). “Clinical pharmacokinetics of probenecid”. Clinical Pharmacokinetics6 (2): 135–151. doi:10.2165/00003088-198106020-00004PMID 7011657S2CID 24497865.
  6. ^ “Probenecid”PubChem. U.S. National Library of Medicine. Retrieved 2022-06-12.
  7. Jump up to:a b Silverman W, Locovei S, Dahl G (September 2008). “Probenecid, a gout remedy, inhibits pannexin 1 channels”American Journal of Physiology. Cell Physiology295 (3): C761 – C767. doi:10.1152/ajpcell.00227.2008PMC 2544448PMID 18596212.
  8. ^ Ho RH (January 2010). “4.25 – Uptake Transporters”. In McQueen CA, Kim RB (eds.). Comprehensive Toxicology (Second ed.). Oxford: Elsevier. pp. 519–556. doi:10.1016/B978-0-08-046884-6.00425-5ISBN 978-0-08-046884-6.
  9. ^ Butler D (November 2005). “Wartime tactic doubles power of scarce bird-flu drug”Nature438 (7064): 6. Bibcode:2005Natur.438….6Bdoi:10.1038/438006aPMID 16267514.
  10. ^ Wilson W, Derse E, eds. (2001). Doping in Elite Sport: The Politics of Drugs in the Olympic Movement. Human Kinetics. p. 86. ISBN 0-7360-0329-0.
Clinical data
Trade namesProbalan
AHFS/Drugs.comMonograph
MedlinePlusa682395
Routes of
administration
By mouth
ATC codeM04AB01 (WHO)
Legal status
Legal statusIn general: ℞ (Prescription only)
Pharmacokinetic data
Protein binding75-95%
Elimination half-life2-6 hours (dose: 0.5-1 g)
Excretionkidney (77-88%)
Identifiers
showIUPAC name
CAS Number57-66-9 
PubChem CID4911
IUPHAR/BPS4357
DrugBankDB01032 
ChemSpider4742 
UNIIPO572Z7917
KEGGD00475 
ChEMBLChEMBL897 
CompTox Dashboard (EPA)DTXSID9021188 
ECHA InfoCard100.000.313 
Chemical and physical data
FormulaC13H19NO4S
Molar mass285.36 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI

/////////probenecid, APPROVALS 2024, FDA 2024, Orlynvah, HC 5006, NSC-18786

#probenecid, #APPROVALS 2024, #FDA 2024, #Orlynvah, #HC 5006, #NSC-18786

Bleximenib


Bleximenib

CAS 2654081-35-1

WeightAverage: 599.796
Monoisotopic: 599.395916661

Chemical FormulaC32H50FN7O3

  • CS-0636752
  • DA-55335
  • HY-148669
  • PHASE 3
  • JNJ-75276617; Menin-MLL inhibitor 24
  • Benzamide, N-ethyl-5-fluoro-2-[[5-[2-[(1R)-4-[(2-methoxyethyl)methylamino]-1-(1-methylethyl)butyl]-2,6-diazaspiro[3.4]oct-6-yl]-1,2,4-triazin-6-yl]oxy]-N-(1-methylethyl)-
  • N-ethyl-5-fluoro-2-{[5-(2-{(3R)-6-[(2-methoxyethyl)(methyl)amino]-2-methylhexan-3-yl}-2,6-diazaspiro[3.4]octan-6-yl)-1,2,4-triazin-6-yl]oxy}-N-(propan-2-yl)benzamide

2866179-95-3 (oxalate)

(R)-N-ethyl-5-fluoro-N-isopropyl-2-((5-(2-(6-((2-methoxyethyl)(methyl)amino)-2-methylhexan-3-yl)-2,6-diazaspiro[3.4]octan-6-yl)-1,2,4-triazin-6-yl)oxy)benzamide oxalate

Chemical Formula: C34H52FN7O7
Exact Mass: 689.39
Molecular Weight: 689.830
Elemental Analysis: C, 59.20; H, 7.60; F, 2.75; N, 14.21; O, 16.23

\Bleximenib is under investigation in clinical trials NCT04811560 (A Phase 1/2 Study of Bleximenib in Participants With Acute Leukemia) and NCT05453903 (A Study of Bleximenib in Combination With Acute Myeloid Leukemia (AML) Directed Therapies)

Bleximenib (JNJ-75276617) is an orally active and selective menin-KMT2A inhibitor, with IC50 values of 0.1 nM, 0.045 nM, and ≤0.066 nM for humans, mice, and dogs, respectively. Bleximenib can inhibit the proliferation and induce apoptosis and differentiation of tumor cells. Bleximenib can be used in the research of tumors such as leukemia.

Bleximenib is an orally bioavailable protein-protein interaction (PPI) inhibitor of the menin-mixed lineage leukemia (MLL; mixed-lineage leukemia 1; MLL1; myeloid/lymphoid leukemia; histone-lysine N-methyltransferase 2A; KMT2A) proteins, with potential antineoplastic activity. Upon oral administration, bleximenib inhibits the interaction between the two proteins menin and MLL and the formation of the menin-MLL complex. This reduces the expression of downstream target genes and results in an inhibition of the proliferation of leukemic cells with either KMT2A alterations such as gene rearrangements (KMT2A-r), duplications, and amplification, or nucleophosmin 1 gene (NPM1) alterations. The menin-MLL complex plays a key role in the survival, growth, transformation and proliferation of certain kinds of leukemia cells.

SCHEME

SIDECHAIN

PATENTS

Janssen Pharmaceutica NV; Johnson & Johnson (China) Investment Ltd.

WO2021121327

WO2022237719

PATENT

WO2022237720

PATENTS

PATENT

US20240261292

Compound A—(R)-N-ethyl-5-fluoro-N-isopropyl-2-((5-(2-(6-((2-methoxyethyl) (methyl)amino)-2-methylhexan-3-yl)-2,6-diazaspiro[3.4]octan-6-yl)-1,2,4-triazin-6-yl)oxy) benzamide

      
 (MOL) (CDX)
      The mixture of (R)-N-ethyl-5-fluoro-N-isopropyl-2-((5-(2-(6-((2-methoxyethyl)amino)-2-methylhexan-3-yl)-2,6-diazaspiro [3.4]octan-6-yl)-1,2,4-triazin-6-yl)oxy)benzamide (compound 11) (40.0 mg, 0.068 mmol), formaldehyde (55.4 mg, 0.683 mol, 37% in water) and AcOH (8.2 mg, 0.137 mmol) in anhydrous MeOH (2 mL) was stirred at 45° C. for 1 h. Then, NaBH 3CN (8.6 mg, 0.137 mmol) was added to the mixture and the resulting mixture was stirred at 45° C. for another 1 h. After cooling to RT, the reaction mixture was treated with sat. aq. NaHCO (40 mL) to adjust the pH value to about 8 and further extracted with DCM (20 mL×3). The combined organic layers were dried over anhydrous Na 2SO 4, filtered and concentrated under reduced pressure to give the crude which was purified by preparative HPLC over Boston Prime (column: C18 150×30 mm Sum, Mobile Phase A: H 2O (0.04% ammonia+10 mM NH 4HCO 3), Mobile Phase B: ACN, Flow rate: 25 m/min, gradient condition B/A from 50% to 80% (50% B to 80% B)) to afford the title compound (9.62 mg, 99.10% purity, 23.3% yield) as yellow oil.

PATENT

WO2022262796

The present invention is directed to (R) -N-ethyl-5-fluoro-N-isopropyl-2- ( (5- (2- (6- ( (2-methoxyethyl) (methyl) amino) -2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) benzamide besylate salt (benzenesulfonate salt) :

[0011]

Preparation of Compound 61

[0140]

tert-butyl (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate

[0141]

[0142]

The mixture 2- ( (5- (2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) -N-ethyl-5-fluoro-N-isopropylbenzamide (intermediate 3) (1.0 g, 2.4 mmol) , tert-butyl (5-methyl-4-oxohexyl) carbamate (intermediate 1) (830 mg, 3.62 mmol) and ZnCl 2(660 mg, 4.84 mmol) in MeOH (15 mL) was stirred at 80 ℃ for 0.5 h. Then NaBH 3CN (310 mg, 4.93 mmol) was added and the resulting mixture was stirred at 80 ℃ for 6 h. After cooled to RT, the mixture was concentrated under reduced pressure to give the crude product, which was further purified by preparative HPLC using a Waters Xbridge Prep OBD (column: C18 150×40 mm 10 um; eluent: ACN/H 2O (0.05%ammonia) from 45%to 75%v/v) to afford the title compound (700 mg, 46%yield) as colorless oil.

reparation of Compounds 62 and 63

[0144]

tert-butyl (R) – (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate

[0145]

tert-butyl (S) – (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate

[0146]

[0147]

tert-butyl (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate (Compound 61) (200 mg, 0.319 mmol) was purified by SFC over DAICEL CHIRALPAK IG (column: 250×30 mm 10 um; isocratic elution: EtOH (containing 0.1%of 25%ammonia) : supercritical CO 2, 40%: 60% (v/v) ) to afford the title compounds (Compound 62) (85 mg, 42%yield) and (Compound 63) (80 mg, 40%yield) both as light yellow oil.

