New Drug Approvals
Follow New Drug Approvals on WordPress.com

FLAGS AND HITS

Flag Counter
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO

Archives

Categories

Join me on Linkedin

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

Join me on Researchgate

Anthony Melvin Crasto Dr.

  Join me on Facebook FACEBOOK   ...................................................................Join me on twitter Follow amcrasto on Twitter     ..................................................................Join me on google plus Googleplus

MYSELF

DR ANTHONY MELVIN CRASTO Ph.D ( ICT, Mumbai) , INDIA 36Yrs Exp. in the feld of Organic Chemistry,Working for AFRICURE PHARMA as ADVISOR earlier with GLENMARK PHARMA at Navi Mumbai, INDIA. Serving chemists around the world. Helping them with websites on Chemistry.Million hits on google, NO ADVERTISEMENTS , ACADEMIC , NON COMMERCIAL SITE, world acclamation from industry, academia, drug authorities for websites, blogs and educational contribution, ........amcrasto@gmail.com..........+91 9323115463, Skype amcrasto64 View Anthony Melvin Crasto Ph.D's profile on LinkedIn Anthony Melvin Crasto Dr.

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 →

Recent Posts

Hetrombopag Olamine


Hetrombopag Olamine, RAFUTROMBOPAG OLAMINE

  • Hetrombopag diolamine
  • SHR8735 olamine
  • Hetrombopag ethanolamine
  • SHR-8735 olamine

  • 580.6 g/mol, C29H36N6O7, V45T2I862X

2-aminoethanol;5-[2-hydroxy-3-[[5-methyl-3-oxo-2-(5,6,7,8-tetrahydronaphthalen-2-yl)-1H-pyrazol-4-yl]diazenyl]phenyl]furan-2-carboxylic acid

(Z)-5-(2-Hydroxy-3-(2- (3-methyl-5-oxo-1-(5,6,7,8-tetrahydronaphthalen- 2-yl)-1H-pyrazol-4(5H)-ylidene)hydrazinyl)phenyl) furan-2-carboxylic acid diethanolamine

CAS 1257792-42-9

1257792-41-8 (free acid) 1257792-41-8 (ethanolamine) 1257792-42-9 (olamine)

Jiangsu Hengrui Pharmaceutical, was approved in China in June 2021 for treatment of adult patients with chronic primary immune thrombocytopenia (ITP) and severe aplastic anemia who have not responded well to other treatments

Hetrombopag Olamine is the orally active ethanolamine salt of hetrombopag, a small-molecule, nonpeptide thrombopoietin receptor (TPO-R or CD110) agonist, with megakaryopoiesis-stimulating activity. Upon oral administration, hetrombopag targets, binds to and stimulates the transmembrane domain of the platelet TPO-R, a member of the hematopoietin receptor superfamily. Activation of TPO-R leads to the proliferation and differentiation of cells in the megakaryocytic lineage and an increase in platelet production. This may prevent or treat chemotherapy-induced thrombocytopenia.

  • OriginatorJiangsu Hengrui Medicine Co.
  • DeveloperAtridia; Jiangsu Hengrui Medicine Co.
  • ClassAntianaemics; Antihaemorrhagics; Aza compounds; Carboxylic acids; Furans; Pyrazolones; Small molecules; Tetrahydronaphthalenes
  • Mechanism of ActionThrombopoietin receptor agonists
  • Orphan Drug StatusYes – Thrombocytopenia
  • MarketedAplastic anaemia; Idiopathic thrombocytopenic purpura
  • Phase IIIThrombocytopenia
  • No development reportedUnspecified
  • 07 Dec 2024Efficacy and adverse events data from a phase-III trial in Aplastic anaemia presented at the 66th American Society of Hematology Annual Meeting and Exposition (ASH-Hem-2024)
  • 31 Jul 2024Phase-III clinical trials in Thrombocytopenia in China (PO) (NCT06507436)
  • 25 Jul 2024Jiangsu Hengrui Medicine plans a phase III trial in Thrombocytopenia (PO) in July 2024 (NCT06507436)

SYN

CN 113929668

https://patentscope.wipo.int/search/en/detail.jsf?docId=CN349207982&_cid=P21-MDCUSL-44897-1

Example 1. Synthesis of 5-(2-carbonyl-2,3-dihydrobenzoxazol-7-yl)furan-2-carboxylic acid
         
        Add purified water to the batching barrel, add 4.0kg of compound a under stirring, then add 10L of hydrochloric acid, stir, pump the material into a 50L reactor, add 10L of purified water to the batching barrel and pump it into the reactor. Turn on stirring, start cooling, the temperature drops to -5~2°C, start adding sodium nitrite aqueous solution (6.4L purified water, 1840g sodium nitrite), keep the temperature in the reactor no higher than 5°C during the process; after adding, continue stirring for 10~20min; add 800g of urea, continue stirring for 10~20min, the obtained diazonium salt solution is ready for use, and the temperature in the whole process is kept no higher than 5°C.
        44kg of acetone was pumped into a 200L reactor, and 15.0kg of compound b and 463.5g of copper chloride dihydrate were added in sequence under stirring. The temperature was raised to 30-35°C, and the obtained diazonium salt solution was added. The temperature was maintained at 30-40°C during the period. After the addition was completed, the temperature was maintained at 30-40°C and the reaction was continued with stirring for 1-1.5h. 120.0L of purified water was added, the temperature was raised to 40-45°C, and stirring was continued for a period of time. Filter, wash the filter cake with purified water until the filtrate is neutral, filter again, and collect the filter cake. 80L of purified water was added to the reactor, stirring was started, and the filter cake was added. Sodium hydroxide aqueous solution was added to the reactor to adjust the pH, the pH value was maintained at 8-10 for a period of time, and the filtrate was pumped into the reactor, and the filter was pressed into the material barrel through the filter press. Then 10L of purified water was pumped into the reactor and filtered into the material barrel. The material in the material barrel was pumped into the reactor, and then ethyl acetate was pumped in, stirred, and allowed to stand for 30-40 minutes. The aqueous phase was separated and collected, and the aqueous phase was pumped into the reactor, and the pH was adjusted to 3-4 with hydrochloric acid solution, and the filter cake was washed with purified water until the filtrate was neutral, and then the filter cake was collected. The filter cake was dried to obtain compound c. The yield of this step was 3.59 kg, and the yield was 55%.
        Example 2: Synthesis of 5-(3-amino-2-hydroxyphenyl)furan-2-carboxylic acid
         
        Purified water was pumped into the 50L reactor, stirring was started, 3.53kg of sodium hydroxide was added, and compound c obtained in the previous step was added. Under nitrogen protection, the reaction mixture was heated to reflux in the reactor for reaction. After the reaction, the reaction solution was cooled, the temperature was lowered to 0-10°C, and hydrochloric acid solution was added to adjust the pH value to 5-6. The filter cake was filtered, and the filtrate was washed with purified water until neutral, and then filtered again to collect the filter cake. The filter cake was dried to obtain compound d. The yield in this step was 2.78kg, with a yield of 90%.
        Example 3. Synthesis of (Z)-5-(2-hydroxy-(2-(3-methyl-5-oxo-1-(5,6,7,8-tetrahydronaphthalen-2-yl)-1H-pyrazol-4(5H)-ylidene)hydrazino)phenyl)furan-2-carboxylic acid
         
        Purified water was added to the batching barrel, and compound d was added in sequence under stirring, and then 6.3L hydrochloric acid was added, and the materials were pumped into a 200L reactor. Purified water was added to the batching barrel again, and then pumped into the reactor. Stirring was started, and the temperature was lowered to -5 to 2°C. Sodium nitrite aqueous solution (sodium nitrite to compound d molar ratio is 1:1) was added, and the internal temperature was kept at no more than 5°C during the process. After the addition was completed, stirring was continued; urea was added, and stirring was continued to obtain a diazonium salt solution for use, and the internal temperature was kept at no more than 5°C during the whole process.
        Add 36L purified water and 4000g sodium hydroxide to the batching barrel, stir to dissolve, and set aside. Take 26kg of the above sodium hydroxide aqueous solution, add compound e (the molar ratio of compound e to compound d is 0.9:1), stir, and add the resulting solution to the diazonium salt solution, keeping the temperature not exceeding 8°C. Add the above-prepared sodium hydroxide aqueous solution dropwise, adjust the pH to 8-10, and keep the temperature at 5-10°C for 3-4h. Add hydrochloric acid solution dropwise to adjust the pH to 2-3, keep the temperature not exceeding 25°C, filter, wash the filter cake with purified water until the filtrate is neutral, filter again, and collect the filter cake. Pump 48.0kg of tetrahydrofuran aqueous solution (22.5kg tetrahydrofuran, 25.5L purified water) into the reactor, add the above-obtained filter cake, beat, filter, wash the filter cake with tetrahydrofuran aqueous solution, wash the filter cake with purified water, filter again, and collect the filter cake. Dry the filter cake.
        Ethyl acetate was pumped into the reactor, and the above-obtained materials were added to the reactor for slurrying, and the filter cake was washed with ethyl acetate, and the filter cake was washed until no obvious droplets flowed out of the mirror, and the filter cake was collected and dried to obtain the compound of formula (I-2). The yield in this step was 5.34 kg, and the yield was 97.5%.
        Example 4. Synthesis of (Z)-5-(2-hydroxy-(2-(3-methyl-5-oxo-1-(5,6,7,8-tetrahydronaphthalen-2-yl)-1H-pyrazol-4(5H)-ylidene)hydrazino)phenyl)furan-2-carboxylic acid
        The compound of formula (I-2) was prepared by using a method substantially the same as in Example 3 (except that the equivalent of compound e was adjusted from 0.9 in Example 3 to the current 0.95, other conditions remained unchanged).
        Comparative Example 1: Synthesis of (Z)-5-(2-hydroxy-(2-(3-methyl-5-oxo-1-(5,6,7,8-tetrahydronaphthalen-2-yl)-1H-pyrazole-4(5H)-ylidene)hydrazino)phenyl)furan-2-carboxylic acid
         
        The compound of formula (I-2) was prepared by using a method substantially the same as in Example 3 (except that the step of adding urea was changed to starch potassium iodide test paper to indicate the reaction endpoint, and other conditions remained unchanged).
        Test Example 1: Effect of urea on the preparation process of the compound of formula (I-2)
        HPLC conditions:
        Chromatographic column: Welch Ultimate XB-C18 (4.6*150mm, 3μm), column temperature: 40℃,
        Flow rate: 1.0ml/min
        Injection volume: 10 μl
        Detector: UV detector
        Detection wavelength: 251nm
        Mobile phase: 0.1% trifluoroacetic acid aqueous solution was used as mobile phase A, acetonitrile was used as mobile phase B, and elution was performed at a ratio of 50%/50% of mobile phase A/mobile phase B.

PATENT

EP 2441457

PATENT

WO 2010142137

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2010142137&_cid=P21-MDCUXF-51461-1

PATENT

WO 2018133818

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018133818&_cid=P21-MDCUYN-53075-1

Example 1. Preparation of 3-methyl-1-(5,6,7,8-tetrahydronaphthalen-2-yl)-1H-pyrazol-5-ol hydrochloride 

[0107]

[0108](5,6,7,8-tetrahydronaphthalene-2-yl)hydrazine hydrochloride (1.3 kg, prepared according to the method in patent application WO2009092276A1) and ethyl acetoacetate (1.17 L) were added to ethyl acetate (5.2 L). The mixture was heated under reflux for 2 hours. The reaction solution was cooled to room temperature, then cooled to 0-5°C, stirred for 1 hour, filtered, and the solid was washed with a small amount of ethyl acetate to obtain a white solid product (1.4 kg, yield 81%). 

[0109]

MS m/z(ESI):229.26[M-HCl+H]

[0110]Example 2. Preparation of (Z)-5-(2-hydroxy-3-(2-(3-methyl-5-oxo-1-(5,6,7,8-tetrahydronaphthalen-2-yl)-1,5-dihydro-4H-pyrazol-4-ylidene)hydrazino)phenyl)furan-2-carboxylic acid (V-1) 

[0111]

[0112]Step 1: Synthesis of intermediate (II-1) 

[0113]Purified water (14.80 kg), 7-aminobenzo[d]oxazol-2(3H)-one (2.00 kg, prepared according to the method in patent application WO2005016898A2), and hydrochloric acid (5.33 kg) were added to the reaction kettle, the temperature was raised to 40-45°C, stirred for 10 min, cooled to -3-5°C, and sodium nitrite aqueous solution (sodium nitrite 940 g, water 3.20 kg) was added dropwise, the internal temperature was kept at no more than 5°C, the end point was controlled by starch potassium iodide test paper, and stirring was continued for 15 min; 

[0114]Add acetone (28L) to the reactor, then add furoic acid (4.57kg) and cupric chloride dihydrate (232g), stir at 35-40℃ until dissolved, add diazonium salt solution dropwise, keep the internal temperature at 35-40℃, and continue stirring for 1.5h. Add purified water (60L), heat to 35-40℃ and stir for 30min. Filter, wash the filter cake with 45-50℃ purified water. Add the filter cake to purified water (40kg), adjust the pH to 8-9 with 15% sodium hydroxide aqueous solution, filter, adjust the pH of the filtrate to 3-4 with 6mol/L hydrochloric acid, filter, wash the filter cake with purified water, and dry to obtain a solid (1.63kg, yield 50%). 

[0115]Step 2: Synthesis of intermediate (III-1) 

[0116]The product from the previous step (1.4 kg) and 15% aqueous sodium hydroxide solution (9.7 kg) were heated to reflux under argon protection and reacted for 28 hours. The reaction solution was poured into ice water (5-6 kg), and hydrochloric acid (6N, 3 L) was slowly added to adjust the pH value to 5-6. The temperature was maintained below 20°C. During this period, ethyl acetate was added to eliminate bubbles. The mixture was filtered, washed with purified water, and dried to obtain a solid (1.18 kg, yield 94%). 

[0117]Step 3: Synthesis of intermediate (V-1) 

[0118]Add the product of the previous step (1.10kg), purified water (27.5kg), and hydrochloric acid (2.92kg) to the reactor in sequence, stir and dissolve, cool to -4 to -1°C, add sodium nitrite aqueous solution (346g sodium nitrite, 5.5kg water), and continue to react for 15min after the addition is completed. Cool to -8 to -5°C. Dissolve sodium hydroxide (1.48kg) in purified water (13.2kg) to obtain a 10% sodium hydroxide aqueous solution. Add 5-methyl-2-(5,6,7,8-tetrahydronaphthalen-2-yl)-2H-pyrazole-3-ol hydrochloride (1.26kg) to the above sodium hydroxide aqueous solution (10kg) to dissolve, and add the resulting solution to the diazonium salt solution at once, keeping the temperature not higher than 10°C. Add the remaining 10% sodium hydroxide aqueous solution, adjust the pH to 8 to 9, naturally heat to 8 to 12°C for reaction, and react for 4h. Add 6N hydrochloric acid, adjust pH=2-3, keep the temperature not more than 20°C, filter, and wash the filter cake with water until pH=6-7. Add the filter cake to 50% tetrahydrofuran aqueous solution (19kg), slurry at room temperature for 2h, filter, wash with 50% tetrahydrofuran aqueous solution, wash with water, and dry. Add ethyl acetate (20kg) to the solid, slurry at 40-45°C for 2h under argon protection, cool to room temperature, filter, wash with ethyl acetate, add the solid to ethyl acetate (20kg), slurry at 40-45°C for 2h under argon protection, cool to room temperature, filter, wash with ethyl acetate, and dry to obtain a solid (2.18kg, yield 95%, purity 99.5%). 

[0119]MS m/z(ESI):457.1[M-1] 

[0120]Example 3. Preparation of (Z)-5-(2-hydroxy-3-(2-(3-methyl-5-oxo-1-(5,6,7,8-tetrahydronaphthalen-2-yl)-1,5-dihydro-4H-pyrazol-4-ylidene)hydrazino)phenyl)furan-2-carboxylic acid ethanolamine salt (1:2) 

[0121]

[0122]Preparation of crude product 

[0123]The compound of formula (V-1) (1.8 kg) was suspended in a tetrahydrofuran/ethanol (14.5 kg, V/V = 2:1) mixed solvent at room temperature, stirred for 0.5 h, cooled to 10-15 ° C, and a tetrahydrofuran ethanol solution of ethanolamine (479.6 g) (tetrahydrofuran 91 g and ethanol 41 g) was added dropwise. The mixture was naturally heated to room temperature and reacted for 20 h. Filtered, washed with a tetrahydrofuran/ethanol (V/V = 2:1) mixed solvent, washed with ethyl acetate, filtered, and dried to obtain a dark red solid (1.73 kg, yield 76%, purity 99.7%). 