[0148]

Compound 64

[0149]

(R) -2- ( (5- (2- (6-amino-2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) -N-ethyl-5-fluoro-N-isopropylbenzamide

[0150]

[0151]

To the solution of tert-butyl (R) – (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate (Compound 62) (550 mg, 0.876 mmol) in DCM (4 mL) was slowly added TFA (4 mL) , and the resulting mixture was stirred at 25 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted in DCM (40 mL) and the pH value was adjusted to around 12 by aq. NaOH (2 M, 16 mL) solution. The aqueous layer was extracted with DCM (10 mL x 2) . The combined organic layers were dried over anhydrous Na 2SO 4, filtered and concentrated in vacuo to afford the title compound (460 mg, crude) as yellow solid, which was used directly in next step without further purification.

[0152]

Compound 11

[0153]

(R) -N-ethyl-5-fluoro-N-isopropyl-2- ( (5- (2- (6- ( (2-methoxyethyl) amino) -2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) benzamide

[0154]

[0155]

The mixture of (R) -2- ( (5- (2- (6-amino-2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) -N-ethyl-5-fluoro-N-isopropylbenzamide (Compound 64) (120 mg, crude) , 1-bromo-2-methoxyethane (32 mg, 0.23 mmol) , Cs 2CO 3(222 mg, 0.681 mmol) , NaI (102 mg, 0.680 mmol) in DMF (1 mL) was stirred at 80 ℃ via microwave irradiation for 1 h. After cooling to RT, the mixture was diluted with H 2O (10 mL) and extracted with EtOAc (3 x 10 mL) . The combined organic layers were washed with H 2O (10 mL) , dried over Na 2SO 4, filtered and concentrated under reduced pressure to afford the crude product which was further purified by HPLC over a Phenomenex Gemini-NX (column: 150×30 mm 5 μm; eluent: ACN/H 2O (10mM NH 4HCO 3) from 51%to 71% (v/v) ) and further purified by SFC over DAICEL CHIRALCEL OD-H (column: 250×30 mm 5 um; eluent: supercritical CO 2in EtOH (0.1%v/v ammonia) 25/25, v/v) to afford the title compound (5.13 mg, 96%purity) as yellow solid.

[0156]

LC-MS (ESI) (Method 1) : R t= 2.997 min, m/z found 586.3 [M+H] +.

[0157]

Compound A

[0158]

(R) -N-ethyl-5-fluoro-N-isopropyl-2- ( (5- (2- (6- ( (2-methoxyethyl) (methyl) amino) -2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) benzamide

[0159]

[0160]

The mixture of (R) -N-ethyl-5-fluoro-N-isopropyl-2- ( (5- (2- (6- ( (2-methoxyethyl) amino) -2- methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) benzamide (Compound 11) (40.0 mg, 0.068 mmol) , formaldehyde (55.4 mg, 0.683 mol, 37%in water) and AcOH (8.2 mg, 0.137 mmol) in anhydrous MeOH (2 mL) was stirred at 45 ℃ for 1 h. Then, NaBH 3CN (8.6 mg, 0.137 mmol) was added to the mixture and the resulting mixture was stirred at 45 ℃ for another 1 h. After cooling to RT, the reaction mixture was treated with sat. aq. NaHCO 3(40 mL) to adjust the pH value to about 8 and further extracted with DCM (20 mL x 3) . The combined organic layers were dried over anhydrous Na 2SO 4, filtered and concentrated under reduced pressure to give the crude which was purified by preparative HPLC over Boston Prime (column: C18 150x30mm 5um, Mobile Phase A: H 2O (0.04%ammonia+10mM NH 4HCO 3) , Mobile Phase B: ACN, Flow rate: 25 mL/min, gradient condition B/A from 50%to 80% (50%B to 80%B) ) to afford the title compound (9.62 mg, 99.10%purity, 23.3%yield) as yellow oil.

////////Bleximenib, CS-0636752, DA-55335, HY-148669, JNJ-75276617, Menin-MLL inhibitor 24

Tegomil fumarate


Tegomil fumarate

cas 1817769-42-8

dimethyl (2E,19E)-4,18-dioxo-5,8,11,14,17-pentaoxahenicosa-2,19-diene-1,21-dioate

4-O-[2-[2-[2-[2-[(E)-4-methoxy-4-oxobut-2-enoyl]oxyethoxy]ethoxy]ethoxy]ethyl] 1-O-methyl (E)-but-2-enedioate

Chemical Formula: C18H26O11
Exact Mass: 418.15
Molecular Weight: 418.395
Elemental Analysis: C, 51.67; H, 6.26; O, 42.06

MXD6KMG2ZP

SCHEME

Patent

Ratiopharm GmbH, WO2015158817

PATENT

WO2017108960

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017108960

Scheme 2: Synthesis of (E)-But-2-enedioic acid 2-(2-{2-[2-((E)-3-methoxycarbonyl- acryloyloxy)-ethoxy]-ethoxy}-ethoxy)-ethyl ester methyl ester

Step 1: Synthesis of (Z)-But-2-enedioic acid mono-[2-(2-{2-[2-((Z)-3- carboxy-acryloyloxy)-ethoxy]-ethoxy}-ethoxy)-ethyl] ester

///////////Tegomil fumarate, MXD6KMG2ZP

Bivamelagon


Bivamelagon

CAS 2641595-54-0

NO1Y8WRA8N, 629.3 g/mol, C35H53ClN4O4

MC-4R Agonist 2

  • N-[(3S,5S)-1-[[(3S,4R)-4-(4-Chlorophenyl)-1-(1,1-dimethylethyl)-3-pyrrolidinyl]carbonyl]-5-(4-morpholinylcarbonyl)-3-pyrrolidinyl]-2-methyl-N-(cis-4-methylcyclohexyl)propanamide
  • N-((3S,5S)-1-((3S,4R)-1-(tert-Butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-5-(morpholine-4-carbonyl)pyrrolidin-3-yl)-N-(cis-4-methylcyclohexyl)isobutyramide
  • N-[(3S,5S)-1-[(3S,4R)-1-tert-butyl-4-(4-chlorophenyl)pyrrolidine-3-carbonyl]-5-(morpholine-4-carbonyl)pyrrolidin-3-yl]-2-methyl-N-(4-methylcyclohexyl)propanamide

LB54640; LB-54640; LR-19021; LR19021

MC-4R Agonist 2 (Example 1) is a MC4R agonist. MC-4R Agonist 2 can be used in the study of obesity, diabetes, inflammation, and erectile dysfunction[1].

SCHEME

PATENT

WO2021091283A1.

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021091283&_cid=P21-M9NZN0-90342-1

Step D: Preparation of N -((3 S ,5 S )-1-((3 S ,4 R )-1-( tert -butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-5-(morpholine-4-carbonyl)pyrrolidin-3-yl)- N -((1 s ,4 R )-4-methylcyclohexyl)isobutyramide hydrochloride

[173]

 N -((3  S ,5  S )-1-((3  S ,4  R )-1-(  tert -butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-5-(morpholine-4-carbonyl)pyrrolidin-3-yl)-  N -((1  s ,4  R )-4-methylcyclohexyl)isobutyramide (5.0 g, 7.95 mmol) obtained in Step C was dissolved in ethyl acetate (50 ml), and a 2N hydrochloric acid ethyl acetate solution (3.97 ml, 15.89 mmol) was slowly added. After stirring at room temperature for 30 minutes, the reaction solvent was concentrated under reduced pressure. The resulting crude solid was purified by trituration using hexane and diethyl ether to obtain the title compound (5.23 g, 99%).

[174]

MS [M+H] = 630 (M+1)

[175]

1H NMR (500 MHz, CD 3OD) δ 7.49-7.44 (m, 4H), 4.83 (m, 1H), 4.23-4.20 (m, 1H), 3.95-3.91 (m, 2H), 3.79-3.47 (m, 14H), 3.03-3.00 (m, 1H), 2.86-2.82 (m, 1H), 2.73-2.67 (m, 1H), 2.20-2.14 (m, 1H), 1.97 (m, 1H), 1.80-1.62 (m, 5H), 1.50 (s, 9H), 1.44-1.27 (m, 3H), 1.06-1.04 (m, 9H)

PATENT

WO2022235103

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2022235103&_cid=P21-M9NZHZ-87240-1

Preparation of cyclohexyl-3-carbonyl-l)-5-(morpholine-4-carbonyl)pyrrolidin-3-yl)-N-((1s,4R)-4-methylcyclohexyl)isobutyramide (MC70)

[141]

[142]The title compound was obtained through the following steps A, B, and C. 

[143]

Step A: Preparation of methyl (2S,4S)-1-((3S,4R)-1-(tert-butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-4-(N-((1s,4R)-4-methylcyclohexyl)isobutyramido)pyrrolidine-2-carboxylate

[144]Methyl (2S,4S)-4-(N-((1s,4R)-4-methylcyclohexyl)isobutyramido)pyrrolidine-2-carboxylate hydrochloride (28.7 g, 82.73 mmol) obtained in Manufacturing Example 1, (3S,4R)-1-(tert-butyl)-4-(4-chlorophenyl)pyrrolidine-3-carboxylic acid (24.5 g, 86.87 mmol), 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (22.2 g, 115.83 mmol) and 1-hydroxybenzotriazole hydrate (15.7 g, 115.83 mmol) obtained in Manufacturing Example 2 were dissolved in N,N’-dimethylformamide (400 ml) and N,N’-diisopropylethylamine (72.0 ml, 413.66 mmol) was slowly added. The mixture was stirred at room temperature for 16 hours and the reaction solvent was concentrated under reduced pressure. A 0.5 N aqueous sodium hydroxide solution was added, and extraction was performed twice with ethyl acetate. The organic layer was washed twice with an aqueous sodium chloride solution and water, dried over anhydrous magnesium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was purified by column chromatography to obtain methyl (2S,4S)-1-((3S,4R)-1-(tert-butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-4-(N-((1s,4R)-4-methylcyclohexyl)isobutyramido)pyrrolidine-2-carboxylate (41.19 g, 87%). 

[145]

MS [M+H] = 575 (M+1)

[146]

Step B: Preparation of (2S,4S)-1-((3S,4R)-1-(tert-butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-4-(N-((1s,4R)-4-methylcyclohexyl)isobutyramido)pyrrolidine-2-carboxylic acid

[147]Methyl (2S,4S)-1-((3S,4R)-1-(tert-butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-4-(N-((1s,4R)-4-methylcyclohexyl)isobutyramido)pyrrolidine-2-carboxylate (39.4 g, 68.62 mmol) obtained in the above step A was dissolved in methanol (450 ml), and 6N sodium hydroxide aqueous solution (57.2 ml, 343.09 mmol) was added. The mixture was stirred at room temperature for 16 hours, and the pH was adjusted to about 5 with 6N hydrochloric acid aqueous solution, and then the reaction solution was concentrated under reduced pressure. The concentrate was dissolved in dichloromethane, and the insoluble solid was filtered through a paper filter. The filtrate was concentrated under reduced pressure to obtain the crude title compound (38.4 g, 99%), which was used in the next step without purification. 

[148]

MS [M+H] = 561 (M+1)

[149]

Step C: Preparation of N-((3S,5S)-1-((3S,4R)-1-(tert-butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-5-(morpholine-4-carbonyl)pyrrolidin-3-yl)-N-((1s,4R)-4-methylcyclohexyl)isobutyramide

[150](2S,4S)-1-((3S,4R)-1-(tert-butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-4-(N-((1s,4R)-4-methylcyclohexyl)isobutyramido)pyrrolidine-2-carboxylic acid (38.4 g, 68.60 mmol), 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (18.4 g, 96.04 mmol) and 1-hydroxybenzotriazole hydrate (13.0 g, 96.04 mmol) obtained in the above step B were dissolved in N,N’-dimethylformamide (200 ml), and then morpholine (5.9 ml, 68.80 mmol) and N,N’-diisopropylethylamine were sequentially added. (59.7 ml, 343.02 mmol) was slowly added. The mixture was stirred at room temperature for 16 hours and the reaction solution was concentrated under reduced pressure, 0.5 N aqueous sodium hydroxide solution was added, and extraction was performed twice with ethyl acetate. The organic layer was washed twice with aqueous sodium chloride solution and water, dried over anhydrous magnesium sulfate, and filtered. The filtrate was concentrated under reduced pressure and purified by column chromatography to obtain N-((3S,5S)-1-((3S,4R)-1-(tert-butyl)-4-(4-chlorophenyl)pyrrolidine-3-carbonyl)-5-(morpholine-4-carbonyl)pyrrolidin-3-yl)-N-((1s,4R)-4-methylcyclohexyl)isobutyramide (37.05 g, 86%,  

MC70  ). 

[151]

MS [M+H] = 630 (M+1)

Bivamelagon (INNTooltip International Nonproprietary Name; developmental code names LB54640LR-19021) is a small-molecule melanocortin MC4 receptor agonist under development by LG Chem Life Sciences for the treatment of hypothalamic obesity, .[1][2] Unlike the older drug with the same mechanism of actionsetmelanotide, it can be taken orally.[3][4][5] As of March 2024, it is in phase 2 clinical trials.[1]

References

  1. Jump up to:a b “Rhythm Pharmaceuticals”AdisInsight. 13 March 2024. Retrieved 25 February 2025.
  2. ^ “Delving into the Latest Updates on Bivamelagon with Synapse”Synapse. 23 January 2025. Retrieved 25 February 2025.
  3. ^ Aronne, Sarah R. Barenbaum, Louis J. (2023). “Antiobesity Medications on the Horizon”. Handbook of Obesity – Volume 2 (5 ed.). CRC Press. pp. 394–401. doi:10.1201/9781003432807-42ISBN 978-1-003-43280-7.
  4. ^ First-in-Human Study of Safety, Pharmacodynamics of LB54640, An Oral Melanocortin-4 Receptor Agonist Mirza, Victoria, MD, MPH; Lee, Jisoo, MD; Gwak, Heemin; Yang, Yunjeong; Kim, Mina.  Obesity; Silver Spring Vol. 30, (Nov 2022): 145-146.
  5. ^ Piper, Noah B.C.; Whitfield, Emily A.; Stewart, Gregory D.; Xu, Xiaomeng; Furness, Sebastian G.B. (August 2022). “Targeting appetite and satiety in diabetes and obesity, via G protein-coupled receptors”. Biochemical Pharmacology202: 115115. doi:10.1016/j.bcp.2022.115115PMID 35671790S2CID 249452717.
Clinical data
Other namesLB54640; LB-54640; LR-19021; LR19021
Routes of
administration
Oral
Drug classMelanocortin MC4 receptor agonist
Legal status
Legal statusInvestigational
Identifiers
showIUPAC name
CAS Number2641595-54-0
PubChem CID165152355
DrugBankDB18331
ChemSpider129440355
UNIINO1Y8WRA8N
Chemical and physical data
FormulaC35H53ClN4O4
Molar mass629.28 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI

[1]. Seung Wan Kang, et al. Melanocortin-4 receptor agonists. Patent WO2021091283A1.

////////Bivamelagon, LB54640, LB-54640, LR-19021, LR19021, NO1Y8WRA8N

Tisolagiline


Tisolagiline

CAS 1894207-44-3

PCH79KLX33

(2S)-2-[[4-[4-(trifluoromethyl)phenyl]phenyl]methylamino]propanamide

322.32 g/mol

SCHEME

Tisolagiline (INNTooltip International Nonproprietary Name; developmental code names KDS-2010SeReMABI) is a potent, highly selective, and reversible monoamine oxidase B (MAO-B) inhibitor which is under development for the treatment of Alzheimer’s disease and obesity.[1][2][3][4] It is taken by mouth.[1] Tisolagiline is being developed by NEUROBiOGEN and Scilex Bio.[1][2] As of December 2024, it is in phase 2 clinical trials for Alzheimer’s disease and obesity.[1][2]

Parkinson’s disease is a progressive disease that ranks second among degenerative neurological diseases, and the incidence rate is estimated to be about 6.3 million patients worldwide, and about 1 in 1,000 people develop Parkinson’s disease. The incidence rate is usually higher in the elderly, but it is now developing in young people as well. Parkinson’s disease is not easy to distinguish from other diseases because the symptoms progress slowly, and it is difficult to detect in the early stages. Clinical characteristics include tremors, rigidity, bradykinesia, postural instability, stooped posture, freezing of gait, depression, sleep disorders, urination disorders, and dementia. 