[0124]

1H-NMR(500MHz,D 2O+NaOH)δ7.725-7.741(d,1H),7.298-7.316(d,3H),7.183-7.198(d,1H),7.131-7.149(m,2H),6.612-6.643(t,1H),3.574-3.596(t,4H),2.759-2.778(br,4H),2.698-2.721(t,4H),2.428(s,3H),1.772(br,4H).

SYN
J.Med.Chem.2024,67,4376−4418

HetrombopagOlamine (Hengqu).

Hetrombopag olamine (6), an oral nonpeptide thrombopoietin receptor
(TpoR)agonistdevelopedby JiangsuHengruiPharmaceutical, was approved in China in June2021 for treatment of adult patients with chronic primary immune thrombocytopenia (ITP) and severe aplastic anemiawhohave not responded well to other treatments.46Hetrombopag, like other TpoR agonists, increases platelet production by binding to the transmembranedomainofTpoRinprogenitorcells, inducing
megakaryocytes.Theeffectisadditivewiththeactionofnative thrombopoietin, whichbinds to the extracellular domainof TpoR.Hetrombopag is structurallyrelatedtoeltrombopag, a previously approvedTpoR, withmodifications to enhance potencyandminimizetoxicity.46−48InaPhaseIIIclinicaltrial, ITPpatients demonstratedadurableplatelet response, with reducedbleedingriskanduseof rescuetherapycomparedto
placebo.49 Akilo-scale, chromatography-freesynthesisofhetrombopag has been reported by researchers at Jiangsu Hengrui Pharmaceutical in the Chinese-language patent literature (Scheme 12).50,51 Commercially available aniline 6.1 was coupledwith furoic acid (6.2) using aMeerwein arylation reaction togive intermediate6.3.This process first involves diazotizationof the anilineusing sodiumnitrite andhydrochloricacid.Ureawasusedtoquenchtheresidualnitrousacid, animprovement thatultimatelygavetheproductwithhigher purity and lower levels of specific impurities; the crude
diazoniumsalt solutionwas carried forwarddirectlywithout furthermanipulation.Furoicacid(6.2)inacetonewastreated withcopper(II)chloridedihydratefollowedbyadditionofthe
diazonium salt solution to affect the arylation. The crude productwaspurifiedbyacid−baseextractionandisolatedby filtrationtoprovide6.3 in55%yield.Basichydrolysisof the
cycliccarbamateunveiledthefreeanilineandphenolmoieties in arene 6.4. Nucleophilic attack of the enolate anion of pyrazolone 6.5 (see Scheme 13) on the diazoniumsalt of aniline6.4 formed the central hydrazonemoiety ina JappKlingemann-like reaction. The crude product was triturated withethylacetatetorapidlyprovidehetrombopagfreebasein
97.5%yield.TreatmentwithethanolamineinTHFandEtOH thengeneratedhetrombopagolamine (6) in76%yieldand 99.7%purity.51 Pyrazolone intermediate6.5was synthesized in two steps
(Scheme 13).52,53 5,6,7,8-Tetrahydronaphthalen-2-yl amine (6.6)was converted to the diazoniumion and reduced in situ to the corresponding hydrazine 6.7 using stannous chloridedihydrate.Condensationof thehydrazinewithethyl acetoacetate in ethyl acetate and in situ cyclization gave pyrazolone6.5.While the synthesis fromaniline6.1 to the activepharmaceutical ingredient(API)6wasreportedonthe
kilo-scale, thesynthesisofpyrazolone6.5wasreportedonlyon gram-scale

(46) Syed, Y. Y. Hetrombopag: First approval. Drugs 2021, 81, 1581−1585.
(47) Xie, C.; Zhao, H.; Bao, X.; Fu, H.; Lou, L. Pharmacological characterization of hetrombopag, a novel orally active human thrombopoietin receptor agonist. J. Cell. Mol. Med. 2018, 22, 5367−5377.
(48) Zheng, L.; Liang, M.-z.; Zeng, X.-l.; Li, C.-z.; Zhang, Y.-f.; Chen, X.-y.; Zhu, X.; Xiang, A.-b. Safety, pharmacokinetics and pharmacodynamics of hetrombopag olamine, a novel TPO-R agonist, in healthy individuals. Basic Clin. Pharmacol. Toxicol. 2017, 121, 414−422.
(49) Mei, H.; Liu, X.; Li, Y.; Zhou, H.; Feng, Y.; Gao, G.; Cheng, P.; Huang, R.; Yang, L.; Hu, J.; Hou, M.; Yao, Y.; Liu, L.; Wang, Y.; Wu, D.; Zhang, L.; Zheng, C.; Shen, X.; Hu, Q.; Liu, J.; Jin, J.; Luo, J.; Zeng, Y.; Gao, S.; Zhang, X.; Zhou, X.; Shi, Q.; Xia, R.; Xie, X.; Jiang, Z.; Gao, L.; Bai, Y.; Li, Y.; Xiong, J.; Li, R.; Zou, J.; Niu, T.; Yang, R.;
Hu, Y. A multicenter, randomized phase III trial of hetrombopag: a novel thrombopoietin receptor agonist for the treatment of immune thrombocytopenia. J. Hematol. Oncol. 2021, 14, 37.
(50) Shi, A.; Diao, A.; Du, Y. Preparation of bicyclic substituted pyrazolone azo derivatives. China Patent CN 113929668, 2022.
(51) Diao, A.; Gao, X.; Bian, L. Method for preparing bicyclo substituted pyrazolone azo derivatives and intermediates. WO 2018133818, 2018.
(52) Tang, P. C.; Lue, H.; Fei, H.; Chen, Y. Preparation of pyrazole derivatives as thrombopoietin receptor agonists. WO 2010142137, 2010.
(53) Tang, P. C.; Lue, H.; Fei, H.; Chen, Y. Salts of bicyclo substituted pyrazolon azo derivatives, preparation method and use
thereof. European Patent EP 2441457, 2014.

.

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

……

//////////Hetrombopag Olamine, CHINA 2021, APPROVALS 2021, Hetrombopag diolamine, SHR 8735 olamine, Hetrombopag ethanolamine, SHR-8735 olamine, V45T2I862X, RAFUTROMBOPAG OLAMINE

SOVESUDIL


SOVESUDIL

PHP-201; AMA 0076, C23O3R93BM

CAS 1333400-14-8

Molecular Weight407.44
FormulaC23H22FN3O3

Sovesudil (PHP-201) is a potent, ATP-competitive, locally acting Rho kinase (ROCK) inhibitor with IC50s of 3.7 and 2.3 nM for ROCK-I and ROCK-II, respectively. Sovesudil lowers intraocular pressure (IOP) without inducing hyperemia.

SCHEME

PATENTS

Bioorganic & Medicinal Chemistry Letters (2013), 23(23), 6442-6446

https://www.sciencedirect.com/science/article/abs/pii/S0960894X13011141

10.1016/j.bmcl.2013.09.040

Figure 2. Synthetic scheme for synthesis of compounds 10–35. (a) H2SO4, MeOH, 60 C, 16 h; (b) NBS, AIBN, CCl4, reflux, 16 h; (c) Boc2NH, t-BuOK, DMF, rt, 16 h; (d) DCM/TFA
(50:1), 0 C ? rt, 4 h; (e) NaOH, MeOH, 50 C, 2 h; (f) HATU, DMAP, NEt3, DMA, 30 C, 16 h; (g) Pd(dppf)Cl2, Na2CO3, H2O, DMF, 100 C; 16 h; (h) ROH, TBTU, HOBT, DIEA, DMF,
rt, 16 h or ROH, DCC, DMAP, DCM, rt, 16 h or Me2C@C(Cl)NMe2, THF or DCM, rt, followed by ROH, 16 h; (i) DCM/TFA (7:1), 30 C, 16 h or HCl(g) in DCM, 30 C, 16 h.

PATENT

WO2011107608

////////SOVESUDIL, 1333400-14-8, PHP 201, AMA 0076, C23O3R93BM

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

……

Contezolid


Contezolid

WeightAverage: 408.337
Monoisotopic: 408.104539468

Chemical FormulaC18H15F3N4O4

Shanghai MicuRx Pharmaceutical Co. Ltd

Contezolid was approved for use by the National Medical Products Administration (NMPA) of China in 2021

  • OriginatorMicuRx Pharmaceuticals
  • ClassAntibacterials; Oxazolidinones; Skin disorder therapies
  • Mechanism of ActionProtein synthesis inhibitors
  • Phase IIIDiabetic foot; Skin and soft tissue infections
  • No development reportedGram-positive infections
  • 28 Jan 2025No recent reports of development identified for phase-I development in Gram-positive-infections(In volunteers) in China (IV)
  • 28 Jan 2025No recent reports of development identified for phase-I development in Gram-positive-infections(In volunteers) in China (PO)
  • 29 Nov 2024Phase-III clinical trials in Skin and soft tissue infections in China (IV), prior to November 2024

Contezolid (trade name Youxitai) is an antibiotic of the oxazolidinone class.[1][2] It is effective against Staphylococcus aureusmethicillin-resistant Staphylococcus aureus (MRSA), Streptococcus pyogenesStreptococcus agalactiae, and other bacteria.[3]

In 2021, it was approved by the National Medical Products Administration of China for the treatment of complicated skin and soft tissue infections (cSSTI).[3][4]

prodrug of contezolid, contezolid acefosamil, which is formulated for IV administration[5] is in Phase III clinical trials for diabetic foot infection.[6]

Chemical structure of contezolid acefosamil

SYN

https://pubs.acs.org/doi/abs/10.1021/acsmedchemlett.2c00191

Abstract

Abstract Image

New oral antibiotic contezolid (CZD) is effective against Gram-positive infections but unsuitable for intravenous (IV) administration due to its modest solubility. To address the medical need for an IV form of CZD, its isoxazol-3-yl phosphoramidate derivatives have been explored, and contezolid acefosamil (CZA, 8), the first representative of a novel O-acyl phosphoramidate prodrug class, has been identified. CZA exhibits high aqueous solubility (>200 mg/mL) and good hydrolytic stability at media pH suitable for IV administration. CZA rapidly converts into the active drug CZD in vivo. In a pharmacokinetic (PK) rat model, the exposure of active drug CZD after IV administration of the prodrug CZA was similar to or higher than that from the IV administration of CZD. The prodrug CZA is bioequivalent to or better than CZD in several preclinical infection models. CZA is likewise active upon its oral administration. To date, CZA has been evaluated in Phase 1 and Phase 2 clinical trials in the USA. It is advancing into further clinical studies including step-down therapy with in-hospital intravenous CZA administration followed by outpatient oral CZD treatment.

SYN

Contezolid (Youxitai). Contezolid (4), also referred to as MRX-I, is an orally administered oxazolidinone
antibacterial agent developed by Shanghai MicuRx Pharmaceutical Co. Ltd. Contezolid was developed to overcome the myelosuppression and monoamine oxidase (MAO) inhibition limitations of the structurally similar linezolid. 32 Contezolid is used to treat complicated skin and soft tissue infections arising
from multidrug-resistant Gram-positive bacterial infections including methicillin-susceptible Staphylococcus aureus (MSSA), methicillin-resistant Staphylococcus aureus (MRSA), Streptococcus pyogenes, Streptococcus agalactiae, and vancomycin-resistant enterococci.3334 Contezolid was approved for use by the National Medical Products Administration (NMPA) of China in 2021.
As with most antibacterial oral therapies, high 35 dosage is required; the drug is given twice daily for 7−14 days.36,37
The synthesis of contezolid builds on prior research from other groups.
A sequence developed by Pharmaciawith a facile SN38began Ar reaction between polyfluorinated nitro
benzene 4.1 and piperidine-4-one 4.2 to furnish 4.3 in good yield (Scheme 9). Silyl enol ether formation afforded 4.4, which was subjected to Tsuji’s 39 method to give the α,βunsaturated ketone in excellent yield. Subsequent reduction of the nitro group gave aryl amine 4.5. Treatment of 4.5 with isobutyl chloroformate gave carbamate 4.6, which was treated with optically pure epoxide 4.7 to give xazolidinone 4.8. 38Mesylation of the free alcohol and displacement with N-Bocaminoisoxazole 4.9 afforded the Boc-protected contezolid 4.10. Simple acidic removal of the Boc group provided contezolid 4.

(32) Wang, W.; Voss, K. M.; Liu, J.; Gordeev, M. F. Nonclinical
evaluation of antibacterial oxazolidinones contezolid and contezolid
acefosamil with low serotonergic neurotoxicity. Chem. Res. Toxicol.
2021, 34, 1348−1354.
(33) Hoy, S. M. Contezolid: First approval. Drugs 2021, 81, 1587−
1591.
(34) MicuRx Pharmaceuticals. China NMPA approves MicuRx’s
contezolid for treatment of drug-resistant bacterial infection. http://www.
micurx.com/703.html (accessed 2023-06).
(35) MSD Pharmaceuticals. Usual dosages of commonly prescribed
antibiotics. https://www.msdmanuals.com/en-jp/professional/
multimedia/table/usual-dosages-of-commonly-prescribed-antibioticsa
(accessed 2023-06).
(36) Barbachyn, M. R.; Hutchinson, D. K.; Brickner, S. J.; Cynamon,
M. H.; Kilburn, J. O.; Klemens, S. P.; Glickman, S. E.; Grega, K. C.;
Hendges, S. K.; Toops, D. S.; et al. Identification of a novel
oxazolidinone (U-100480) with potent antimycobacterial activity. J.
Med. Chem. 1996, 39, 680−685.
(37) Im, W. B.; Choi, S. H.; Park, J. Y.; Choi, S. H.; Finn, J.; Yoon, S.
H. Discovery of torezolid as a novel 5-hydroxymethyl-oxazolidinone
antibacterial agent. Eur. J. Med. Chem. 2011, 46, 1027−1039.
(38) Manninen, P. R.; Brickner, S. J. Preparation of N-aryl-5R
hydroxymethyl-2-oxazolidinones from N-aryl carbamates: N-phenyl
(5R)-hydroxymethyl-2-oxazolidinone. Organic Synth 2005, 81, 112.
(39) Tsuji, J.; Minami, I.; Shimizu, I. A novel palladium-catalyzed
preparative method of α,β-unsaturated ketones and aldehydes from
saturated ketones and aldehydes via their silyl enol ethers. Tetrahedron
Lett. 1983, 24, 5635−5638.

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

……

Clinical data
Trade namesYouxitai
Other namesMRX-I
Legal status
Legal statusRx in China
Identifiers
IUPAC name
CAS Number1112968-42-9
PubChem CID25184541
IUPHAR/BPS10795
DrugBankDB12796
ChemSpider34217570
UNIIB669M62ELP
KEGGD11297
ChEMBLChEMBL3287379
CompTox Dashboard (EPA)DTXSID901353186 
Chemical and physical data
FormulaC18H15F3N4O4
Molar mass408.337 g·mol−1
3D model (JSmol)Interactive image
SMILES
InChI

References

  1.  Gordeev MF, Yuan ZY (June 2014). “New Potent Antibacterial Oxazolidinone (MRX-I) with an Improved Class Safety Profile”. Journal of Medicinal Chemistry57 (11): 4487–4497. doi:10.1021/jm401931ePMID 24694071.
  2.  Zhao X, Huang H, Yuan H, Yuan Z, Zhang Y (May 2022). “A Phase III multicentre, randomized, double-blind trial to evaluate the efficacy and safety of oral contezolid versus linezolid in adults with complicated skin and soft tissue infections”. The Journal of Antimicrobial Chemotherapy77 (6): 1762–1769. doi:10.1093/jac/dkac073PMID 35265985.
  3.  Hoy SM (September 2021). “Contezolid: First Approval”Drugs81 (13): 1587–1591. doi:10.1007/s40265-021-01576-0PMC 8536612PMID 34365606.
  4.  Mak E (3 June 2021). “Micurx wins China approval for antibacterial contezolid”BioWorld.
  5.  Liu J, Wang W, Wang C, Zhang L, Zhang X, Liu S, et al. (July 2022). “Discovery of Antibacterial Contezolid Acefosamil: Innovative O-Acyl Phosphoramidate Prodrug for IV and Oral Therapies”ACS Medicinal Chemistry Letters13 (7): 1030–1035. doi:10.1021/acsmedchemlett.2c00191PMC 9290071PMID 35859881.
  6.  “Contezolid acefosamil by MicuRx Pharmaceuticals for Diabetic Foot Infection (DFI): Likelihood of Approval”GlobalData. 31 May 2023 – via Pharmaceutical Technology.