[3]Parkinson’s disease has an unknown cause, but it is known to be a disease that occurs when nerve cells that secrete the neurotransmitter dopamine in the brain are destroyed, resulting in a lack of dopamine. The most widely developed and used drug is levodopa therapy, which is generally administered by administering levodopa, which is converted into dopamine in the body. Levodopa is the most effective treatment for Parkinson’s disease, but there are cases where the drug-related effects decrease or various movement disorders occur during the treatment process. Other drugs used include COMT inhibitors and MAO-B inhibitors, which suppress dopamine metabolism and maintain the concentration of dopamine in the brain. 

[4]MAO-B is known to play an important role in dopamine metabolism in the brain and to suppress damage to brain neurons. Although there is no clear evidence that MAO-B inhibitors actually slow down the progression of Parkinson’s disease, it is known that inhibiting MAO-B has an effect of suppressing degeneration or death of dopamine neurons, as it plays an important role in the development of Parkinson’s disease caused by MPTP or similar environmental toxicants. In addition, evidence from animal and clinical trials suggests that MAO-B inhibitors have a brain protective effect, unlike other drugs. 

[5]The most representative MAO-B inhibitor approved is selegiline, which is prescribed as a treatment for Parkinson’s disease, but when taken, it is metabolized into amphetamine in the body, causing liver toxicity, and as an irreversible inhibitor, it has various side effects. Azilect, which contains rasagiline, was first marketed in Israel in 2005 and has recently been released in about 50 countries including Europe and the United States. Azilect does not have amphetamine side effects in the body when taken and is said to be more effective than other dopaminergic drugs. However, rasagiline, like selegiline, is an irreversible MAO-B inhibitor, so although it has an excellent MAO-B inhibition effect, it has the disadvantage of safety issues. Therefore, recently, drugs that are effective and can reversibly inhibit activity are being developed as alternatives to complement these shortcomings, but no notable reversible inhibitors have been prescribed to date. 

[6]Meanwhile, obesity is a medical condition in which excessive fat accumulates in the body to the extent that it has a negative impact on health. Excessive weight can appear in combination with various diseases as the remaining energy is accumulated excessively due to the difference between energy consumed and energy used. 

[7]Previous studies on the hypothalamus in relation to food regulation have focused on neurons that make up a portion of the brain, which has limited our understanding of the brain’s function in controlling food and obesity. Therefore, in order to comprehensively understand brain function, studies on glial cells, which make up the majority, must also be conducted in parallel. In addition, astrocytes, which are the most numerous among glial cells, have recently emerged as cells that can activate or inhibit surrounding neurons by secreting various signaling substances such as GABA (gamma-aminobutyric acid), glutamate, D-serine, and ATP. Astrocytes in the hypothalamus also interact closely with POMC (pro-opiomelanocortin) neurons and express leptin receptors, which can contribute to leptin signaling. 

[8]There are two groups of POMC neurons in the hypothalamus: those that induce appetite reduction and those that induce energy consumption. Under normal circumstances, astrocytes help activate nearby POMC neurons that induce energy consumption. However, in obese states, unlike normal astrocytes, they are transformed into reactive astrocytes due to excessive leptin signals, and putrescine is converted into GABA by MAO-B (mono-aminoxidase B) and secreted. In addition, POMC neurons that induce energy consumption express GABAa receptors outside the synapse containing a4, a5, and a6 subunits due to excessive leptin signals, and are affected by persistent GABA secreted from anti-responsive astrocytes. As a result, POMC neurons are inhibited, energy consumption is reduced, and fat accumulation occurs. 

[9]At this time, if MAOBI, the causal enzyme of GABA production, is inhibited, GABA production and secretion are inhibited, the inhibition of POMC neurons is relieved, and they are reactivated to promote energy consumption. However, POMC neurons that induce appetite reduction do not express GABAa receptors outside the synapse, so they are not continuously affected by GABA. Therefore, MAOBI inhibitors selectively act on POMC neurons that induce energy consumption and exhibit the effect of obesity treatment. However, most of the existing MAOBI inhibitors are irreversible inhibitors, and there is a problem that they are accompanied by various side effects. Accordingly, drugs that can reversibly inhibit MAOBI are being researched and developed, but no notable reversible MAOBI inhibitor that can effectively act on obesity has been prescribed to date.

REF

Regulatory Toxicology and Pharmacology (2020), 117, 104733

Toxicological Research (Cham, Switzerland) (2023), 39(4), 693-709

Combinatorial Chemistry & High Throughput Screening (2020), 23(9), 836-841 

KR2023027416,

WO2023022256

WO2023022256

WO2016052928

PATENT

WO2016052928

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2016052928&_cid=P20-M8XX0L-81795-1

Using L-Alaninamide hydrochloride or D-Alaninamide hydrochloride, a reductive amination reaction was performed with the compound of step (a) to obtain an imine compound (step b, reaction scheme 1b), which was then reduced with sodium cyanoborohydride to obtain an amine compound (step c, reaction scheme 1c). 

[112]Add 1.2 equivalents of Glycinamide hydrochloride or L-Alaninamide hydrochloride or D-Alaninamide hydrochloride or L-Valinamide hydrochloride or L-Leucinamide hydrochloride to anhydrous methanol at a concentration of 0.92 molar, and then add 1.5 equivalents of triethylamine. When the solution becomes transparent, add 1.0 equivalent of the aldehyde synthesized in step (a). After two hours, wash with ethyl acetate and distilled water. Dry the organic layer with sodium sulfate and concentrate in vacuo. Dissolve the concentrated reaction solution in anhydrous methanol at a concentration of 1.0 molar, and add 4.0 equivalents of sodium cyanoborohydride at 0 ℃. Then, react at room temperature for 18 hours, and after completion of the reaction, wash the reaction solution with ethyl acetate and distilled water. The organic layer was dried over sodium sulfate, concentrated in vacuo, and separated and purified using silica gel column chromatography.

References

  1. Jump up to:a b c d “KDS 2010”AdisInsight. 6 February 2025. Retrieved 24 February 2025.
  2. Jump up to:a b c “Delving into the Latest Updates on KDS-2010 with Synapse”Synapse. 23 January 2025. Retrieved 24 February 2025.
  3. ^ Nam MH, Sa M, Ju YH, Park MG, Lee CJ (April 2022). “Revisiting the Role of Astrocytic MAOB in Parkinson’s Disease”International Journal of Molecular Sciences23 (8): 4453. doi:10.3390/ijms23084453PMC 9028367PMID 354572724.4. KDS2010 A recently developed KDS2010, which is ~12,500-fold more selective to MAOB than MAOA, differentiates the role of MAOB from MAOA and reports that MAOB does not contribute to DA degradation [39]. KDS2010 is a potent (IC50 = 7.6 nM), and selective MAOB inhibitor named shows no known off-target effect (no other enzymes or channels causing >40% inhibition) or toxicity for 4 weeks of repeated dosing in non-human primates [16,41]. KDS2010 was turned out to be highly effective for alleviating the PD-related motor symptoms and PD-like pathology, including reactive astrogliosis, excessive astrocytic GABA, and nigrostriatal DAergic neuronal loss in multiple rodent models of PD [41]. Its clinical efficacy is still waiting to be tested in future studies.
  4. ^ Duarte P, Cuadrado A, León R (2021). “Monoamine Oxidase Inhibitors: From Classic to New Clinical Approaches”. Handbook of Experimental Pharmacology264: 229–259. doi:10.1007/164_2020_384ISBN 978-3-030-68509-6PMID 32852645KDS2010 is a novel compound highly potent and selective reversible MAO-B inhibitor (Fig. 2). It has demonstrated learning and memory improvements, promotion of synaptic transmission, and reduction of astrogliosis and astrocytic GABA levels in APP/presenilin 1 mice (Park et al. 2019).

Clinical data
Other namesKDS-2010; KDS2010; SeReMABI
Drug classReversible monoamine oxidase B (MAO-B) inhibitor
Identifiers
showIUPAC name
CAS Number1894207-44-3
PubChem CID132023446
ChemSpider128942408
UNIIPCH79KLX33
ChEMBLChEMBL5314546
Chemical and physical data
FormulaC17H17F3N2O
Molar mass322.331 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI

///////////Tisolagiline, PCH79KLX33

Bemfivastatin, PPD 10558, RBx 10558


Bemfivastatin, PPD 10558, RBx 10558

cas 805241-79-6

Molecular Weight588.67
FormulaC34H37FN2O6
  • PPD-10558 calcium salt
  • Ppd-10558(calcium salt)
  • 805241-64-9
  • ppd-10558 calcium
  • 3I8G750MW3
  • calcium;(3R,5R)-7-[2-(4-fluorophenyl)-4-[[4-(hydroxymethyl)phenyl]carbamoyl]-3-phenyl-5-propan-2-ylpyrrol-1-yl]-3,5-dihydroxyheptanoate
  • C68H72CaF2N4O12

Bemfivastatin (PPD 10558) is an orally active, HMG-CoA Reductase (HMGCR) inhibitor, also known as Statin. Bemfivastatin enhances the activity of liver extraction. Bemfivastatin exhibits little developmental toxicity effects in pregnant rats and rabbits via daily oral doses during organogenesis period. The no observed adverse effect level (NOAEL) are ≥320 mg/kg/day for rats developmental toxicity, 12.5 mg/kg/day for rabbits maternal toxicity, and 25 mg/kg/day for rabbits developmental toxicity, respectively. Bemfivastatin can be used for research on Statin-related hypercholesterolemic myalgia with inability to tolerate statins.

Korean Patent No. 10-1329113 describes a method for preparing (3R,5R)-7-[2-(4-fluorophenyl)-5-isopropyl-3-phenyl-4-[(4-hydroxymethylphenylamino)carbonyl]-pyrrol-1-yl]-3,5-dihydroxy-heptanoic acid hemicalcium salt, as shown in the following reaction scheme.

SCHEME

MAIN

PATENT

WO2020040614

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2020040614&_cid=P11-M8VDBE-14315-1

Step 3: Preparation of tert-butyl (3R,5R)-7-(2-(4-fluorophenyl)-4-((4-(hydroxymethyl)phenyl)carbamoyl)-5-isopropyl-3-phenyl-1H-pyrrol-1-yl)-3,5-dihydroxyheptanoate

[499]In step 2, tert-butyl 2-((4R,6R)-6-(2-(3-((4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)carbamoyl)-5-(4-fluorophenyl)-2-isopropyl-4-phenyl-1H-pyrrol-1-yl)ethyl)-2,2-dimethyl-1,3-dioxan-4-yl)acetate (5 g) was dissolved in methanol (37 ml) and THF (37 ml), 1 N HCl aqueous solution (37 ml) was added, and the mixture was stirred at room temperature for 2 hours. EA was added to the reaction solution, diluted, and washed several times with distilled water and brine. The extracted organic layer was dried over Na 

2 SO 

4 and filtered under reduced pressure. The filtrate was concentrated under reduced pressure, EA and hexane were added, and the mixture was purified by recrystallization to obtain the title compound. 

[500]White solid 4.6 g (yield quantitative); 

[501]

1H NMR (500 MHz, CDCl 3): 7.24-7.14 (m, 9H), 7.06 (d, J = 8.5 Hz, 2H), 6.99 (t, J = 8.5 Hz, 2H), 6.87 (br s, 1H), 4.57 (s, 2H), 4.45-4.08 (m, 2H), 3.96-3.90 (m, 1H), 3.75-3.71 (m, 1H), 3.58 (sep, J = 7.0 Hz, 1H), 2.32 (d, J = 6.5 Hz, 2H), 1.73-1.65 (m, 1H), 1.64-1.58 (m, 1H), 1.54 (d, J = 7.0 Hz, 6H), 1.45 (s, 9H), 1.27-1.22 (m, 2H), MH+ 645.

Step 4: Preparation of (3R,5R)-7-(2-(4-fluorophenyl)-5-isopropyl-3-phenyl-4-((4-hydroxymethylphenylamino)carbonyl)-pyrrol-1-yl)-3,5-dihydroxyheptanoic acid hemicalcium salt

[503]In step 3, tert-butyl (3R,5R)-7-(2-(4-fluorophenyl)-4-((4-(hydroxymethyl)phenyl)carbamoyl)-5-isopropyl-3-phenyl-1H-pyrrol-1-yl)-3,5-dihydroxyheptanoate (4.19 g) obtained was dissolved in MeOH (65 ml) and THF (65 ml), and stirred in an ice bath. NaOH pellets (5 eq, 1.3 g) were added, and the mixture was stirred for 1 more hour at room temperature. After concentrating the reaction solution under reduced pressure, distilled water (44 ml) was added until the formed solid was completely dissolved. After concentrating the reaction solution under reduced pressure, distilled water (430 ml) was added until the solid was completely dissolved. 1 M Ca(OAc) 

2 aqueous solution (3.6 ml) was slowly added dropwise, and the mixture was stirred for 15.5 hours at room temperature. After the generated solid was filtered under reduced pressure, it was washed several times with distilled water and the filtered solid was dried in an oven. 

[504]2.98 g of white solid (yield 76%); 

[505]

1H NMR (500 MHz, DMSO-d 6) δ 9.78 (br s, 1H), 7.46 (d, J = 8.5 Hz, 2H), 7.26-7.23 (m, 2H), 7.19 (t, J = 9.0 Hz, 2H), 7.15 (d, J = 8.5 Hz, 2H), 7.09-7.05 (m, 4H), 7.02-6.98 (m, 1H), 6.41 (br s, 1H), 5.04 (t, J = 5.5 Hz, 1H), 4.75 (br s, 1H), 4.39 (d, J = 5.5 Hz, 2H), 3.98-3.91 (m, 1H), 3.79-3.69 (m, 2H), 3.55-3.50 (m, 1H), 3.22 (sep, J = 7.0 Hz, 1H), 2.03 (dd, J = 15.0 Hz, 4.0 Hz, 1H), 1.90 (dd, J = 15.0 Hz, 8.0 Hz, 1H), 1.63-1.57 (m, 1H), 1.54-1.47 (m, 1H), 1.41-1.36 (m, 1H), 1.37 (d, J = 7.0 Hz, 6H), 1.23-1.16 (m, 1H), MH+ (acid+1) 589.

Step 5: Preparation of (3R,5R)-7-(2-(4-fluorophenyl)-5-isopropyl-3-phenyl-4-((4-hydroxymethylphenylamino)carbonyl)-pyrrol-1-yl)-3,5-dihydroxyheptanoic acid hemicalcium salt

[540]The title compound was prepared in the same manner as in step 4 of Example 15. 

[541]

1H NMR (500 MHz, DMSO-d 6) δ 9.78 (br s, 1H), 7.46 (d, J = 8.5 Hz, 2H), 7.26-7.23 (m, 2H), 7.19 (t, J = 9.0 Hz, 2H), 7.15 (d, J = 8.5 Hz, 2H), 7.09-7.05 (m, 4H), 7.02-6.98 (m, 1H), 6.41 (br s, 1H), 5.04 (t, J = 5.5 Hz, 1H), 4.75 (br s, 1H), 4.39 (d, J = 5.5 Hz, 2H), 3.98-3.91 (m, 1H), 3.79-3.69 (m, 2H), 3.55-3.50 (m, 1H), 3.22 (sep, J = 7.0 Hz, 1H), 2.03 (dd, J = 15.0 Hz, 4.0 Hz, 1H), 1.90 (dd, J = 15.0 Hz, 8.0 Hz, 1H), 1.63-1.57 (m, 1H), 1.54-1.47 (m, 1H), 1.41-1.36 (m, 1H), 1.37 (d, J = 7.0 Hz, 6H), 1.23-1.16 (m, 1H), MH+ (acid+1) 589.