/////////Contezolid, CHINA 2021, APPROVALS 2021, MRX-I, 1112968-42-9, MRX 1, B669M62ELP, コンテゾリド ,

AZVUDINE


AZVUDINE

CAS


1011529-10-4

WeightAverage: 286.223
Monoisotopic: 286.082581021

Chemical FormulaC9H11FN6O4

  • FNC
  • HY-19314
  • RO 0622
  • RO-0622
  • SB17040

  • IJ2XP0ID0K
  • DTXSID901027757

4-amino-1-[(2R,3S,4R,5R)-5-azido-3-fluoro-4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-1,2-dihydropyrimidin-2-one

Azvudine is an antiviral drug which acts as a reverse transcriptase inhibitor.[3] It was discovered for the treatment of hepatitis C[4] and has since been investigated for use against other viral diseases such as AIDS and COVID-19,[2][5] for which it was granted conditional approval in China.[6][7]

Azvudine was first discovered in 2007.[8] It costs 350 Chinese yuan per 7 days for COVID, as of November 2022.[9]

Azvudine is under investigation in clinical trial NCT04668235 (Study on Safety and Clinical Efficacy of AZVUDINE in COVID-19 Patients (Sars-cov-2 Infected)).

Azvudine (RO-0622) is a potent nucleoside reverse transcriptase inhibitor (NRTI), with antiviral activity on HIVHBV and HCV. Azvudine exerts highly potent inhibition on HIV-1 (EC50s ranging from 0.03 to 6.92 nM) and HIV-2 (EC50s ranging from 0.018 to 0.025 nM). Azvudine inhibits NRTI-resistant viral strains. Azvudine is a click chemistry reagent, it contains an Azide group and can undergo copper-catalyzed azide-alkyne cycloaddition reaction (CuAAc) with molecules containing Alkyne groups. It can also undergo strain-promoted alkyne-azide cycloaddition (SPAAC) reactions with molecules containing DBCO or BCN groups.

SYN

https://patents.google.com/patent/CN111892636A/en

According to the inventor’s research and understanding, at present, the synthesis of azvudine mainly includes the following methods according to different raw materials:

1. It is prepared by using a ribonucleotide as a raw material. The method requires a total of 15 steps of reaction to obtain the target product. The inventor’s research found that DAST is used as a fluorination reagent in the fluorination process of this method, which has large steric hindrance and is difficult to fluoride, and the route process is complicated and the route is long. Low cost, high cost, not suitable for industrial production.

2. It is prepared by using 1,3,5-O-tribenzoyl-2-deoxy-2-fluoro-D-arabinofuranoside as raw material. The inventors have found that the preparation raw materials are not easy to obtain, and the route involves uridine to In the conversion reaction of cytidine, the reaction route is further extended, thus limiting the further application of this method.

3. Using ribonucleotides as raw materials to synthesize, the inventors found that this method requires 12 steps to synthesize the target product, and DAST is also used as a fluorination reagent in the fluorination process, which has large steric hindrance and low fluorination efficiency.

4. It is prepared by using uracil nucleotide as raw material. The inventors have found that the raw material cost of this method is relatively high, it involves the conversion process of uridine to cytidine, and the reaction yield is not high.

To sum up, the inventors have found that the currently known processes for preparing azvudine have the following disadvantages: synthesizing uracil nucleotides with ribonucleotides as raw materials, and then converting uracil nucleotides into target products, synthesizing uracil nucleotides. The process routes all exceed 12 steps, and the fluorination reaction uses DAST (diethylaminosulfur trifluoride) reagent, which increases the difficulty of the reaction due to steric hindrance, reduces the yield, and brings difficulties to industrial production.

Example 1

Figure BDA0002623551980000063
Figure BDA0002623551980000063

Accurately weigh 4.86 g of cytidine (compound 1), dissolve it in 20 mL of ethanol, then add 4.52 g of benzoic anhydride, raise the temperature to 60° C., and stir overnight. After the reaction was completed, the solvent was removed under reduced pressure, and 100 mL of deionized water was added to wash and filter to obtain compound 2 (96.5% yield). The structural characterization is shown in Figure 1 .

Example 2

Figure BDA0002623551980000071
Figure BDA0002623551980000071

Weigh 3.5 g of compound 2, dissolve in 25 mL of pyridine, ice-water bath at 0°C, add 3.0 mL of TIPDS under nitrogen protection, react for 1 h, decompose the reaction mixture with water, remove pyridine under reduced pressure, extract with chloroform, and wash with saturated aqueous sodium bicarbonate solution , and dried over anhydrous sodium sulfate to obtain compound 3 (83.6% yield). The structural characterization is shown in Figure 2.

Example 3

Figure BDA0002623551980000072
Figure BDA0002623551980000072

Weigh 5.9 g of compound 3, dissolve it in 100 mL of tetrahydrofuran, add 2.82 g of trifluoromethanesulfonic anhydride (Tf 2 O), and react at room temperature for 2 h under nitrogen protection. Then, the reaction temperature was lowered to -20°C, 2.46 g of tetrabutylammonium fluoride (Bu 4 NF) was added, and the reaction was continued for 10 h. After the reaction was completed, tetrahydrofuran was removed under reduced pressure, extracted with chloroform, washed with saturated aqueous sodium bicarbonate solution, and dried over anhydrous sodium sulfate to obtain compound 4 (86.8% yield). The structural characterization is shown in Figure 3.

Example 4

Figure BDA0002623551980000073
Figure BDA0002623551980000073

5.9 g of compound 3 was weighed, dissolved in 100 mL of dichloromethane and 10 mL of anhydrous pyridine, cooled to -50°C under nitrogen protection, added with 1.61 g of DAST, and reacted for 12 h. After the reaction was completed, the solvent was removed under reduced pressure, extracted with chloroform, washed with saturated aqueous sodium bicarbonate solution, and dried over anhydrous sodium sulfate to obtain compound 4 (76.0% yield).

Example 5

Figure BDA0002623551980000074
Figure BDA0002623551980000074

Weigh 3.5g of compound 4, add 100mL of tetrahydrofuran, add 0.5g of imidazole, 0.5g of triphenylphosphine, slowly add 3.75g of tetrahydrofuran solution containing 10wt% iodine, stir at room temperature for 5h, the reaction is complete, remove the solvent under reduced pressure, Compound 5 was prepared (84% yield) and the structural characterization is shown in Figure 4 .

Example 6

Figure BDA0002623551980000081
Figure BDA0002623551980000081

Weigh 4.59g of compound 5, dissolve it in 100mL of methanol, add 0.5g of DBU, control the temperature to 60°C, react for 12h, cool to room temperature, add saturated aqueous sodium chloride solution, adjust the acidic pH=3 with 1M hydrochloric acid, extract with ethyl acetate, It was dried over anhydrous sodium sulfate and concentrated under reduced pressure to obtain compound 6 (77.4% yield). The structural characterization is shown in Figure 5 .

Example 7

Figure BDA0002623551980000082
Figure BDA0002623551980000082

Weigh 3.3 g of compound 6, add 50 mL of DMF solution dissolved with 0.6 g of sodium azide, add 50 mL of DMF solution dissolved with 0.6 g of ICl, control the temperature to 0 ° C, and react for 12 h. After the reaction is completed, add sodium bisulfite until The color of iodine disappears completely. The solvent was removed under reduced pressure to obtain compound 7 (77% yield). The structural characterization is shown in FIG. 6 .

Example 8

Figure BDA0002623551980000083
Figure BDA0002623551980000083

Weigh 2.5 g of compound 7, dissolve it in 50 mL of DMF, add 0.65 g of benzoic acid, add 0.5 g of silver acetate, and stir at room temperature for 12 h. After the reaction was completed, the mixture was filtered, and the solvent was removed under reduced pressure to obtain compound 8 (71.2% yield). The structural characterization is shown in FIG. 7 .

Example 9

Figure BDA0002623551980000084
Figure BDA0002623551980000084

Weigh 4.82 g of compound 8, add 100 mL of methanol, 10 mL of deionized water, 3 mL of triethylamine, stir at room temperature for 5 h, and remove the solvent under reduced pressure to obtain compound 9 (88.7% yield). The structural characterization is shown in Figure 8.

SYN

https://pubs.acs.org/doi/10.1021/acs.oprd.4c00166

Azvudine was approved for the treatment of adult HIV-1 infection in China in 2021, and it was approved for conditional marketing for the treatment of SARS-CoV-2 in China in 2022. In this work, we describe a fully continuous flow synthesis of 2′-deoxy-2′-fluoroarabinoside, a key intermediate for azvudine. The process was accomplished via six chemical transformations, including chlorination, hydrolysis, fluorination, bromination, condensation, and deprotection in six sequential continuous flow devices. Under the optimized process conditions, the total yield was 32.3% with a total residence time of 156 min. Compared with batch conditions, the total yield was doubled, the total reaction time was shortened 16 times, and the E factor was reduced 1.63 times.

1,3,5-Tri-O-benzoyl-D-ribofuranose (FR-1)
To a stirred solution of O-acetyl-2,3,5-tri-O-benzoyl-β-D-ribofuranose (FR-0,
1.008 g, 2.0 mmol) in anhydrous dichloromethane (DCM, 10 mL) was added
dropwise thionyl chloride (0.713 g,6 mmol) at 0 oC, and the resulting mixture
allowed to stir at room temperature for 14 h. The solution was evaporated,
dissolved with toluene (5 mL× 3), and then evaporated. To the residue was
added DCM (10 mL) and water (5 mL), and and continued stirring at room
temperature for 2 h. The solution was then washed with saturated aqueous
NaHCO3 (15 mL) and dried over Na2SO4, filtered and evaporated. DCM (2 mL)
and n-Hexane (20 mL) was added to the residue and the mixture was stirred at
room. A white solid was collected by filtration to give FR-1 (yield: 47.8%).
2-Deoxy-2-fluoro-1,3,5-tri-O-benzoyl-D-ribofuranose (FR-F)
To a stirred solution of FR-1 (0.924 g, 2.0 mmol) in anhydrous DCM (10 mL) was
added dropwise DAST (0.976 g, 6.0 mmol) for 30 min, and the resulting mixture
allowed to stir at 40 °C (16 h). The reaction was cooled and quenched with
saturated aqueous NaHCO3 (15 mL). The solution was extracted with DCM and
water, and then washed with saturated aqueous NaHCO3 (20 mL). This was then
dried over Na2SO4, filtered and evaporated. The crude product was purified by
silica gel column chromatography (ethyl acetate: petroleum ether =1:10) to
obtain a white solid (yield: 41.2%).
α-D-Arabinofuranosyl bromide, 2-deoxy-2-fluoro-, 3,5-dibenzoate (FR-Br)
To a stirred solution of FR-F (0.928 g, 2.0 mmol) in anhydrous DCM (10 mL) was
added dropwise 33.3% HBr-HOAc (1.47 g, 6.0 mmol) at 0 oC, and the resulting
mixture were allowed to stir at room temperature for 7 h. The reaction was
quenched with saturated aqueous NaHCO3 (10 mL), dried over Na2SO4, filtered
and evaporated to yield the product (FR-Br) as a brown oil (yield: 99.8%).

1-(2-deoxy-2-fluoro-3,5-di-O-benzoyl-β-D-arabino-furanosyl)uracil (FAU-Bz)
A mixture of uracil (0.336 g, 3 mmol) and (NH4)2SO4 (10 mg, 0.075 mmol) in
hexamethyldisilazane (HMDS) (6 mL) was refluxed under nitrogen for 5 h. To
the silylated uracil solution was added a solution of FR-Br in dry acetonitrile (10
mL) and the mixture was refluxed under nitrogen for 5 h. The solution was
evaporated, extracted with DCM (20 mL) and washed with saturated aqueous
NaHCO3 (15 mL). This was then dried over Na2SO4, filtered and evaporated.
Ethyl acetate (10 mL) and petroleum ether (50 mL) was added to the residue
and the mixture was stirred at room temperature. A light yellow solid was
collected by filtration to give FAU-Bz (yield: 83.2%).
2′-deoxy-2′-fluoro- arabinoside (FAU)
To a solution of FUA-Bz (0.908 g, 2.0 mmol) in anhydrous methanol (MeOH, 10
mL) was added NH3-MeOH (5 mL), stirred at room temperature for 15 h and
evaporated to dryness under reduced pressure. White solid

SYN

: J. Med. Chem. 2024, 67, 4376−4418

Azvudine (1). Azvudine (1) is an antiviral manufactured by China-based Genuine Biotech. It was
approved in China in 2021 as a first-in-class treatment for human immunodeficiency virus (HIV). It has a dual function, acting as a reverse transcriptase inhibitor and targeting the viral infectivity factor/apolipoprotein B mRNA-editing enzyme, catalytic subunit 3G (Vif/A3G) protein−protein interaction.6 Azvudine has activity against both wild-type and drug-resistant strains of HIV due to the presence of a 3′-hydroxy group and substitution in the 4′-position of the ribose core.7 Due to its known antiviral activity, azvudine was repurposed as a treatment for COVID-19 and approved for this indication inChina in 2022. It acts as an RNA-dependent RNA polymerase (RdRp) inhibitor, the same mechanism as the previously approved molnupiravir and remdesivir. In addition to its antiviral activity, concentration of the drug in the thymus has suggested immune-targeting activity; this dual function is unique among RdRp inhibitors.8 Several syntheses of azvudine have been reported in the scientific and patent literature. Scheme 1 highlights a 100 g scale synthesis from a patent filed by Shandong University.9 Other syntheses are similar, containing the furanose functional group manipulations in 1.5−1.8, though these routes differ in choice of nucleobase and protecting group strategy, and were reported on a smaller scale.10−12 The synthesis began from benzoyl-protected fluoro-furanose 1.1. Bromination with HBr in acetic acid followed by displacement of the bromide 1.2 with protected cytosine 1.3 yielded intermediate 1.4. Deprotection of the benzoyl groups with ammonia in MeOH formed diol 1.5, and a Mitsunobu reaction converted the
primary alcohol to alkyl iodide 1.6. Elimination of the iodide with sodium methoxide followed by addition of sodium azide and iodine monochloride across the resulting alkene produced substitution in the 4′-position of the ribose core.7 Due to its known antiviral activity, azvudine was repurposed as a treatment for COVID-19 and approved for this indication in China in 2022. It acts as an RNA-dependent RNA polymerase (RdRp) inhibitor, the same mechanism as the previously approved molnupiravir and remdesivir. In addition to its antiviral activity, concentration of the drug in the thymus has suggested immune-targeting activity; this dual function is unique among RdRp inhibitors.8 Several syntheses of azvudine have been reported in the scientific and patent literature. Scheme 1 highlights a 100 g scale synthesis from a patent filed by Shandong University.9 Other syntheses are similar, containing the furanose functional group manipulations in 1.5−1.8, though these routes differ in choice of nucleobase and protecting group strategy, and were reported on a smaller scale.10−12 The synthesis began from
benzoyl-protected fluoro-furanose 1.1. Bromination with HBr in acetic acid followed by displacement of the bromide 1.2 with protected cytosine 1.3 yielded intermediate 1.4. Deprotection of the benzoyl groups with ammonia in MeOH formed diol 1.5, and a Mitsunobu reaction converted the primary alcohol to alkyl iodide 1.6. Elimination of the iodide with sodium methoxide followed by addition of sodium azide
and iodine monochloride across the resulting alkene producedazide 1.7. Both the alcohol and amine were reprotected with benzoyl chloride, and the iodide was displaced with metachlorobenzoic acid in an oxidative nucleophilic substitution reaction to yield penultimate intermediate 1.9. All protecting
groups were then removed with ammonia in MeOH to yield
azvudine (1).