KR2001835  63%

KR2016103248

/////////Bemfivastatin, PPD 10558,  PPD-10558, RBx-10558; PPD10558, RBx10558, PPD 10558, RBx 10558, bemfivastatin CA, RBx 10558

Umifoxolaner, ML 878


Umifoxolaner, ML 878

CAS 2021230-37-3

Molecular Weight643.86
FormulaC26H16ClF10N3O3
  • 4-[(5S)-5-[3-Chloro-4-fluoro-5-(trifluoromethyl)phenyl]-4,5-dihydro-5-(trifluoromethyl)-3-isoxazolyl]-N-[2-oxo-2-[(2,2,2-trifluoroethyl)amino]ethyl]-1-naphthalenecarboxamide (ACI)
  • 4-{(5S)-5-[3-chloro-4-fluoro-5-(trifluoromethyl)phenyl]-5-(trifluoromethyl)-4,5-dihydroisoxazol-3-yl}-N-{2-oxo-2-[(2,2,2-trifluoroethyl)amino]ethyl}naphthalene-1-carboxamide
  • ML 878
  • 4-[(5S)-5-[3-chloro-4-fluoro-5-(trifluoromethyl)phenyl]-5-(trifluoromethyl)-4H-1,2-oxazol-3-yl]-N-[2-oxo-2-(2,2,2-trifluoroethylamino)ethyl]naphthalene-1-carboxamide
  • WHO 11642

umifoxolaner (ML-878) is a γ-aminobutyric acid (GABA) regulated chloride channels antagonist. Umifoxolaner is an anti-parasitic agent

Animals such as mammals and birds are often susceptible to parasite infestations/infections. These parasites may be ectoparasites, such as insects, and endoparasites such as filariae and other worms. Domesticated animals, such as cats and dogs, are often infested with one or more of the following ectoparasites:

– fleas (e.g. Ctenocephalides spp., such as Ctenocephalides felis and the like);

– ticks (e.g. Rhipicephalus spp., Ixodes spp., Dermacentor spp., Amblyomma spp., and the like);

– mites (e.g. Demodex spp., Sarcoptes spp., Otodectes spp., and the like);

– lice (e.g. Trichodectes spp., Cheyletiella spp., Linognathus spp. and the like);

– mosquitoes (Aedes spp., Culex spp., Anopheles spp. and the like); and

– flies (Haematobia spp., Musca spp., Stomoxys spp., Dermatobia spp., Cochliomyia spp. and the like).

Fleas are a particular problem because not only do they adversely affect the health of the animal or human, but they also cause a great deal of psychological stress. Moreover, fleas are also vectors of pathogenic agents in animals and humans, such as dog tapeworm {Dipylidium caninum).

Similarly, ticks are also harmful to the physical and psychological health of the animal or human. However, the most serious problem associated with ticks is that they are the vector of pathogenic agents in both humans and animals. Major diseases which are caused by ticks include borreliosis (Lyme disease caused by Borrelia burgdorferi), babesiosis (or piroplasmosis caused by Babesia spp.) and rickettsioses (also known as Rocky Mountain spotted fever). Ticks also release toxins which cause inflammation or paralysis in the host. Occasionally, these toxins are fatal to the host.

Likewise, farm animals are also susceptible to parasite infestations. For example, cattle are affected by a large number of parasites. A parasite which is very prevalent among farm animals is the tick genus Rhipicephalus {Boophilus), especially those of the species microplus (cattle tick), decolor atus and annulatus. Ticks, such as Rhipicephalus {Boophilus) microplus, are particularly difficult to control because they live in the pasture where farm animals graze.

Animals and humans also suffer from endoparasitic infections including, for example, helminthiasis which is most frequently caused by a group of parasitic worms categorized as cestodes (tapeworm), nematodes (roundworm) and trematodes (flatworm or flukes). These parasites adversely affect the nutrition of the animal and cause severe economic losses in pigs, sheep, horses, and cattle as well as affecting domestic animals and poultry. Other parasites which occur in the gastrointestinal tract of animals and humans include Ancylostoma, Necator, Ascaris, Strongyloides, Trichinella, Capillaria, Toxocara, Toxascaris, Trichuris, Enterobius and parasites which are found in the blood or other tissues and organs such as filarial worms and the extra intestinal stages of Strongyloides, Toxocara and Trichinella.

SCHEME

Patents

WO2017176948

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017176948&_cid=P12-M8S60W-88110-1

Cinchonanium, 9-hydroxy-6′-methoxy-1-[[3,4,5-tris(phenylmethoxy)phenyl]methyl]-, chloride (1:1), (8α,9R)- 2138407-51-7,  HYDROXYL AMINE, NAOH, MDC , WATER]

Example 5: Synthesis of (R)-IA-3 using chiral phase transfer catalyst (IIIb-13-1)

Step 1 : Synthesis of intermediate 4-2.

1) The substituted iodobenzene starting material (SM) (200.0 g, 1.0 eq.) and THF (400 ml, 10 volumes) were placed into a 1 L reactor and the mixture was cooled to -10 to -5° C.

2) /-PrMgCl (340 ml, 1.1 eq.) added dropwise over 1.5 hours at -10 to -5°C to the mixture. 3) After the addition was complete, the mixture was stirred for 1 h at -10 to -5°C.

4) TLC analysis showed the complete consumption of SM (quenching sample with 1 M HCl).

5) CF3COOMe (94.7 g, 1.2 eq.) was added over an hour at -10~-5°C to the reaction mixture.

6) The mixture was stirred for another 12 hours -10~-5°C.

7) TLC analysis showed the almost complete consumption of intermediate 4-1 (quench with 1M HCl).

8) 1 M HCl 1000 ml was added dropwise to the reaction mixture slowly at 0~5°C over 2 hours.

9) The reaction mixture was extracted with hexane twice (1000 ml, 500 ml).

10) Add ^-toluenesulfonic acid 1.0 g to the organic layer and then the mixture was refluxed for 30 min.

11) The resulting mixture was then concentrated under vacuum at 20~25°C to remove the hexane.

12) Sodium bicarbonate (NaHC03, 300mg) was added and the mixture distilled in vacuum to afford compound 4-2 at 80~82°C, as a red liquid (85.0 grams, purity was 92.5% by HPLC, and the yield was 47.0%).

Step 2: Preparation of the compound of Formula (IIA-3):

4-1 IIA-3

1) Compound 4-2 (70.0 g, 1.0 eq.) and acetonitrile (ACN, 350ml, 5 volumes) were placed into a 1 L reactor. The solid was dissolved completely.

2) Compound 4-1 (70.2 g, 1.2 eq.) was then added to the mixture, and the mixture was heated to 90-95° C.

3) The ACN/water azeotrope was removed by distillation (b.p. 79°C).

4) K2C03 (2.0 g, 0.1 eq.) was then added to the mixture.

5) Distillation was continued to remove ACN/water at 90~95°C for about 6 hours.

6) After this time, about 28% Compound 4-2 remained by HPLC.

7) The mixture was cooled to 15~20°C over 1.5 hours and solid precipitated.

8) Water (50 ml) was added and then the mixture was cooled further to 0° C over 40 min.

9) The mixture was then held at 0° C for 40 minutes.

10) The mixture was filtered and the cake was washed with 100 ml of cold ACN/water (ACN/water, 25:6v/v) to yield 75.0 g of a yellow solid after drying (purity: 95.1%, yield: 50.0%).

Step 3 : Preparation of (R)-IA-3 using chiral phase transfer catalyst IIIb-13-1

1) The Compound of Formula IIA-3 (40.0 g, 1.0 eq.) and DCM (1.2 L, 30 volumes) were placed into a 2 L reactor; the solid was dissolved completely.

2) The mixture was cooled to 0° C and some starting material precipitated out.

3) The catalyst of formula IIIb-13-1 (1.47g, 3% mol) was added to the mixture and the mixture was cooled to -10° C.

4) Hydroxylamine (21. Og, 5.0 eq., 50% in water) was added to a solution of NaOH (15.3 g, 6.0 eq., in 5 volumes of water) in another reactor and stirred for 30 minutes.

5) The hydroxylamine/NaOH solution was then added dropwise to the 2 L reactor over about 4 hours.

6) The resulting reaction mixture was stirred for 16 h at -10°C.

7) In-process samples were taken and analyzed by HPLC until the content of starting material was < 1.0%.

8) When the reaction was complete, the mixture was warmed to 10°C and 200 ml of water was added. The mixture was stirred for 10 minutes.

9) After mixing, the mixture was allowed to stand to separate the aqueous and organic layers and the organic layer was collected.

10) The organic layer was washed with 200 ml of 5% KH2PO4.

11) The two layers were allowed to separate and organic layer was collected.

12) The organic layer was then washed with 200 ml brine, the two layers allowed to separate and the organic layer was again collected.

13) The resulting organic layer was concentrated under vacuum at 25-30°C to about 2 volumes.

14) Toluene (400 ml, 10 volumes) was charged to the vessel and concentration under vacuum was continued at 40~45°C to about 3 volumes. The solvent exchange was repeated twice more using the same procedure.