(6) Sun, L.; Peng, Y.; Yu, W.; Zhang, Y.; Liang, L.; Song, C.; Hou, J.;
Qiao, Y.; Wang, Q.; Chen, J.; et al. Mechanistic insight into
antiretroviral potency of 2’-deoxy-2’-beta-fluoro-4’-azidocytidine
(FNC) with a long-lasting effect on HIV-1 prevention. J. Med.
Chem. 2020, 63, 8554−8566.
(7) Chang, J. 4’-Modified nucleosides for antiviral drug discovery:
achievements and perspectives. Acc. Chem. Res. 2022, 55, 565−578.
(8) Zhang, J.-L.; Li, Y.-H.; Wang, L.-L.; Liu, H.-Q.; Lu, S.-Y.; Liu, Y.;
Li, K.; Liu, B.; Li, S.-Y.; Shao, F.-M.; Wang, K.; Sheng, N.; Li, R.; Cui,
J.-J.; Sun, P.-C.; Ma, C.-X.; Zhu, B.; Wang, Z.; Wan, Y.-H.; Yu, S.-S.;
Che, Y.; Wang, C.-Y.; Wang, C.; Zhang, Q.; Zhao, L.-M.; Peng, X.-Z.;
Cheng, Z.; Chang, J.-B.; Jiang, J.-D. Azvudine is a thymus-homing
anti-SARS-CoV-2 drug effective in treating COVID-19 patients. Signal
Transduct. Target Ther. 2021, 6, 414.
(9) Chen, X.; Wang, Z.; Yu, H.; Liu, X. Preparation method of
azvudine and its intermediates. China Patent CN 115960147, 2023.
(10) Smith, D. B.; Kalayanov, G.; Sund, C.; Winqvist, A.; Maltseva,
T.; Leveque, V. J.-P.; Rajyaguru, S.; Pogam, S. L.; Najera, I.;
Benkestock, K.; Zhou, X.-X.; Kaiser, A. C.; Maag, H.; Cammack, N.;
Martin, J. A.; Swallow, S.; Johansson, N. G.; Klumpp, K.; Smith, M.
The design, synthesis, and antiviral activity of monofluoro and
difluoro analogues of 4’-azidocytidine against hepatitis C virus
replication: The discovery of 4’-azido-2’-deoxy-2’-fluorocytidine and4’-azido-2’-dideoxy-2’,2’-difluorocytidine. J. Med. Chem. 2009, 52,
2971−2978.
(11) Wang, Q.; Hu, W.; Wang, S.; Pan, Z.; Tao, L.; Guo, X.; Qian,
K.; Chen, C. H.; Lee, K. H.; Chang, J. Synthesis of new 2’-deoxy-2’-
fluoro-4’-azido nucleoside analogues as potent anti-HIV agents. Eur. J.
Med. Chem. 2011, 46, 4178−83.
(12) Deng, W.; Jiang, S.; Yu, T.; Zhai, D. Synthesis method of
azvudine. China Patent CN 111892636, 2020.

Medical uses

In July 2021, azvudine became conditionally approved in China for the following indication: “to treat high-viral-load cases of HIV-1, in combination with a nucleoside reverse-transcriptase inhibitor and a non-nucleoside reverse-transcriptase inhibitor”. The approval text describes it as a dual reverse transcriptase and Vif inhibitor.[10]

In July 2022, azvudine received emergency conditional approval for COVID-19 in adults.[11] It is believed to work by inhibiting the RNA-dependent RNA polymerase (RdRp) enzyme in the SARS-CoV-2 virus.[12][13]

Adverse effects

According to the manufacturer, phase II trials of azvudine in combination with doravirine and tenofovir disoproxil fumarate in HIV patients found an adverse effect profile similar to, but milder, than lamivudine combined with the two drugs. Very common (> 10%) side effects include dizziness, elevated liver enzymes, vomiting, and elevated alkaline phosphatase. Common (> 1%) side effects include nausea, elevated blood lipids, fever, insomnia, tiredness, and diarrhea. Detailed numbers are provided by Genuine in the slides and the medication package insert.[14][15] A boxed warning is present at the beginning of the Chinese package insert, describing a risk of “decrease in absolute neutrophil count, increase in total bilirubin, increase in glutathione aminotransferase, and increase in blood glucose”.[15]

The small (n=10) open-label pilot study for azvudine used alone in COVID reported no adverse events.[16]

Non-human models

Azvudine is found to be mutagenic in in vitro in the Ames test, CHL test, and in vitro in the mice micronucleus test.[17]

Azvudine is toxic to the reproductive system of rats and rabbit. The minimum reproductive NOAEL found for males is 5.0 mg/kg/d and for females 0.5 mg/kg/d. It is excreted in rat breast milk; the NOAEL for rat pups is 1.5 mg/kg/d.[17]

Azvudine is mainly toxic to the immune system, bone marrow, and digestive system of model animals. The chronic NOAELs are 0.5 mg/kg/d (rat, 3 months), 0.3 mg/kg/d (rat, 26 weeks), and 0.1 mg/kg/d (beagle dog, 1 month and 39 weeks).[17] For comparison, the chronic human dose for HIV treatment is 0.05 mg/kg/d, using the reference 3 mg dose and an average Chinese body mass of 59.5 kg (2014).

History

Azvudine was first found in literature in a patent filed by Chang Jun-biao of Zhengzhou University.[8] It received its current name in 2009, when researchers at Roche independently discovered it as a Hep C RNA polymerase inhibitor in vitro.[4] In the following years, Chinese scientists tested it in vitro for a number of targets, most importantly HBV (human and duck) and HTLV-1, two viruses with a reverse transcriptase.[18][19][20]

It was first proposed as an HIV treatment in 2011, when in vitro tests by the Chang group provided positive results.[21] In 2014, its oral pharmokinetics in rats was elucidated.[1] A phase II study (NCT04109183) was finished in March 2019 by Genuine Biotech. In August 2020, the Chang group found that the substance inhibits vif in vitro.[22] In the same month, China’s drug regulator (NMPA) decided to fast-track the approval process, labelling it a first-in-class medication.[14] In July 2021, NMPA granted conditional approval for HIV-1.[7] It was included in the 2021 HIV treatment recommendnation by the Chinese Medical Association and Chinese CDC, published October that year.[14] Curiously, no full results of the trial have been made available for this study in any journal detailing the experiment design as of December 2022.[23] Parts of the results are shown on the drug monograph as well as a 2022 slides deck produced by Genuine for the NHSA available on the latter’s website.[14]

Azvudine was found to inhibit some coronaviruses in vitro around 2020, leading to an interest in its use in COVID. An open-label pilot study on mild and moderate cases was performed in 2020, with mildly positive results.[16] A phase III trial was performed in 2022 in China. In July 2022, China’s drug regulator granted conditional approval for it to be used to treat COVID-19, following a local phase III trial.[6] Initially, no detailed description of the said trial was published in any journals, but state media quoted some numbers from the developer: “40% clinical improvement in 7 days by FNC group, compared to 11% in control”.[7] It is unclear how such “improvement” is defined.

Four phase III clinical trials investigated azvudine’s efficacy and safety in adults with mild-to-moderate COVID-19. The findings indicate that azvudine may reduce the time to eliminate detectable levels of virus (viral load) and improve symptoms faster than standard treatment. In trials, it was reported to be safe with few side effects. However, some studies produced inconsistent results in terms of symptom improvement and severe illness prevention. Additionally, the studies tended to use a smaller number of participants than other major COVID-19 drug trials.[12][13]

Society and culture

Genuine owns two different tradenames for this medication: 双新艾克 (literally “dual new AIDS inhibitor”) for HIV use[14] and 捷倍安 (literally “fast extra safe”) for COVID use.[7] No generics are available.

Geniune holds one patent related to the drug: the original 2007 patent on the entire class of 2′-fluorine-4′-substituted nucleotides, purchased from Zhengzhou University.[8] Two other Chinese patents on synthesizing the drug are found on Google Patents, but the owners do not appear to be connected to Geniune.[24] Roche held one 2002 patent, CNA028118480A (CN1516590A), over the broader class of 4′-substituted nucleotides. The patent was voided in 2019 after Riboscience, its new holder, stopped paying fees.[25]

References

  1.  Peng Y, Cheng T, Dong L, Zhang Y, Chen X, Jiang J, et al. (September 2014). “Quantification of 2′-deoxy-2′-β-fluoro-4′-azidocytidine in rat and dog plasma using liquid chromatography-quadrupole time-of-flight and liquid chromatography-triple quadrupole mass spectrometry: Application to bioavailability and pharmacokinetic studies”. Journal of Pharmaceutical and Biomedical Analysis98: 379–386. doi:10.1016/j.jpba.2014.06.019PMID 24999865.
  2.  Liu Y, Liu B, Zhang Y, Peng Y, Huang C, Wang N, et al. (July 2017). “Intestinal absorption mechanisms of 2′-deoxy-2′-β-fluoro-4′-azidocytidine, a cytidine analog for AIDS treatment, and its interaction with P-glycoprotein, multidrug resistance-associated protein 2 and breast cancer resistance protein”. European Journal of Pharmaceutical Sciences105: 150–158. doi:10.1016/j.ejps.2017.05.009PMID 28487144S2CID 4252337.
  3.  Wang RR, Yang QH, Luo RH, Peng YM, Dai SX, Zhang XJ, et al. (2014). “Azvudine, a novel nucleoside reverse transcriptase inhibitor showed good drug combination features and better inhibition on drug-resistant strains than lamivudine in vitro”PLOS ONE9 (8): e105617. Bibcode:2014PLoSO…9j5617Wdoi:10.1371/journal.pone.0105617PMC 4140803PMID 25144636.
  4.  Smith DB, Kalayanov G, Sund C, Winqvist A, Maltseva T, Leveque VJ, et al. (May 2009). “The design, synthesis, and antiviral activity of monofluoro and difluoro analogues of 4′-azidocytidine against hepatitis C virus replication: the discovery of 4′-azido-2′-deoxy-2′-fluorocytidine and 4′-azido-2′-dideoxy-2′,2′-difluorocytidine”. Journal of Medicinal Chemistry52 (9): 2971–2978. doi:10.1021/jm801595cPMID 19341305.
  5.  Harrison C (April 2020). “Coronavirus puts drug repurposing on the fast track”Nature Biotechnology38 (4): 379–381. doi:10.1038/d41587-020-00003-1PMID 32205870.
  6.  Ye Y (July 2022). “China approves first homegrown COVID antiviral”. Naturedoi:10.1038/d41586-022-02050-xPMID 35883009S2CID 251104078.
  7.  “首个国产抗新冠口服药附条件获批上市” [First domestic oral anti-Covid drug conditionally approved]. 新华网. 证券日报. 2022-07-26. Archived from the original on 2022-08-09. Retrieved 2022-07-26.
  8.  Chang J, Bao X, Wang Q, Guo X, Wang W, Qi X. Preparation of 2′-fluoro-4′-substituted nucleoside analogs as antiviral agents. 20070807. Chinese Patent Application No: CN 2007-10137548. Chinese Patent No: CN 101177442A, 20080514.
  9.  “新冠口服药阿兹夫定片线上开售, 每瓶售价350元” [Oral COVID drug Azvudine tablet available online at 350 yuan/bottle]. Xinmin Evening News. 19 November 2022. Retrieved 28 December 2022 – via Beijing Daily (repost).
  10.  “国家药监局附条件批准阿兹夫定片上市” [NMPA conditionally approvals azvudine tablets]. http://www.nmpa.gov.cn (in Chinese). 2021-07-21.
  11.  “国家药监局应急附条件批准河南真实生物科技有限公司阿兹夫定片增加新冠肺炎治疗适应症注册申请” [NMPA grants emergency conditional approval for additional indication registration of azvudine tablets (Hebei Genuine Biotech Co., Ltd.)]. http://www.nmpa.gov.cn. 2022-07-25.
  12.  Zhu KW (2023). “Efficacy and safety evaluation of Azvudine in the prospective treatment of COVID-19 based on four phase III clinical trials”Frontiers in Pharmacology14: 1228548. doi:10.3389/fphar.2023.1228548PMC 10484631PMID 37693894.
  13.  Wang Y, Xie H, Wang L, Fan J, Zhang Y, Pan S, Zhou W, Chen Q, Liu X, Wu A, Zhang H, Wang J, Tian X (February 2024). “Effectiveness of azvudine in reducing mortality of COVID-19 patients: a systematic review and meta-analysis”Virology Journal21 (1): 46. doi:10.1186/s12985-024-02316-yPMC 10893615PMID 38395970.
  14.  Genuine Biotech (July 11, 2022). “阿兹夫定片(双新艾克)” [Azvudine Tablets (Shuāngxīnàikè)]. NHSA.gov.cn. Archived from the original on 2022-09-06.
  15.  “阿兹夫定片说明书” [Azvudine Tablets, Monograph] (PDF). WUXU DATA. Retrieved 2023-01-03.
  16.  Ren Z, Luo H, Yu Z, Song J, Liang L, Wang L, et al. (October 2020). “A Randomized, Open-Label, Controlled Clinical Trial of Azvudine Tablets in the Treatment of Mild and Common COVID-19, a Pilot Study”Advanced Science7 (19): e2001435. doi:10.1002/advs.202001435PMC 7404576PMID 35403380.
  17.  Drug Review Center (China) (June 30, 2022). “阿兹夫定片 (CXHS2000016-17) 申请上市技术审评报告” [Azvudine tabs (CHXS2000016-17) request for marketing technical review report] (PDF). WUXU DATA. Retrieved 2023-01-03.
  18.  Wang Q, Liu X, Wang Q, Zhang Y, Jiang J, Guo X, et al. (April 2011). “FNC, a novel nucleoside analogue inhibits cell proliferation and tumor growth in a variety of human cancer cells”. Biochemical Pharmacology81 (7): 848–855. doi:10.1016/j.bcp.2011.01.001PMID 21219886.
  19.  Zheng L, Wang Q, Yang X, Guo X, Chen L, Tao L, et al. (2012). “Antiviral activity of FNC, 2′-deoxy-2′-β-fluoro-4′-azidocytidine, against human and duck HBV replication”. Antiviral Therapy17 (4): 679–687. doi:10.3851/IMP2094PMID 22452880S2CID 25576607.
  20.  Zhou Y, Zhang Y, Yang X, Zhao J, Zheng L, Sun C, et al. (2012). “Novel nucleoside analogue FNC is effective against both wild-type and lamivudine-resistant HBV clinical isolates”Antiviral Therapy17 (8): 1593–1599. doi:10.3851/IMP2292PMID 22910281S2CID 29382902.
  21.  Wang Q, Hu W, Wang S, Pan Z, Tao L, Guo X, et al. (September 2011). “Synthesis of new 2′-deoxy-2′-fluoro-4′-azido nucleoside analogues as potent anti-HIV agents”European Journal of Medicinal Chemistry46 (9): 4178–4183. doi:10.1016/j.ejmech.2011.06.020PMC 3164908PMID 21745701.
  22.  Sun L, Peng Y, Yu W, Zhang Y, Liang L, Song C, et al. (August 2020). “Mechanistic Insight into Antiretroviral Potency of 2′-Deoxy-2′-β-fluoro-4′-azidocytidine (FNC) with a Long-Lasting Effect on HIV-1 Prevention”. Journal of Medicinal Chemistry63 (15): 8554–8566. doi:10.1021/acs.jmedchem.0c00940PMID 32678592S2CID 220631451.
  23.  Li G, Wang Y, De Clercq E (April 2022). “Approved HIV reverse transcriptase inhibitors in the past decade”Acta Pharmaceutica Sinica. B12 (4): 1567–1590. doi:10.1016/j.apsb.2021.11.009PMC 9279714PMID 35847492.
  24.  Google Patents Search, “阿兹夫定” (with quotes), CN114149475A, CN111892636A.
  25.  Guokr.com (10 August 2022). “真实生物的真实面目”Huxiu.com. Retrieved 30 December 2022.

Further reading

Clinical data
Trade names捷倍安, 双新艾克
Other names2′-Deoxy-2′-β-fluoro-4′-azidocytidine (FNC), RO-0622
Legal status
Legal statusUS: Investigational drugCN: Conditional use Rx
Pharmacokinetic data
Bioavailability83% (rat, dog)[1]
Metabolismliver (CYP3A)[2]
Elimination half-life4 hours (dog)[1]
Identifiers
IUPAC name
CAS Number1011529-10-4 
PubChem CID24769759
DrugBankDB16407
ChemSpider24717759
UNIIIJ2XP0ID0K
ChEMBLChEMBL519846
Chemical and physical data
FormulaC9H11FN6O4
Molar mass286.223 g·mol−1
3D model (JSmol)Interactive image
SMILES
InChI

///////AZVUDINE, Genuine Biotech, APPROVALS 2022, CHINA 2022, FNC, HY 19314, RO 0622, RO-0622, SB17040, IJ2XP0ID0K, DTXSID901027757

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

……

Sontigidomide 


Sontigidomide 

CAS 2560577-69-5

Molecular Weight513.47
FormulaC26H22F3N3O5

N-[[2-(2,6-Dioxo-3-piperidinyl)-2,3-dihydro-1-oxo-1H-isoindol-5-yl]methyl]-α-oxo-4-[1-(trifluoromethyl)cyclopropyl]benzeneacetamide

enzeneacetamide, N-[[2-(2,6-dioxo-3-piperidinyl)-2,3-dihydro-1-oxo-1H-isoindol-5-yl]methyl]-α-oxo-4-[1-(trifluoromethyl)cyclopropyl]-

FDD2NVW84X, Sontigidomida

Sontigidomide (Compound 5) is an antineoplastic compound. Sontigidomide inhibits MOLM-13 cell proliferation more than 80% at 1 μM (3 days).

SCHEME

COUPLER………….

MAIN……….

PATENTS

WO2023070120  BioTheryX, Inc.

PATENT

US20200369679

https://patentscope.wipo.int/search/en/detail.jsf?docId=US311579044&_cid=P20-MD87Y5-18242-1

Example 5

Compound I-5: N-((2-(2,6-Dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)methyl)-2-oxo-2-(4-(1-(trifluoromethyl)cyclopropyl)phenyl)acetamide

Compound I-5 was synthesized as shown in Scheme 5.

   To a solution of 3-(5-(aminomethyl)-1-oxoisoindolin-2-yl)piperidine-2,6-dione 8 (80 mg, 0.258 mmol) in DCM (4 mL) at 0° C. was added TEA (52.2 mg, 0.516 mmol). After stirring for 2 min, 2-oxo-2-(4-(1-(trifluoromethyl)cyclopropyl)phenyl)acetyl chloride 13 (71.3 mg, 0.258 mmol) was added and the mixture was stirred at RT for 2 h. After concentration, the residue was purified using prep-HPLC eluting with ACN/H 2O (0.1% TFA) from 10% to 95% to afford compound I-5 (16.1 mg) in 12% yield. MS (ESI) m/z: 514.0 [M+H] +1H NMR (400 MHz, DMSO-d 6) δ 10.98 (s, 1H), 9.57 (t, J=6.0 Hz, 1H), 8.03-8.01 (m, 2H), 7.74-7.48 (m, 7H), 5.13-5.09 (m, 1H), 4.59-4.57 (m, 2H), 4.49-4.31 (m, 2H), 2.95-2.87 (m, 1H), 2.63-2.58 (m, 1H), 2.45-2.38 (m, 1H), 2.03-1.99 (m, 1H), 1.43-1.40 (m, 2H), 1.24-1.21 (m, 2H).

[1]. Kyle W.H. Chan, et al. Protein-targeting compounds and pharmaceutical compositions thereof, and their therapeutic applications. US20200369679.

////////Sontigidomide, FDD2NVW84X, CANCER, Sontigidomida

..

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

……

Keverprazan


  • P-CAB agent 2
  • keverprazan
  • 1978371-23-1
  • Keprason
  • SOC12UY3ZP

Keverprazan

432.5 g/mol

1-[5-(2-fluorophenyl)-1-[3-(3-methoxypropoxy)phenyl]sulfonylpyrrol-3-yl]-N-methylmethanamine

C22H25FN2O4S

CAS 2209911-80-6

Molecular Weight468.97
FormulaC22H26ClFN2O4S

Jiangsu Carephar Pharmaceuticals, is
a potassium ion competitive acidblocker (P-CAB) that was approved inFebruary2023inChina for thetreatment of refluxesophagitis or duodenal ulcer inadults

P-CAB agent 2 hydrochloride is a potent and orally active potassium-competitive acid blocker and a gastric acid secretion inhibitor. P-CAB agent 2 hydrochloride inhibits H+/K+-ATPase activity with an IC50 value of <100 nM. P-CAB agent 2 hydrochloride inhibits the hERG potassium channel with an IC50 value of 18.69 M. P-CAB agent 2 hydrochloride shows no acute toxicity and inhibits histamine (HY-B1204)-induced gastric acid secretion.

P-CAB agent 2 is a potent and orally active potassium-competitive acid blocker and a gastric acid secretion inhibitor. P-CAB agent 2 inhibits H+/K+-ATPase activity with an IC50 value of <100 nM. P-CAB agent 2 inhibits the hERG potassium channel with an IC50 value of 18.69 M. P-CAB agent 2 shows no acute toxicity and inhibits histamine (HY-B1204)-induced gastric acid secretion[1].

REF

https://pubs.acs.org/doi/10.1021/acs.jmedchem.4c02079?ref=PDF

KeverprazanHydrochloride. Keverprazan hydrochloride(4),developedby Jiangsu CarepharPharmaceuticals,is
apotassiumioncompetitiveacidblocker (P-CAB) thatwas approvedinFebruary2023inChina forthetreatmentofrefluxesophagitisorduodenal ulcer inadults.36Those illnesses arecaused by gastric acid entering the esophagus, leading toregurgitation and heartburn.37 Current treatments mainly
employ acid-suppressing therapies such as proton pumpinhibitors (PPIs, e.g., lansoprazole); however, PPIs do notacidicenvironments.38Toaddress those issues,P-CABshavebeen developed as a new class of acid suppressants.39
Keverprazan, a novel P-CAB, reversibly binds to gastricH+,K+-ATPase in competition against K+ ions, therebyinhibitingtheenzyme.40Itprovidesstableandsustainedgastricacidinhibitioneffectat20mgdose,whilebeingsafeandwelltoleratedupto60mginasingle-ascendingdosestudy.41Theoriginal synthesisof keverprazanhydrochloridedevelopedbyQinetal. isillustratedinScheme7.42TheroutebeganwithSN2alkylationof3-nitrophenol4.1withalkylbromide4.2
toformphenylether4.3.Thenitrogroupwasreducedtoaniline 4.4underRaney-Ni-mediatedhydrogenation.Aniline4.4wasconverted to sulfonyl chloride 4.5 via copper-mediated
sulfonylationin55%yield.Thesulfonyl chloridewascoupledwithpyrrolederivative4.6toformsulfonamide4.7in60%yield.

Finally, reductiveaminationofthealdehydewithmethylamine and sodium cyanoborohydride, followed by HCl salt formation,43furnishedkeverprazanhydrochloride(4)in18%yield.
The synthesis of pyrrole fragment 4.6was reported byArikawaetal.andillustratedinScheme8.39SN2displacementofα-bromoketone 4.8 with ethyl cyanoacetate 4.9 affordedintermediate 4.10. Treatment of 4.10 with anhydrous HCl in EtOAc followed by hydrogenation effected cyclization to form pyrazole 4.11. The ester was reduced to the corresponding
alcohol with DIBAL-H,andthealcoholoxidized to aldehyde4.6 with NMO and TPAP.

(36) Kang, C.Keverprazan Hydrochloride: first approval. Drugs 2023,
83, 639−643.
(37) Chen, S.; Liu, D.; Chen, H.; Liao, A.; Li, F.; Liu, C.; Li, X.; Li, S.;
Zhang, Y.; Wang, Y.; et al. The efficacy and safety of keverprazan, a
novel potassium-competitive acid blocker, in treating erosive
oesophagitis: a phase III, randomised, double-blind multicentre
study. Aliment. Pharmacol. Ther. 2022, 55, 1524−1533.
(38)Tan,N.-d.; Liu, X.-w.; Liu, C.-x.; Li, S.-b.; Chen, H.-h.; Li, X.; Wu,
H.; Liao, A.-J.; Zhen, Y.-b.; Shen, P.-z.; et al. Efficacy of keverprazan for
duodenal ulcer: a phase II randomized, double-blind, parallel
controlled trial. J. Gastroenterol. Hepatol. 2022, 37, 2060−2066.
(39) Arikawa, Y.; Nishida, H.; Kurasawa, O.; Hasuoka, A.; Hirase, K.;
Inatomi, N.; Hori, Y.; Matsukawa, J.; Imanishi, A.; Kondo, M.; et al.
Discovery of a novel pyrrole derivative 1-[5-(2-fluorophenyl)-1
(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamine fuma
rate (TAK-438) as a potassium-competitive acid blocker (P-CAB). J.
Med. Chem. 2012, 55, 4446−4456.
(40) Parsons, M. E.; Keeling, D. J. Novel approaches to the
pharmacological blockade of gastric acid secretion. Expert Opin. Invest.
Drugs 2005, 14, 411−421.
(41) Zhou,S.;Xie, L.; Zhou, C.; Zhao, Y.; Wang,L.;Ding,S.;Chen, J.;
Zhu, B.; Su, M.; Shao, F. Keverprazan, a novel potassium-competitive
acid blocker: single ascending dose safety, tolerability, pharmacoki
netics, pharmacodynamics and food effect in healthy subjects. Eur. J.
Pharm. Sci. 2023, 190, No. 106578.
(42) Qin, Y.; Su, M.; Jin, Q.; Chen, T.; Jiang, J. Pyrrole sulfonyl
derivative, and preparation method and medical use thereof. EP
3248963 B1, 2019.

SYN

WO2016119505

PATENT

WO2016119505A1]

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2016119505&_cid=P12-MD5D4X-02931-1

Embodiment 1: 

[0042]Preparation of 1-(5-(2-fluorophenyl)-1-((3-(3-methoxypropoxy)phenyl)sulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine (EXP 1)

1) Preparation of 1-(3-methoxypropoxy)-nitrobenzene (Compound 2) 

[0045]3-Nitrophenol (Compound 1, 1.0 g, 7.19 mmol), potassium carbonate (2.9 g, 21.6 mmol) and 1-bromo-3-methoxypropane (1.65 g, 10.79) were dissolved in anhydrous DMF (20 mL), stirred at 90 °C overnight, added with water (50 mL), and extracted with ethyl acetate (50 mL x 3). The organic phases were combined, dried, and concentrated to give a yellow solid 1-(3-methoxypropoxy)-nitrobenzene (Compound 2, 1.5 g, yield 100%). 

[0046]2) Preparation of 1-(3-methoxypropoxy)-aniline (Compound 3) 

[0047]1-(3-methoxypropoxy)-nitrobenzene (Compound 2, 1.0 g, 4.74 mmol) and Raney Ni (100 mg) were dissolved in anhydrous methanol (20 mL) and stirred overnight at room temperature under a hydrogen atmosphere. The mixture was filtered and the filtrate was dried to obtain solid 1-(3-methoxypropoxy)-aniline (Compound 3, 0.80 g, 91% yield). 

[0048]3) Preparation of 1-(3-methoxypropoxy)-benzenesulfonyl chloride (Compound 4) 

[0049]At 0°C, sodium nitrite (571 mg, 8.29 mmol) was added to acetic acid (10 mL) and aqueous hydrochloric acid solution (2N, 10 mL) of 1-(3-methoxypropoxy)-aniline (compound 3, 1.0 g, 5.52 mmol) in batches. After the addition was completed, the mixture was stirred at 0°C for 25 min to obtain solution I. At 0°C, cuprous chloride (190 mg, 1.1 mmol) was added to an acetic acid solution of sulfur dioxide (10 mL, 2N) to obtain solution II. At 0°C, solution I was added dropwise to solution II, and after the addition was completed, the mixture was naturally warmed to room temperature and stirred for 3 hours. The mixture was extracted with ethyl acetate (150 mL x 3), the organic phases were combined, dried, concentrated, and column chromatography (petroleum ether: ethyl acetate = 20:1) was performed to obtain 1-(3-methoxypropoxy)-benzenesulfonyl chloride (compound 4, 800 mg, yield 55%) as a yellow oil. 

[0050]4) Preparation of 5-(2-fluorophenyl)-1-((3-(3-methoxypropoxy)phenyl)sulfonyl)-1H-pyrrole-3-carbaldehyde (Compound 5) 

[0051]At 0°C, t-BuOK (233 mg, 2.08 mmol) was added to a solution of 5-(2-fluorophenyl)-1H-pyrrole-3-carboxaldehyde (200 mg, 1.04 mmol) in anhydrous THF (5 mL), and the mixture was stirred at 0°C for 30 min. After the reaction was completed, 15-crown-5 (542 mg, 2.08 mmol) and 1-(3-methoxypropoxy)-benzenesulfonyl chloride (compound 4, 412 mg, 2.08 mmol) were added respectively. After the addition was completed, the mixture was naturally heated to room temperature and stirred for 90 min. After the reaction was completed, the mixture was quenched with ice water (50 g) and extracted with ethyl acetate (50 mL x 3). The organic phases were combined, dried, concentrated, and purified by column chromatography (petroleum ether:ethyl acetate=8:1) to give a yellow solid 5-(2-fluorobenzene)-1-((3-(3-methoxypropoxy)phenyl)sulfonyl)-1H-pyrrole-3-carbaldehyde (260 mg, yield 60%). 

[0052]5) Preparation of 1-(5-(2-fluorophenyl)-1-((3-(3-methoxypropoxy)phenyl)sulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine (EXP 1) 

[0053]5-(2-Fluorobenzene)-1-((3-(3-methoxypropoxy)phenyl)sulfonyl)-1H-pyrrole-3-carbaldehyde (Compound 5, 500 mg, 1.19 mmol), acetic acid (144 mg, 2.39 mmol), and methylamine alcohol solution (1 mL) were dissolved in 3 mL of anhydrous methanol and stirred at room temperature for 4 h. NaBH 

3 CN (212 mg, 3.59 mmol) was added. The mixture was stirred for 60 min. Ice water (30 g) was added to quench the reaction and the mixture was extracted with ethyl acetate (50 mL x 3). The organic phases were combined, dried, concentrated, and subjected to column chromatography to obtain 1-(5-(2-fluorobenzene)-1-((3-(3-methoxypropoxy)phenyl)sulfonyl)-1H-pyrrole-3-yl)-N-methylmethanamine (EXP 1, 100 mg, 20%) as a white solid. 

[0054]

HPLC:99.4%;MS(ESI)m/z:[M+H] +=433.0; 1H-NMR(400MHz,DMSO-d6)δ:8.72(s,1H),7.78(d,1H),7.46-7.55(m,2H),7.21-7.32(m,3H),6.85-7.11(m,2H),6.83-6.85(m,1H),6.44(d,1H),3.95-4.02(m,4H),3.47(t,2H),3.32(s,3H),2.52(m,3H),1.94(t,2H)ppm。

[References]

[1] YinLin Qin, et al. Pyrrole sulfonyl derivative, and preparation method and medical use thereof. WO2016119505A1.

[1]. YinLin Qin, et al. Pyrrole sulfonyl derivative, and preparation method and medical use thereof. WO2016119505A1.

///////////KEVERPRAZAN, Keverprazan, CHINA 2023, APPROVAL 2023, Jiangsu Carephar Pharmaceuticals, ULCER

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

……

Risevistinel


Risevistinel

NYX-783 CAS 2591344-26-0, UNII-52TU5MZG22

NYX 2925, 2012536-16-0, X062KF5ZV3

C14H23N3O4, 297.35

UNII-52TU5MZG22,
X062KF5ZV3

(αS,4R)-α-[(1R)-1-Hydroxyethyl]-5-(2-methyl-1-oxopropyl)-1-oxo-2,5-diazaspiro[3.4]octane-2-acetamide

2,5-Diazaspiro[3.4]octane-2-acetamide, α-[(1R)-1-hydroxyethyl]-5-(2-methyl-1-oxopropyl)-1-oxo-, (αS,4S)-

(2S,3R)-3-hydroxy-2-[(4S)-5-(2-methylpropanoyl)-3-oxo-2,5-diazaspiro[3.4]octan-2-yl]butanamide

  • NYX-783, NYX 2925
  • CS-0113907
  • HY-135741
  • NYX-2925

Risevistinel (NYX-783) is a positive allosteric modulator of N-methyl-D-aspartate (NMDA) receptor. Nevadistinel can be used to inhibit cognitive impairment associated with neurodegenerative diseases, such as mild cognitive impairment, mild Alzheimer’s disease, Parkinson’s disease, Lewy body disease.