15) When the solvent exchange was complete, the solution was heated to 55-60°C.

16) The mixture was then cooled to 40° C over 1.5 hours and stirred at 40°C for 3 hours.

17) The mixture was then cooled to 25°C over 2 hours and stirred at 25°C for 3hours.

18) The mixture was finally cooled to 5-10°C over 1 hour and stirred at 8° C for 12 hours.

19) After this time, the mixture was filtered and the filter cake was washed with cold toluene (80 ml, 2 volumes).

20) The product was dried under vacuum at 70~75°C for 12h to yield a white solid (22.0 g, chiral purity: 98.0% by area using the chiral HPLC method described in Example 3, chemical purity: 97.1% by area (HPLC), yield: 48.8%). The 1H MR and LCMS spectra are consistent with the structure of the product.

Example 6: Preparation of (S)-IA-3 using chiral phase transfer catalyst IIIa-13-1

) The compound of Formula IIA-3 (11.6 g, 1.0 eq.) and DCM 360 ml, 30 volumes) were placed into a 1 L reactor; the solid was dissolved completely.

) The mixture was cooled to 0°C and some starting material was precipitated out.

) The catalyst (0.43 g, 3% mol) was added to the resulting mixture, and the mixture was cooled to -10° C.

) Hydroxylamine (6.1 g, 5.0 eq., 50% in water) was added to a solution of NaOH (4.4 g, 6.0 eq., in 5 volumes of water) in another reactor, and the mixture was stirred for 30 minutes.

) The hydroxylamine and NaOH solution was added dropwise to the 1 L reactor over about 2 hours, after which the mixture was stirred for 16 h at -10° C.

) Samples were taken and analyzed by HPLC to monitor the extent of reaction until the content of starting material was < 1.0%.

) When the reaction was complete, the mixture was warmed to 10°C and 50 ml of water was added. The mixture was stirred for 10 minutes.

) The mixture was allowed to settle to separate the aqueous and organic layers and the organic layer was collected.

) The organic layer was washed with 50 ml of 5% KH2PO4.

0) The mixture was allowed to separate and the organic layer was collected.

1) The organic layer was washed with 50 ml brine and the organic layer was again collected. 2) The organic layer was concentrated under vacuum at 25-30°C to about 2 volumes.

3) Toluene (230 ml, 10 volumes) was charged and concentration under vacuum was continued at 40~45°C to about 3 volumes. The solvent exchange was repeated twice more using the same procedure.

14) After the solvent exchange was complete, the solution was heated to 55-60°C.

15) The mixture was then cooled to 40° C over 1.5 hours and stirred at 40° C for 3 hours.

16) The mixture was cooled to 25° C over 2 hours and stirred at 25° C for 3 hours.

17) Finally, the mixture was cooled to 5-10° C over 1 hour and stirred at 8° C for 12 hours, after which the mixture was filtered.

18) The filter cake was washed with cold toluene (25 ml, 2 volumes).

19) The product was dried under vacuum at 85~90°C for 24h, resulting in the product as a white solid (6.8 g, chiral purity: 98.7% by area using the chiral FTPLC method described in Example 3, chemical purity: 99.3% by area (FTPLC), yield: 52.1%).

SEE ALSO US20170239218 

[1]. Cady, Susan Mancini; Cheifetz, Peter; Galeska, Izabela; Le Hir de Fallois, Loic.Long-acting injectable formulations comprising isoxazoline for prevention and treatment of parasitic infections.WO2016164487A1.

//////////Umifoxolaner, ML 878, ML878, CS072E2C38, ML-878, WHO 11642

Bavtavirine


Bavtavirine, CAS 1956373-71-9

  • KAJ2CK6ZYE
  • 4-((4-Amino-8-(4-((1E)-2-cyanoethenyl)-2,6-dimethylphenyl)-2-quinazolinyl)amino)benzonitrile
  • Benzonitrile, 4-((4-amino-8-(4-((1E)-2-cyanoethenyl)-2,6-dimethylphenyl)-2-quinazolinyl)amino)-

C26H20N6 416.48

Benzonitrile, 4-[[4-amino-8-[4-[(1E)-2-cyanoethenyl]-2,6-dimethylphenyl]-2-quinazolinyl]amino]-

Gilead Sciences, Inc.; Institute of Organic Chemistry and Biochemistry of the AS CR, v.v.i.

Bavtavirine is a potent non-nucleoside reverse transcriptase inhibitors (NNRTIs). Bavtavirine is part of highly active antitiretroviral therapy (HAART) treatment regimen. Bavtavirine can be used for HIV disease research.

SCHEME

PATENT

WO2016105564

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2016105564&_cid=P11-M8QXHF-67832-1

A mixture of compound 2a (100 mg, 0.30 mmol), 4-cyanoaniline (46 mg, 0.388 mmol, Sigma-Aldrich) and hydrogen chloride solution in 1,4-dioxane (4M, 7 μL, 0.03 mmol) in dry N-methyl-2-pyrrolidone (2 mL) was heated at 120 °C for 2 hours. The reaction mixture was cooled down to room temperature and triethylamine (0.1 mL, 0.72 mmol) was added. After 15 minutes, water (5 mL) was added and the solid product was filtered off and washed with water. The crude residue was taken up in a mixture of dichloromethane and diethyl ether (1:1,5 mL) and then treated in a sonic bath for 3 minutes. The solid compound was filtered off and washed with diethyl ether (5 mL) to afford the title compound 2. 1H NMR (400 MHz, DMSO-d6) δ 9.44 (s, 1H), 8.18 (dd, J = 8.2, 1.5 Hz, 1H), 7.74 (d, J = 16.7 Hz, 1H), 7.70 (d, J = 8.9 Hz, 2H), 7.51 (s, 2H), 7.48 (dd, J = 7.1, 1.3 Hz, 1H), 7.34 (dd, J = 8.2, 7.1 Hz, 1H), 7.26 (d, J = 8.9 Hz, 2H), 6.54 (d, J = 16.7 Hz, 1H), 1.91 (s, 6H). HRMS: (ESI+) calculated for C26H2,N6 [M+H] 417.1822, found 417.1820. LCMS (m/z) 417.2 [M+H], Tr = 4.68 min (LCMS method 1).

[1]. Jansa P, et al. Quinazoline derivatives used to treat hiv. The United States, WO2016105564 A1. 2016-06-30.

////////////Bavtavirine

Uplarafenib


Uplarafenib

1425485-87-5

494.5 g/mol

Molecular FormulaC22H21F3N4O4S
Molecular Weight494.487
  • B-Raf IN 10
  • TQU3V7CXC3
  • N-[2,4,5-trifluoro-3-(3-morpholin-4-ylquinoxaline-6-carbonyl)phenyl]propane-1-sulfonamide
  • B-Raf IN 10; B-Raf IN-10; B-Raf-IN-10

UPLARAFENIB is a small molecule drug with a maximum clinical trial phase of II and has 1 investigational indication. Neupharma, Inc.

 There are at least 400 enzymes identified as protein kinases. These enzymes catalyze the phosphorylation of target protein substrates. The phosphorylation is usually a transfer reaction of a phosphate group from ATP to the protein substrate. The specific structure in the target substrate to which the phosphate is transferred is a tyrosine, serine or threonine residue. Since these amino acid residues are the target structures for the phosphoryl transfer, these protein kinase enzymes are commonly referred to as tyrosine kinases or serine/threonine kinases.

The phosphorylation reactions, and counteracting phosphatase reactions, at the tyrosine, serine and threonine residues are involved in countless cellular processes that underlie responses to diverse intracellular signals (typically mediated through cellular receptors), regulation of cellular functions, and activation or deactivation of cellular processes. A cascade of protein kinases often participate in intracellular signal transduction and are necessary for the realization of these cellular processes. Because of their ubiquity in these processes, the protein kinases can be found as an integral part of the plasma membrane or as cytoplasmic enzymes or localized in the nucleus, often as components of enzyme complexes. In many instances, these protein kinases are an essential element of enzyme and structural protein complexes that determine where and when a cellular process occurs within a cell.