NYX-2925 is an N-methyl-D-aspartate receptor (NMDAR) modulator, and at low concentrations of endogenous agonist (glycine or D-serine) and in the presence of glutamate, NYX-2925 partially activates the NMDAR. NYX-2925 appears to act at a binding site that is distinct from NMDAR agonists or antagonists studied to date, such as D-cycloserine, ketamine, MK-801, or kynurenic acid. The mode of action of NYX-2925 seems to be distinct from that of all existing and emerging drugs that are indicated for the treatment of neuropathic pain. While current medications target individual elements of pain signal transmission or modulation, NYX-2925 can modulate multiple synaptic relays within pain circuits.


NYX-2925 is under investigation in clinical trial NCT04146896 (Efficacy and Safety of NYX-2925 in Subjects With Neuropathic Pain Associated With Diabetic Peripheral Neuropathy (DPN)).

SCHEME

COUPLER

MAIN

REF

US20210139489 Aptinyx Inc.

https://patentscope.wipo.int/search/en/detail.jsf?docId=US323750708&_cid=P11-MD3X0E-31059-1

Synthesis of methyl pyrrolidine-2-carboxylate (2S-E)

      To a stirring solution of L-proline (50 g, 434 mmol) in methanol was added thionyl chloride (37.5 ml, 521 mmol) at 0° C. and heated to 70° C. for 16 h. The reaction mixture was brought to RT and concentrated under vacuum to afford compound 2S-E as (70 g, 99%) as thick syrup (hydrochloride salt).
       1H-NMR: (500 MHz, DMSO-d 6): δ 4.15-4.13 (m, 1H), 3.65 (s, 3H), 3.35-3.30 (m, 2H), 2.23-2.15 (m, 1H), 1.86-1.78 (m, 3H), 1.41 (s, 9H);
      LCMS m/z: 129 [M ++1]

Synthesis of 1-tert-butyl 2-methyl pyrrolidine-1,2-dicarboxylate (2S-F)

      To a stirring solution of compound 2S-E (70 g, 422 mmol) in CH 2Cl (700 mL) were added Et 3N (183 mL, 1.26 mol) at 0° C. and stirred for 10 min. After added Boc-anhydride (184 mL, 845 mmol) at 0° C. and the reaction mixture was stirred at RT for 16 h. After consumption of the starting material (by TLC), the reaction was diluted with water (200 mL) and extracted with CH 2Cl (2×200 mL). The combined organic layer was washed with citric acid (1×150 mL), brine (1×200 mL). The organic layer was dried over Na 2SO and concentrated under reduced pressure to afford crude compound which was purified by column chromatography by eluting 50% EtOAC/n-hexane to obtain compound 2S-F (80 g, 83%) as thick syrup.
       1H-NMR: (400 MHz, DMSO-d 6): δ 4.15-4.13 (m, 1H), 3.65 (s, 3H), 3.35-3.30 (m, 2H), 2.23-2.15 (m, 1H), 1.86-1.78 (m, 3H), 1.41 (s, 9H);
      LCMS m/z: 229 [(M ++1)-Boc].

Synthesis of 1-tert-butyl 2-methyl 2-((benzyloxy) methyl) pyrrolidine-1,2-dicarboxylate (2S-G)

      To a stirring solution of compound 2S-F (25 g, 109 mmol) in THF (250 mL) was added LiHMDS (240 mL, 240 mmol) at −20° C. and stirred for 2 h. To this BOM-chloride (23 mL, 163 mmol) was added drop wise at −30° C. and stirred for 2 h. After consumption of the starting material (by TLC), the reaction was quenched with aqueous NH 4Cl solution (100 mL) and extracted with EtOAc (2×200 mL). The combined organic layer was washed with water (2×150 mL) followed by brine solution (2×100 mL). The organic layer was dried over Na 2SO and concentrated to obtain crude compound which was purified by column chromatography by eluting 10% EtOAc/n-hexane to afford compound 2S-G (30 g, 79%) as thick syrup.
       1H-NMR: (500 MHz, DMSO-d 6): δ 7.36-7.22 (m, 5H), 4.59-4.48 (m, 2H), 4.02-3.88 (m, 1H), 3.63 (s, 3H), 3.49-3.35 (m, 2H), 3.34-3.30 (m, 1H), 2.31-2.23 (m, 1H), 2.04-1.89 (m, 2H), 1.82-1.78 (m, 1H);
      LCMS m/z: 349.4 [(M ++1)-Boc]

Synthesis of 2-((benzyloxy) methyl)-1-(tert-butoxycarbonyl) pyrrolidine-2-carboxylic acid (2S-H)

      To a stirring solution of compound 2S-G (30 g, 86 mmol) in methanol (70 mL) was added NaOH solution (6.88 g in 70 mL H 2O) at RT. The reaction mixture was heated to 70° C. for 16 h. After consumption of the starting material (by TLC), the solvent from the reaction was evaporated under reduced pressure and diluted with EtOAc (2×200 mL). The separated aqueous layer was acidified using citric acid solution (pH-3) and extracted with EtOAc (2×250 mL). The combined organic layer was dried over Na 2SO and concentrated to afford crude was triturated with n-hexane to obtain compound 2S-H (25 g, 86.8%) as an off-white solid.
       1H-NMR: (400 MHz, DMSO-d 6): δ 12.35 (br s, 1H), 7.37-7.29 (m, 5H), 4.56-4.48 (m, 2H), 4.06-4.00 (m, 1H), 3.92-3.89 (m, 1H), 3.66-3.45 (m, 1H), 3.37-3.28 (m, 1H), 2.31-2.20 (m, 1H), 2.05-1.97 (m, 1H), 1.87-1.75 (m, 2H), 1.38 (s, 9H);
      LCMS m/z: 335.3 [M ++1]

Synthesis of 1-(tert-butoxycarbonyl)-2-(hydroxymethyl) pyrrolidine-2-carboxylic acid (2S-I)

      To a stirring solution of compound 2S-H (25 g, 74 mmol) in methanol (150 mL) was added 50% wet 10% Pd/C (7 g) at RT and stirred for 10 h under H atmosphere. After consumption of the starting material (by TLC), the reaction mixture was filtered through a pad of celite and the pad was washed with methanol (100 mL). Obtained filtrate was concentrated under reduced pressure to afford compound 2S-I (15 g, 82.8%) as white solid.
       1H-NMR: (400 MHz, DMSO-d 6): δ 4.66 (br s, 1H), 3.96-3.83 (m, 1H), 3.63-3.59 (m, 1H), 3.49-3.41 (m, 1H), 3.34-3.25 (m, 1H), 2.30-2.17 (m, 1H), 1.95-1.72 (m, 3H), 1.38 (s, 9H).
      Mass (ESI): m/z 245 [M ++1]

Synthesis of tert-butyl 2-(((2S,3R)-1,3-bis (benzyloxy)-1-oxobutan-2-yl) carbamoyl)-2-(hydroxymethyl) pyrrolidine-1-carboxylate (2S-J)

      To a stirring solution of compound 2S-I (18 g, 73.4 mmol) in CH 2Cl (180 mL) were added DIPEA (40 mL, 220 mmol), 2S-D (21.9 g, 73.4 mmol), HATU (41.8 g, 110 mmol) at RT and stirred for 16 h. After consumption of the starting material (by TLC), the reaction mixture was diluted with water (50 mL) and extracted with CH 2Cl (2×100 mL). The combined organic layer was washed with brine, dried over anhydrous Na 2SO 4, filtered and concentrated under reduced pressure. Obtained crude material was purified by silica gel column chromatography eluting with 30% EtOAc/n-hexane to afford compound 2S-J (20 g, 52%) as pale yellow thick syrup.
       1H-NMR: (400 MHz, DMSO-d 6): δ 8.25-8.12 (m, 1H), 7.31-7.27 (m, 10H), 5.85 (t, J=4.8 Hz, 1H), 5.14 (s, 2H), 4.54-4.49 (m, 2H), 4.31-4.20 (m, 1H), 4.15-4.07 (m, 1H), 3.91-3.50 (m, 1H), 3.52-3.37 (m, 1H), 3.31-3.27 (m, 2H), 2.35-2.07 (m, 1H), 1.95-1.90 (m, 1H), 1.73-1.52 (m, 2H), 1.39 (s, 9H), 1.19 (d, J=6.4 Hz, 3H);
      Mass (ESI): m/z 527.4 [M ++1]

Synthesis of tert-butyl 2-((2S,3R)-1,3-bis (benzyloxy)-1-oxobutan-2-yl)-1-oxo-2,5-diazaspiro [3.4] octane-5-carboxylate (2S-K)

      To a stirring solution of triphenylphosphine (24.7 g, 94 mmol) in THF (100 mL) was added DIAD (15.3 g, 75 mmol) at RT and stirred for 30 min. To this added compound 2S-J (20 g, 37.9 mmol) in (10 mL) THF slowly and reaction mixture was stirred at RT for 2 h. After consumption of the starting material (by TLC), the reaction was concentrated under reduced pressure. The crude material was purified by silica gel column chromatography eluting 25% EtOAc/n-hexane to afford compound 2S-K (17 g, 88%) as pale yellow thick syrup.
       1H-NMR: (400 MHz, DMSO-d 6): δ 7.33-7.26 (m, 5H), 7.23-7.18 (m, 5H), 5.10 (s, 2H), 4.80-4.73 (m, 2H), 4.60 (s, 2H), 4.31 (s, 2H), 4.05-4.00 (m, 2H), 1.80-1.68 (m, 4H), 1.39 (s, 9H), 1.18 (d, J=6.0 Hz, 3H);
      Mass (ESI): m/z 509.4 [M ++1]

Synthesis of (2S,3R)-2-(5-(tert-butoxycarbonyl)-1-oxo-2,5-diazaspiro [3.4] octan-2-yl)-3-hydroxybutanoic acid (2S-L)

      To a stirring solution of compound 2S-K (7 g, 13.7 mmol) in methanol (100 mL) was added 10% Pd/C (4 g) at RT and stirred for 6 h under H atmosphere. After consumption of the starting material (by TLC), the reaction mixture was filtered through a pad of celite and the pad was washed with methanol (50 mL). Obtained filtrate was concentrated under reduced pressure to obtained crude, which was triturated with n-pentane (50 mL) to afford compound 2S-L (4 g, 88%) as white solid.
       1H-NMR: (500 MHz, DMSO-d 6): δ 12.80 (br s, 1H), 4.78-4.73 (m, 1H), 4.21-4.19 (m, 1H), 4.09 (s, 2H), 3.55-3.46 (m, 2H), 2.09-2.05 (m, 2H), 1.80 (d, J=7.0 Hz, 1H), 1.38 (s, 9H), 1.35-1.28 (m, 2H), 1.17 (d, J=6.5 Hz, 3H)
      LCMS m/z: 329.6 [M ++1]
      

Synthesis of tert-butyl 2-((2S,3R)-1-amino-3-hydroxy-1-oxobutan-2-yl)-1-oxo-2,5-diazaspiro [3.4] octane-5-carboxylate (2S-FNL-2)

      To a stirring solution of compound 2S-L (500 mg, 1.52 mmol) in CH 2Cl (5 mL) were added DIPEA (0.8 mL, 4.57 mmol), EDCI·HCl (350 mg, 1.82 mmol) followed by HOBt (280 mg, 1.82 mmol), NH 4Cl (161 mg, 3.04 mmol) at 0° C. and stirred for 16 h at RT. After consumption of the starting material (by TLC), the reaction mixture was diluted with water (10 mL) and extracted with CH 2Cl (2×30 mL). The combined organic layer was washed with citric acid solution (2×30 mL). The organic layer was dried over anhydrous Na 2SO 4, filtered and concentrated under reduced pressure. Obtained crude material was purified by silica gel column chromatography eluting 2% MeOH/DCM to afford compound (2S-FNL-2) (200 mg, 40%) as colorless liquid.
       1H-NMR: (500 MHz, DMSO-d 6): δ 7.53 (s, 2H), 4.59 (s, 1H), 4.02 (s, 1H), 3.77-3.70 (m, 2H), 3.62-3.53 (m, 2H), 3.46-3.33 (m, 1H), 2.17-2.03 (m, 2H), 1.88-1.71 (m, 2H), 1.38 (s, 9H), 1.18 (d, J=6.5 Hz, 3H);
      Mass (ESI): 328.3 [M ++1]

Synthesis of (2S,3R)-3-hydroxy-2-(1-oxo-2,5-diazaspiro [3.4] octan-2-yl) butanamide (2S-FNL-3)

      To a stirring solution of compound (2S-FNL-2) (200 mg, 0.61 mmol) in CH 2Cl (5 mL) was added TFA (0.5 mL, 6.1 mmol) at 0° C. and stirred at RT for 3 h. After completion of reaction (by TLC), the reaction mixture was concentrated under reduced pressure to obtained crude compound which was triturated with n-pentane/diethylether (5 mL/5 mL) to afford compound (2S-FNL-3) (100 mg) as white solid (TFA salt).
       1H-NMR: (400 MHz, D 2O): δ 4.33-4.29 (m, 2H), 4.09 (d, 1H), 3.95 (d, 1H), 3.57-3.48 (m, 2H), 2.51-2.46 (m, 2H), 2.25-2.19 (m, 2H), 1.31 (d, 3H);
      LCMS, m/z: 455 [2M ++1]

Synthesis of (2S,3R)-3-hydroxy-2-(5-isobutyryl-1-oxo-2,5-diazaspiro [3.4] octan-2-yl) butanamide (NYX-2925)

      To a stirring solution of (2S-FNL-3) (500 mg (crude), 2.20 mmol) in CH 2Cl (10 mL) was added TEA (1 mL, 7.70 mmol) followed by isobutyryl chloride (256 mg, 2.42 mmol) at 0° C. and stirred for 16 h at RT. After consumption of the starting material (by TLC), the reaction mixture was diluted with water (10 mL) and extracted with CH 2Cl (2×30 mL). The combined organic layer was washed with citric acid solution (2×30 mL). The organic layer was dried over anhydrous Na 2SO 4, filtered and concentrated under reduced pressure. Obtained crude material was purified by silica gel column chromatography eluting 2% MeOH/DCM to afford the two diastereomers of (2S-FNL-4) (100 mg, 15.2%) as white solid. The two 2S-FNL-4 diastereomers were separated by repetitive silica gel column chromatography to provide NYX-2925.
       1H-NMR: (500 MHz, DMSO-d 6): δ 7.63 (br s, ex with D20, 1H), 7.18 (br s, ex with D20, 1H), 4.77 (d, J=4.0 Hz, ex with D 2O, 1H), 4.04-4.00 (m, 1H), 3.95 (d, J=6.5 Hz, 1H), 3.76 (d, J=5.5 Hz, 1H), 3.66-3.63 (m, 1H), 3.53 (q, J=8.0 Hz, 1H), 3.39 (d, J=5.5 Hz, 1H), 2.72 (septet, J=6.5 Hz, 1H), 2.14-2.05 (m, 2H), 1.92-1.86 (m, 2H), 1.10 (d, J=6.0 Hz, 3H), 1.02 (d, J=6.5 Hz, 3H), 0.99 (d, J=6.5 Hz, 3H);
      Mass (ESI): 298.4 [M ++1].

PATENT

WO2022086858 

WO2021021996

[1]. Moskal, et al. Methods of treating disorders associated with elevated levels of antibodies that interact with the NMDA receptor by administering a spiro-β-lactam compound. World Intellectual Property Organization, WO2021021996 A1. 2021-02-04.

//////Risevistinel, NYX 783, UNII-52TU5MZG22, Aptinyx Inc, NYX 2925, CS 0113907, HY 135741, NYX-2925

Rezatapopt


Rezatapopt, PC 14586

CAS 2636846-41-6

Molecular Weight545.57
SynonymsPC14586
FormulaC28H31F4N5O2
CAS No.2636846-41-6

4-[3-[4-[[(3S,4R)-3-fluoro-1-methylpiperidin-4-yl]amino]-1-(2,2,2-trifluoroethyl)indol-2-yl]prop-2-ynylamino]-3-methoxy-N-methylbenzamide

5W59S33KC9

Rezatapopt (PC14586) is an orally active antineoplastic agent. Rezatapopt binds to a pocket created by the TP53 Y220C mutation. Rezatapopt restores p53 tumor suppressor functions by stabilization of the p53 protein structure. Rezatapopt demonstrates tumor inhibition and regression in mouse models with established human tumor xenografts harboring the TP53 Y220C mutation.