The identification of effective small compounds which specifically inhibit signal transduction and cellular proliferation by modulating the activity of tyrosine and serine/threonine kinases to regulate and modulate abnormal or inappropriate cell proliferation, differentiation, or metabolism is therefore desirable. In particular, the identification of compounds that specifically inhibit the function of a kinase which is essential for processes leading to cancer would be beneficial

SCHEME

Patent

Compound A [WO2022119905A2]

WO2022119905 69%

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2022119905&_cid=P20-M8O7NY-07177-1

Example 1: Preparation of N-(2,4,5-trifluoro-3-(3-morpholinoquinoxaline-6-carbonyl)phenyl)propane-l-sulfonamide (Compound A)

[181] Step l : To a solution of quinoxalin-2(lH)-one (54.64 g, 374 mmol, 1.0 eq.) in HO Ac (1000 mL) was added a solution of Bn (19.18 mL, 374 mmol, 1.0 eq.) in HOAc (200 mL) dropwise. The resulting mixture was stirred at rt for 12 h, then poured into ice-water. The precipitate was collected by filtration and dried to afford 7-bromoquinoxalin-2(lH)-one as an off-white solid (74 g, 88%).

[182] Step l : To a suspension of 7-bromoquinoxalin-2(lH)-one (224 g, 1 mol, 1.0 eq.) in POCl3 (1000 mL) was added DMF (3.65 g, 0.05 mol, 0.05 eq.). The resulting mixture was stirred at 120 °C for 2 h, then cooled to rt and slowly poured into ice-water with vigorous stirring. The precipitate was collected by filtration and dried to afford 7-bromo-2-chloroquinoxaline as brown solid (180 g, 75%).

[183] Step 3 : To a solution of 7-bromo-2-chloroquinoxaline (50 g, 0.2mol, 1.0 eq.) in CH3CN (200 mL) were added morpholine (89 g, 1.02 mol, 5.0 eq.) and K2CO3 (85 g, 0.61mol, 3.0 eq). The resulting mixture was stirred at 90 °C for 2 h, then cooled and filtered. The filtrate was concentrated and the residue was re-crystallized from EA to afford 4-(7-bromoquinoxalin-2-yl)morpholine (59 g, 98.3%).

[184] Step 4 : To a solution of 4-(7-bromoquinoxalin-2-yl)morpholine (59 g, 0.2 mol, 1.0 eq.) in DMF (500 mL) was added TEA (139 mL, 1.0 mol, 5.0 eq.), EtsSiH (127 mL, 0.8 mol, 4.0 eq) and Pd(dppf)C12.CH2C12 (8.16 g, 0.01 mol, 0.05 eq.). The resulting mixture was stirred at 90 °C for 12h in an autoclave under CO (1 MPa), then cooled and concentrated. The resulting residue was purified by flash column chromatography(EA/PE=l/l) to afford 3-morpholinoquinoxaline-6-carbaldehyde as a yellow solid (40 g, 82.3%).

[185] Step 5 : To a solution of N-(2,4,5-trifluorophenyl)pivalamide (550 mg, 2.4 mmol, E2 eq.) in THF (30 mL) cooled at -78 °C was added LDA (4.1 mL, 4.8mmol, 2.4 eq.) dropwise. The resulting mixture was stirred at -78 °C for 1 h, then a solution of 3-morpholinoquinoxaline-6-carbaldehyde (486 mg, 2.0 mol, 1.0 eq.) in THF (20 mL) was added dropwise. The resulting mixture was stirred at -78 °C for 1 h, then quenched by the addition of NH4CI solution. The mixture was extracted with EA (20 mL X 3) and the combined organic layers were dried over Na2SO4 and concentrated. The resulting residue was purified by flash column chromatography (MeOH/DCM=l/50, v/v) to afford N-(2,4,5-trifluoro-3-(hydroxy(3-morpholinoquinoxalin-6-yl)methyl)phenyl)pivalamide (620 mg, 65.2%).

[186] Step 6 : The solution of N-(2,4,5-trifluoro-3-(hydroxy(3-morpholinoquinoxalin-6-yl)methyl)phenyl)pivalamide (620 mg, 1.3 mmol, 1.0 eq.) in DCM (10 mL) was added MnCb (358 mg, 6.5 mmol, 5.0 eq.). The resulting mixture was stirred at 50 °C overnight, then cooled and filtered. The filtrate was concentrated and the residue was purified by flash column chromatography (PE/EA=l/2,v/v) to afford N-(2,4,5-trifluoro-3-(3-morpholinoquinoxaline-6-carbonyl)phenyl)pivalamide (560 mg, 90%).

[187] Step 7 : To a solution of N-(2,4,5-trifluoro-3-(3-morpholinoquinoxaline-6-carbonyl)phenyl)pivalamide (560 mg , 1.2 mmol, 1.0 eq. ) in HO Ac (10 mL) was added cone. HC1 (50 mL). The mixture was stirred at 110 °C for 4h, then poured onto ice. The mixture was adjusted to pH 10 by the addition of IN NaOH solution, then extracted with DCM (100 mL X 3). The combined organic layers were dried over Na2SO4 and concentrated. The resulting residue was purified by flash column chromatography (PE/EA=l/4,v/v) to afford (3-amino-2,5,6-trifluorophenyl)(3-morpholinoquinoxalin-6-yl)methanone as brown solid (410 mg, 88 % yield).

[188] Step 8 : To a solution of (3-amino-2,5,6-trifluorophenyl)(3-morpholinoquinoxalin-6-yl)methanone (40 mg, 0.1 mmol, 1.0 eq.) in DCM (10 mL) was added TEA (101 mg, 1 mol, 10 eq.) and propane- 1 -sulfonyl chloride (0.5 mL, 0.5 mmol, 5.0 eq.). The resulting mixture was stirred at rt for 1 h, then washed with water and extracted with DCM (lOmL X 3). The combined organic layers were dried over Na2SO4, filtered and concentrated. The resulting residue was purified by flash column chromatography (PE/EA=2/1, v/v) to afford N-(propylsulfonyl)-N-(2,4,5-trifluoro-3-(3-morpholinoquinoxaline-6-carbonyl)phenyl)propane-l-sulfonamide (41 mg, 62.2%).

[189] Step 9 : To a solution of N-(propylsulfonyl)-N-(2,4,5-trifluoro-3-(3-morpholinoquinoxaline-6-carbonyl)phenyl)propane-l -sulfonamide (41 mg, 0.068 mmol, 1.0 eq.) in MeOH/THF (10 mL /10 mL) was added 1 N NaOH (0.15 mmol, 2.2 eq.). The resulting mixture was stirred at rt for 1 h, then concentrated. The resulting residue was purified by flash column chromatography (PE/EA=l/l,v/v) to afford N-(2,4,5-trifluoro-3-(3-morpholinoquinoxaline-6-carbonyl)phenyl)propane-l -sulfonamide (Compound A) (23 mg, 68.9%). LRMS (M+H+) m/z calculated 495.1, found 495.1. XH NMR (CDCh, 400 MHz) 8 8.67 (s, 1 H), 7.98-8.03 (m, 3 H), 7.66-7.73 (m, 1 H), 6.72 (s, 1 H), 3.78-3.88 (m, 8H), 3.12-3.16 (t, 2 H), 1.87-1.92 (q, 2 H), 1.05-1.09 (t, 3 H).

Example 2. Preparation of Crystalline Form I of Compound A

[190] N-(2,4,5-trifluoro-3-(3-morpholinoquinoxaline-6-carbonyl)phenyl)propane-l-sulfonamide (2.53 kg) and ethyl acetate (EA) (9.1 kg) were added to the reactor. The mixture was stirred under refluxing for 2h. The solution was cooled to room temperature. The resulting precipitate was filtered, washed with EA (1 kg), and dried under vacuum at 45 °C to afford Crystalline Form I of N-(2,4,5-trifluoro-3-(3-morpholinoquinoxaline-6-carbonyl)phenyl)propane-1-sulfonamide (1.94 kg, 76.7%).

Example 3. Preparation of Crystalline Form II of Compound A

[191] N-(2,4,5-trifluoro-3-(3-morpholinoquinoxaline-6-carbonyl)phenyl)propane-l-sulfonamide (4.01 kg) was dissolved in EA (60 kg), and water (20 kg) was added. The organic phase was separated and concentrated to 4-6 kg under vacuum at 40-45 °C. The resulting residue was dissolved in EA (6 kg) and stirred for 4 hours at 10-20 oC. The solid was filtered, washed with EA (1.5 kg), and dried under vacuum at 50-55 oC to afford Crystalline Form II of N-(2,4,5-trifluoro-3 -(3 -morpholinoquinoxaline-6-carbonyl)phenyl)propane- 1 -sulfonami de (3.15 kg, 78.6%).

SEE

US20130053384 69%