SCHEME

COUPLER

COUPLER

MAIN

REF

PAPER

https://pubs.acs.org/doi/10.1021/acsmedchemlett.4c00379

2-Iodo-1-(2,2,2-trifluoroethyl)-1H-indol-4-amine 15 was prepared from 4-nitroindole as described in
WO2017143291. 1
H NMR (400 MHz, dimethylsulfoxide [DMSO]-d6) δ ppm 9.19–10.88 (m, 2 H), 7.63
(d, J=8.34 Hz, 1 H), 7.16–7.25 (m, 1 H), 7.04–7.14 (m, 2 H), 5.14–5.33 (m, 2 H). LCMS (ES+
, m/z):
340.9 [(M+H)+
].

SnCl2.2H2O (398.11 mg, 1.76 mmol, 0.20 eq.) was added to a solution of 2-iodo-1-(2,2,2-trifluoroethyl)-
1H-indol-4-amine 15 (3.00 g, 8.82 mmol, 1.00 eq.) and 1-methylpiperidin-4-one (1.20 g, 10.61 mmol,
1.20 eq.) in MeOH (10.00 mL). The mixture was stirred at 25 °C for 3 hours (h), and then NaBH3CN
(2.77 g, 44.1 mmol, 5.00 eq.) was added, stirring at 25 °C for 69 h. Thin-layer chromatography (TLC)
indicated that the starting material was consumed, and the reaction mixture was filtered. The filtrate was
poured into H2O (200 mL) and extracted with ethyl acetate ([EtOAc] 200 mL2). The combined organic layers were washed with H2O (200 mL), dried over Na2SO4, and concentrated under reduced pressure to give a residue. The crude material was purified by flash column chromatography (Silica gel, petroleum ether (PE) : EtOAc = 0:1) and then by preparative high performance chromatography ([prep-HPLC] column: Phenomenex Luna C18 10040mm5 um; mobile phase: [H2O (0.2% Formic acid-acetonitrile [ACN])]; gradient: 10%–50% acetonitrile over 8.0 minutes) to yield 2-iodo-N-(1-methylpiperidin-4-yl)-1- (2,2,2-trifluoroethyl)-1H-indol-4-amine 16 (2.50 g, 5.72 mmol, 64.93% yield) as a light-brown solid. 1 H NMR (400 MHz, DMSO-d6) δ ppm 8.24 (br s, 1 H, formic acid salt), 7.17 (s, 1 H), 6.85–6.95 (m, 1 H), 6.78 (br d, J = 7.99 Hz, 1 H), 6.16 (br d, J = 7.63 Hz, 1 H), 5.44 (br d, J = 2.62 Hz, 1 H), 4.99 (q, J = 8.54 Hz, 2 H), 3.33 (br s, 1 H), 2.85 (br d, J = 9.66 Hz, 2 H), 2.25 (br s, 3 H), 2.07–2.18 (m, 2 H), 1.94 (br d, J = 12.04 Hz, 2 H), 1.46–1.58 (m, 2 H). LCMS (ES+, m/z): 438.0 [(M+H)+]. Boc2O (26.03 g, 119.26 mmol, 6.00 eq.) was added to a solution of 2-methoxy-4-(methylsulfonyl)aniline 17 (4.00 g, 19.88 mmol, 1.00 eq.) in dioxane (40.00 mL) at 25 o C (room temperature). The reaction mixture was stirred at 110 °C for 16 h. TLC and LCMS indicated that the reaction was completed, and it was concentrated in vacuo. The residue was purified by column chromatography (SiO2, PE/EtOAc = 10/1 to 1:1) to yield tert-butyl (2-methoxy-4-(methylsulfonyl)phenyl)carbamate (6.00 g, 19.92 mmol, 72.6% purity, 72% yield) as a yellow gum. 1 H NMR (400 MHz, DMSO-d6) δ ppm 8.33 (s, 1 H), 8.03 (d, J = 8.38 Hz, 1 H), 7.47 (dd, J = 8.38, 2.00 Hz, 1 H), 7.44 (d, J = 2.00 Hz, 1 H), 3.91 (s, 3 H), 3.18 (s, 3 H), 1.47 (s, 9 H). LCMS (ES+, m/z): 324.1 [(M+Na)+ ]. NaH (867.27 mg, 60% purity, 21.69 mmol, 3.00 eq.) was added in portions at 0 °C to a mixture of tert-butyl (2-methoxy-4-(methylsulfonyl)phenyl)carbamate (3.00 g, 7.23 mmol, 1.00 eq.) in dimethylformamide ([DMF] 30.00 mL) and stirred at 0 °C for 0.5 h. 3-Bromoprop-1-yne (3.23 g, 21.69 mmol, 3.00 eq.) was added to the reaction mixture, stirring at 0 °C for 2.5 h. TLC (Plate 1: PE : EtOAc = 1:1) and LCMS indicated that the starting material was consumed, and the product was detected. The reaction mixture was poured into a saturated solution of NH4Cl (200 mL) at 0 o C and was extracted with EtOAc (200 mL3). The combined organic phase was dried over Na2SO4, filtered, and concentrated

in vacuo. The residue was purified by column chromatography (SiO2, PE : EtOAc = 5:1 to 1:2) to give
tert-butyl (2-methoxy-4-(methylsulfonyl)phenyl)(prop-2-yn-1-yl)carbamate (3.00 g, 8.85 mmol,
74% purity, 90% yield) as a light-yellow gum.
1
H NMR (400 MHz, DMSO-d6) δ ppm 7.53–7.56 (m, 1 H), 7.46–7.53 (m, 2 H), 4.10–4.51 (m, 2 H), 3.90
(s, 3 H), 3.27 (s, 3 H), 3.17 (t, J = 2.32 Hz, 1 H), 1.27–1.39 (m, 9 H). LCMS (ES+
, m/z): 283.9 [(M+H-tBu)+].
A solution of 4M HCl/EtOAc (20.00 mL) was added to the solution of tert-butyl (2-methoxy-4-
(methylsulfonyl)phenyl)(prop-2-yn-1-yl)carbamate (3.00 g, 6.54 mmol, 1.00 eq.) in EtOAc (1.00 mL).
The reaction mixture was stirred at 25 °C for 2 h. TLC indicated that the starting material was consumed
completely. The reaction mixture was concentrated in vacuo to yield 2-methoxy-4-(methylsulfonyl)-N-
(prop-2-yn-1-yl)aniline 18 (1.80 g, 7.53 mmol, 85.3% yield, HCl salt) as a yellow solid.
1
H NMR (400 MHz, DMSO-d6) δ ppm 7.38 (dd, J = 8.40, 1.60 Hz, 1 H), 7.22 (d, J = 1.60 Hz, 1 H), 6.75
(d, J = 8.80 Hz, 1 H), 3.99 (d, J = 2.4 Hz, 2 H), 3.87 (s, 3 H) 3.10 (s, 3 H), 3.08 (t, J = 2.31 Hz, 1 H).
LCMS (ES+
, m/z): 240.1 [(M+H)+
].
i-Pr2NH (2.08 g, 20.58 mmol, 2.91 mL, 10 eq.), CuI (392.02 mg, 2.06 mmol, 1 eq), 2-iodo-N-(1-
methylpiperidin-4-yl)-1-(2,2,2-trifluoroethyl)-1H-indol-4-amine 16 (0.9 g, 2.06 mmol, 1 eq.) and
Pd(PPh3)4 (475.71 mg, 411.67 μmol, 0.2 eq.) was added to a solution of 2-methoxy-4-(methylsulfonyl)-N-
(prop-2-yn-1-yl)aniline 18 (622.16 mg, 2.47 mmol, 1.2 eq.) in DMSO (10 mL) at 45 °C under N2. The
reaction mixture was stirred at 45 °C for 1 h. TLC (DCM/MeOH=10:1, Rf = 0.3) indicated that the
starting material was consumed completely. It was poured into ethylenediaminetetraacetic acid ([EDTA]
20 mL) and stirred for 1 h, then extracted with EtOAc (40 mL3). The combined organic phase was washed with brine (40 mL), dried with anhydrous Na2SO4, filtered, and concentrated in vacuo. The crude product was purified by column chromatography (SiO2, PE : EtOAc = 1:1 to dichloromethane (DCM) / MeOH = 10:1, Rf = 0.3), then by prep-HPLC (column: Phenomenex Luna(2) C18 25050 10u; mobile
phase: [water (0.1% trifluoroacetic acid)-ACN]; B%: 30%–50%, 20 min) to yield compound 13 (0.6 g,
1.09 mmol, 53.08% yield, 99.9% purity) as a light-yellow solid.
1 H NMR (400 MHz, DMSO-d6) δ ppm 1.41–1.54 (m, 2 H), 1.91 (br d, J = 11.00 Hz, 2 H), 1.95–2.08 (m,
2 H) 2.17 (s, 3 H), 2.68–2.80 (m, 2 H), 3.10 (s, 3 H), 3.20–3.29 (m, 1 H), 3.89 (s, 3 H), 4.36 (d,
J = 6.24 Hz, 2 H), 4.92 (q, J = 9.09 Hz, 2 H), 5.49 (d, J = 7.95 Hz, 1 H), 6.15 (d, J = 7.83 Hz, 1 H),
6.50 (t, J = 6.24 Hz, 1 H), 6.68 (d, J = 8.19 Hz, 1 H), 6.89 (d, J = 8.44 Hz, 1 H), 6.99 (t, J = 8.01 Hz,
1 H), 7.09 (s, 1 H), 7.25 (d, J = 1.83 Hz, 1 H), 7.39 (dd, J = 8.31, 1.83 Hz, 1 H). LCMS (ES+, m/z):
549.3 [(M+H)+
]

a
Reagents and conditions: (a) Pd(PPh3)4, CuI, diisopropylamine, DMSO, 20 °C, 1 h; (b) TMSCl, DMF, 0 °C, 0.5 h;
(c) BH3.THF, 0 °C, 0.5 h; (d) EtOAc/HCl, 20 °C, 1 h; (e) 10 eq. (CH2O)n, NaBH3CN, MeOH, 20 °C, 16 h; f)
LiOH.H2O, MeOH, 40 °C, 12 h; g) MeNH3Cl, HOBT, EDCI, TEA, DCM, RT, 16 h; h) Chiral SFC separation

PATENTS

WO2023016434  36%

WO2021061643

US20230024905

WO2023016434 Jacobio Pharmaceuticals Co., Ltd.

WO2023225477 PMV Pharmaceuticals, Inc.

 US20230024905 PMV Pharmaceuticals, Inc.

WO2021061643 PMV Pharmaceuticals, Inc.

WO2021262483, PMV Pharmaceuticals, Inc.

WO2023196993 PMV Pharmaceuticals, Inc.

WO2021262484  WO2021262541 

//////////Rezatapopt, PC 14586, 5W59S33KC9

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

Resigratinib


Resigratinib, KIN 3248

CAS 2750709-91-0

C26H27F2N7O3

523.5 g/mol

3-[2-(1-cyclopropyl-4,6-difluorobenzimidazol-5-yl)ethynyl]-1-[(3S,5R)-5-(methoxymethyl)-1-prop-2-enoylpyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide

Resigratinib (KIN-3248) is an experimental anticancer medication which acts as a fibroblast growth factor receptor inhibitor (FGFRi) and is in early stage human clinical trials.[1][2][3]

KIN-3248 is a small molecule that targets and inhibits oncogenic fibroblast growth factor receptors (FGFRs). It was designed to mainly target FGFR2 and FGFR3 alterations, which act as oncogenic drivers in 10-20% of cholangiocarcinoma and 20-35% of urothelial cancers, respectively. While effective, disease progression may occur 6 to 8 months after treatment with currently approved FGFR inhibitors is started, and this effect is usually associated with on-target resistance mutations in the kinase domain of FGFR. Therefore, the broad inhibition of FGFR isoforms may be effective against different types of tumors. The safety, tolerability, pharmacokinetics, and preliminary efficacy of KIN-3248 are currently being evaluated in adults with advanced tumors harboring FGFR2 and/or FGFR3 gene alterations. In February 2023, Kinnate Biopharma received Fast Track designation from the FDA for KIN-3248 to treat unresectable, locally advanced or metastatic cholangiocarcinoma (CCA).

SCHEME

COUPLER

COUPLER

MAIN

CONTINUED………….

REF

US11345681,

https://patents.google.com/patent/US11345681B1/en

Example 78

3-[2-(1-Cyclopropyl-4,6-difluoro-1,3-benzodiazol-5-yl)ethynyl]-1-[(3S,5R)-5-(methoxymethyl)-1-(prop-2-enoyl)pyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide

Step 1: 1-(Tert-butyl) 2-methyl (2R,4R)-4-((tert-butyldiphenylsilyl)oxy)pyrrolidine-1,2-dicarboxylate
      To a stirred solution of 1-(tert-butyl) 2-methyl (2R,4R)-4-hydroxypyrrolidine-1,2-dicarboxylate (8.00 g, 32.62 mmol) and imidazole (4.44 g, 65.23 mmol) in DMF (80.00 mL) was added tert-butyl(chloro)diphenylsilane (13.45 g, 48.93 mmol) at 0° C. over 30 min. The reaction mixture was stirred for 16 h at room temperature. The resulting mixture was diluted with water (400 mL) and extracted with EA (3×300 mL). The combined organic layers was washed with brine (5×500 mL), dried over anhydrous Na 2SO and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (6/1). The fractions contained desired product were combined and concentrated to afford 1-(tert-butyl) 2-methyl (2R,4R)-4-((tert-butyldiphenylsilyl)oxy)pyrrolidine-1,2-dicarboxylate (14.20 g, 90%) as a colorless oil. MS ESI calculated for C 2737NO 5Si [M+H] +, 484.24, found 484.25.
      Step 2: Tert-butyl (2R,4R)-4-[(tert-butyldiphenylsilyl)oxy]-2-(hydroxymethyl)pyrrolidine-1-carboxylate
      To a stirred solution of 1-(tert-butyl) 2-methyl (2R,4R)-4-((tert-butyldiphenylsilyl)oxy)pyrrolidine-1,2-dicarboxylate (30.00 g, 62.02 mmol) in THF (300.00 mL) was added LiBFi (6.08 g, 0.28 mol) in portions at 0° C. under nitrogen atmosphere. The reaction mixture was stirred for 16 h at room temperature under nitrogen atmosphere. The resulting mixture was acidified to pH 5 with HCl (1M) at 0° C. and then basified to pH 8 with saturated NaHCO (aq.). The resulting mixture was extracted with EA (4×500 mL). The combined organic layers was dried over anhydrous Na 2SO and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1). The fractions contained desired product were combined and concentrated to afford tert-butyl (2R,4R)-4-[(tert-butyldiphenylsilyl)oxy]-2-(hydroxymethyl)pynolidine-1-carboxylate (24.00 g, 85%) as a light yellow oil. MS ESI calculated for C 2637NO 4Si [M+H] +, 456.25, found 456.30.
      Step 3: Tert-butyl (2R,4R)-4-[(tert-butyldiphenylsilyl)oxy]-2-(methoxymethyl)pyrrolidine-1-carboxylate
      To a suspension of NaH (0.20 g, 8.33 mmol) in THF (18 mL) was added a solution of tert-butyl (2R,4R)-4-[(tert-butyldiphenylsilyl)oxy]-2-(hydroxymethyl)pyrrolidine-1-carboxylate (2.50 g, 5.49 mmol) in THF (64.00 mL) slowly at 0° C. under nitrogen atmosphere. After stirred at 0° C. for 1 h, to the above mixture was added CH 3I (1.17 g, 8.23 mmol) dropwise at 0° C. The reaction mixture was stirred for additional 3 h at room temperature. The resulting mixture was diluted with water (60 mL), and then extracted with EA (3×30 mL). The combined organic layers was dried over anhydrous Na 2SO and filtered. The filtrate was concentrated under reduced pressure to afford tert-butyl (2R,4R)-4-[(tert-butyldiphenylsilyl)oxyl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (2.30 g, 89%) as a light yellow solid. MS ESI calculated for C 2739NO 4Si [M+H] +, 470.26, found 470.30.
      Step 4: Tert-butyl (2R,4R)-4-hydroxy-2-(methoxymethyl)pyrrolidine-1-carboxylate
      To a stirred solution of tert-butyl (2R,4R)-4-[(tert-butyldiphenylsilyl)oxy]-2-(methoxymethyl)pyrrolidine-1-carboxylate (46.30 g, 98.57 mmol) in THF (375.00 mL) was added tetra-n-butylammonium fluoride (1 M in THF) (146.70 mL, 0.14 mol) at 0° C. The reaction mixture was stirred at room temperature for 3 h. The resulting mixture was diluted with water (1 L) and extracted with EA (3×500 mL). The combined organic layers was dried over anhydrous Na 2SO and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1). The fractions contained desired product were combined and concentrated to afford tert-butyl (2R,4R)-4-hydroxy-2-(methoxymethyl)pyrrolidine-1-carboxylate (16.60 g, 73%) as a light yellow oil. MS ESI calculated for C 1121NO [M+H] +, 232.15, found 232.20.
      Step 5: Tert-butyl (2R)-4-(3,5-dibromo-4-cyanopyrazol-1-yl)-2-methoxymethyl)pyrrolidine-1-carboxylate
      To a stirred solution of 3.5-dibromo-1H-pyrazole-4-carbonitrile (2.00 g, 7.97 mmol), tert-butyl (2R,4R)-4-hydroxy-2-(methoxymethyl)pyrrolidine-1-carboxylate (1.84 g, 7.97 mmol) and triphenylphosphine (3.14 g, 11.95 mmol) in THF (40.00 mL) was added diisopropyi azodicarboxylate (2.42 g, 11.95 mmol) dropwise at 0° C. under argon atmosphere. The reaction mixture was stirred for 2 h at room temperature. The resulting mixture was diluted with water (300 mL). The resulting mixture was extracted with EtOAc (3×200 mL). The combined organic layers was dried over anhydrous Na 2SO and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (5/1). The fractions contained desired product were combined and concentrated to afford tert-butyl (2R)-4-(3,5-dibromo-4-cyanopyrazol-1-yl)-2-(methoxymethyl)pyrrolidine-1-carboxylate (3.50 g, 94%) as a dark yellow solid. MS ESI calculated for C 1520Br 24[M+H−100] +, 361.99, 363.99, 365.99; found 362.10, 364.10, 366.10.
      Step 6: Tert-butyl (2S,4R)-4-[3-bromo-4-cyano-5-(methylamino)pyrazol-1-yl]-2-(methoxymethyl)pyrrollidine-1-carboxylate
      To a stirred solution of tert-butyl (2R)-4-(3,5-dibromo-4-cyanopyrazol-1-yl)-2-(methoxymethyl)pyrrolidine-1-carboxylate (1.00 g, 2.15 mmol) in NMP (10.00 mL) was added CH 3NH 2(2.98 mL, 5.96 mmol) at room temperature under nitrogen atmosphere. The reaction mixture was stirred for 16 h at 50° C. under nitrogen atmosphere. The resulting mixture was diluted with water (30 mL) and extracted with EA (3×50 mL). The combined organic layers was washed with brine (3×20 mL), dried over anhydrous Na 2SO 4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1). The fractions contained desired product were combined and concentrated to afford tert-butyl (2S,4R)-4-[3-bromo-4-cyano-5-(methylamino)pyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (0.67 g, 35%) as an off-white solid. MS ESI calculated for C 1624BrN 5[M+H−100] +, 314.11, 316.11, found 314.10, 316.10.
      Step 7: (2R,4S)-4-[3-bromo-5-[(tert-butoxycarbonyl)(methyl)amino]-4-cyanopyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate
      To a stirred solution of tert-butyl (2R,4S)-4-[3-bromo-4-cyano-5-(methylamino)pyrazol-1-yl]-2-(methoxymethyl)pyrrollidine-1-carboxylate (20.30 g, 49.00 mmol) in DCM (300.00 mL) were added Boc 2O (20.97 mL, 98.01 mmol), DMAP (0.60 g, 4.90 mmol) and Et 3N (20.43 mL, 0.14 mol) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred for 1 h at room temperature. The resulting mixture was diluted with water (3×200 mL) and extracted with DCM (3×200 mL). The combined organic layers was washed with brine (3×100 mL). The organic layer was dried over anhydrous Na 2SO and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1). The fractions contained desired product were combined and concentrated to afford (2R,4R)-4-[3-bromo-5-[(tert-butoxycarbonyl)(methyl)amino]-4-cyanopyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (24.00 g, 95%) as an off-white solid. MS ESI calculated for C 2132BrN 55[M+H] +, 514.16, 516.16, found 514.15, 516.15; 1H NMR (400 MHz, CDCl 3) δ 4.94-4.90 (m, 1H), 4.23-4.19 (m, 1H), 3.75-3.66 (m, 3H), 3.44-3.40 (m, 1H), 3.36 (s, 3H), 3.25 (s, 3H), 2.62-2.58 (m, 1H), 2.41-2.19 (m, 1H), 1.48 (s, 18H).
      Step 8: Tert-butyl (2R,4S)-4-(5-[(tert-butoxycarbonyl)(methyl)amino]-4-cyano-3-[2-(trimethylsilyl)ethynyl]pyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate
      To a stirred mixture of (2R,4S)-4-[3-bromo-5-[(tert-butoxycarbonyl)(methyl)amino]-4-cyanopyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (24.00 g, 46.65 mmol), CuI (1.78 g, 9.33 mmol), Pd(PPh 32Cl (3,27 g, 4.67 mmol) and trimethylsilylacetylene (19.78 mL, 0.20 mol) in DMF (240.00 mL) was added TEA (19,45 mL, 0.19 inol). The reaction mixture was degassed with nitrogen for three times and stirred for 2 h at 90° C. The resulting mixture was concentrated under reduced pressure. The residue was diluted with water (500 mL) and extracted with EA (4×500 mL). The combined organic layers was washed with brine (2×500 mL), dried over anhydrous Na 2SO 4, filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (2/1). The fractions contained desired product were combined and concentrated to afford tert-butyl (2R, 4S)-4-[5-[(tert-butoxycarbonyl)(methyl)amino]-4-cyano-3-[2-(trimethylisilyl)ethynyl]pyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (2.4.00 g, 96%) as a brown solid. MS ESI calculated for C 264155Si [M+H]+, 532.29, found 532.40; 1H NMR (400 MHz, CDCl 3) δ 4.93-4.89 (m, 1H), 4.23-4.17 (m, 1H), 3.68-3.52 (m, 3H), 3.42-3.37 (m, 1H), 3.35-3.33 (m, 3H), 3.25-3.20 (m, 3H), 2.63-2.58 (m, 1H), 2.33-2.13 (m, 1H), 1.46 (s, 18H), 0.27 (s, 9H).
      Step 9: Tert-butyl (2R,4S)-4-(5-[(tert-butoxycarbonyl)(methyl)amino]-4-cyano-3-ethynylpyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate
      To a stirred solution of tert-butyl (2R,4S)-4-[5-[(tert-(butoxycarbonyl)(methyl)amino]-4-cyano-3-[2-(trimethylsilyl)ethynyl]pyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (24.00 g, 45.14 mmol) in THF (200.00 mL) was added TBAF (67.70 mL, 67.70 mmol, 1 M in THF) at 0° C. The reaction mixture was stirred for 1 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was diluted with water (500 mL) and extracted with EA (3×500 mL). The combined organic layers was washed with brine (2×500 mL), dried over anhydrous Na 2SO 4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (2/1). The fractions contained desired product were combined and concentrated to afford tert-butyl (2R,4S)-4-[5-[(tert-butoxycarbonyl)(methyl)amino]-4-cyano-3-ethynylpyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (17.40 g, 83%) as an off-white solid, MS ESI calculated for C 23335[M+H] +, 460.25, found 460.40; 1H NMR (400 MHz, CDCl 3) δ 4.93-4.89 (m, 1H), 4.22-4.18 (m, 1H), 3.84-3.46 (m, 3H), 3.42-3.37 (m, 1H), 3.37-3.31 (m, 4H), 3.24 (s, 3H), 2.62-2.59 (m, 1H), 2.28-2.24 (m, 1H), 1.46 (s, 18H).
      Step 10: Tert-butyl (2R,4S)-4-[5-[(tert-butoxycarbonyl)(methyl)amino]-4-carbamoyl-3-ethynylpyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate
      To a stirred solution of ter tyl (2R,4S)-4-(5-[(tert-butoxycarbonyl)(methyl)amino]-4-cyano-3-ethynylpyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (17.40 g, 37.86 mmol) in DMSO (30.00 mL) and EtOH (150.00 mL) were added 0.5 M NaOH (87.09 mL, 43.54 mmol) and H 22(10.26 mL, 0.13 mol) at 0° C. The reaction mixture was stirred for 0.5 h at 0° C. Then the reaction mixture was warmed up to room temperature and stirred for another 0.5 h at room temperature. The resulting mixture was diluted with water (500 mL) and extracted with EA (3×500 mL). The combined organic layers was washed with brine (2×300 mL), dried over anhydrous Na 2SO 4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/2). The fractions contained desired product were combined and concentrated to afford tert-butyl (2R,4S)-4-[5-[(tert-butoxycarbonyl)(methyl)amino]-4-carbamoyl-3-ethynylpyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (17.20 g, 95%) as an off-white solid. MS ESI calculated for C 23355[M+H] +, 478.26, found 478.25; 1H NMR (300 MHz, CDCl 3) δ 6.80-6.74 (m, 1H), 5.69-5.62 (m, 1H), 5.04-5.00 (m, 1H), 4.23-4.19 (m, 1H), 3.75-3.67 (m, 3H), 3.49-3.42 (m, 1H), 3.39-3.32 (m, 3H), 3.14 (s, 3H), 2.72-2.60 (m, 1H), 2.32-2.21 (m, 1H), 1.62-1.31 (m, 18H).
      Step 11: 3-Ethynyl-1-[(3S,5R)-5-(methoxymethyl)pyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide dihydrochloride
      To a stirred mixture of tert-butyl (2R,4S)-4-[5-[(tert-butoxycarbonyl)(methyl)amino]-4-carbamoyl-3-ethynylpyrazol-1-yl]-2-(methoxymethyl)pyrrolidine-1-carboxylate (17.20 g, 36.02 minor) in DCM (170,00 mL) was added HCl (180.08 mL, 0.72 mol, 4 M in EA). The reaction mixture was stirred for 1 h at room temperature under argon atmosphere. The resulting mixture was concentrated and dried to afford 3-ethynyl-1-[(3S,5R)-5-(methoxymethyl)pyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide dihydrochloride (12.50 g, crude) as an off-white solid which was used in the next step directly without further purification. MS ESI calculated for C 1321Cl 252[M+H−2 HCl] +, 278.15, found 278.05.
      Step 12: 3-Ethynyl-1-[(3S,5R)-5-(methoxymethyl)-1-(prop-2-enoyl)pyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide
      To a stirred mixture of 3-ethynyl-1-[(3S,5R)-5-(methoxymethyl)pyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide dihydrochloride (12.50 g, 35.69 mmol) and K 2CO (172 mL, 0.43 mol, 2.5 M) in THF (250.00 mL) was added acryloyl chloride (2.89 g, 32.15 mmol) dropwise at 0° C. under argon atmosphere. The reaction mixture was stirred for 10 min at 0° C. The resulting mixture was diluted with water (500 mL) and extracted with EA (3×500 mL). The combined organic layers was washed with brine (2×300 mL), dried over anhydrous Na 2SO 4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1). The fractions contained desired product were combined and concentrated to afford 3-ethynyl-1-[(3S,5R)-5-(methoxymethyl)-1-(prop-2-enoyl)pyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide (11.10 g, 84%) as an off-white solid. MS ESI calculated for C 162153[M+H] +, 332.16, found 332.20; 1H NMR (400 MHz, CDCl 3) δ 6.76 (s, 1H), 6.60-6.36 (m, 2H), 5.74-5.68 (m, 1H), 5.50-5.20 (m, 2H), 4.55-4.39 (m, 1H), 4.06-3.83 (m, 3H), 3.53-3.40 (m, 2H), 3.36-3.35 (m, 3H), 3.03-2.99 (m, 3H), 2.68-2.60 (m, 1H), 2.37-2.23 (m, 1H).
      Step 13: N-cyclopropyl-3,5-difluoro-2-nitroaniline
      To a stirred solution of 1,3,5-trifluoro-2-nitrobenzene (4.50 g, 25.41 mmol) in EtOH (45.00 mL) was added aminocyclopropane (2.90 g, 50.82 mmol) dropwise at 0° C. under nitrogen atmosphere. The reaction mixture was stirred for 1 h at 0° C. The resulting mixture was diluted with water (100 mL) and extracted with EA (3×100 mL). The combined organic layers was washed with brine (2×50 mL), dried over anhydrous Na 2SO and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (2/1). The fractions contained desired product were combined and concentrated to afford N-cyclopropyl-3,5-difluoro-2-nitroaniline (4.11 g, 85%) as a yellow solid. 1H NMR (400 MHz, CDCl 3) δ 7.64 (s, 6.80-6.74 (m, 1H), 6.30-6.27 (m, 1H), 2.59-2.54 (m, 1H), 1.01-0.83 (m, 2H), 0.76-0.61 (m, 2H).
      Step 14: N-cyclopropyl-3,5-difluoro-4-iodo-2-nitroaniline
      To a stirred mixture of N-cyclopropyl-3,5-difluoro-2-nitroaniline (4.11 g, 19.19 mmol) in methanesulfonic acid (45.00 mL) was added NIS (4.53 g, 20.15 mmol) in portions at 0° C. The reaction mixture was stirred for 2 h at room temperature. The resulting mixture was quenched with ice/water (100 mL) at 0° C. The resulting mixture was basified to pH 8 with sat. NaOH and extracted with EA (3×100 mL). The combined organic layers was washed with brine (3×100 mL), dried over anhydrous Na 2SO and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1). The fractions contained desired product were combined and concentrated to afford N-cyclopropyl-3,5-difluoro-4-iodo-2-nitroaniline (4.50 g, 69%) a yellow solid. 1H NMR (400 MHz, CDCl 3) δ 7.53 (s, 1H), 6.90-6.88 (m, 1H), 2.57-2.54 (m, 1H), 1.07-0.85 (m, 2H ), 0.83-0.58 (m, 2H).
      Step 15: N1-cyclopropyl-3,5-difluoro-4-iodobenzene-1,2-diamine
      To a stirred mixture of N-cyclopropyl-3,5-difluoro-4-iodo-2-nitroaniline (4.40 g, 12.94 mmol) and NH 4Cl (2.77 g, 51.76 mmol) in EtOH (44.00 mL) and H 2O (8.80 mL) was added Fe (2.89 g, 51.76 mmol). The reaction mixtue was stirred at 70° C. for 6 h. The resulting mixture was diluted with water (150 mL) and extracted with EA×100 mL). The combined organic layers was washed with brine (2×50 mL), dried over anhydrous Na 2SO and filtered. The filtrate was concentrated under reduced pressure to afford N-cyclopropyl-3,5-difluoro-4-iodobenzene-1,2-diamine (3.30 g, crude) as a brown oil which was used in the next step directly without further purification. MS ESI calculated for C 992IN [M+H] +, 310.98, found 311.00; 1H NMR (300 MHz, CDCl 3) δ 6.68-6.61 (m, 1H), 2.50-2.41 (m, 1H), 0.89-0.73 (m, 2H), 0.78-0.51 (m, 2H).
      Step 16: 1-Cyclopropyl-4,6-difluoro-5-iodo-1,3-benzodiazole
      To a stirred solution of N 1-cyclopropyl-3,5-difluoro-4-iodobenzene-1,2-diamine (3.30 g, 10.64 mmol) in MeOH (33.00 mL) was added trimethyl orthoformate (3.39 g, 31.92 mmol) at room temperature under nitrogen atmosphere. The reaction mixture was stirred for 16 h at 70° C. The resulting mixture was cooled down to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/2). The fractions contained desired product were combined and concentrated to afford 1-cyclopropyl-4,6-difluoro-5-iodo-1,3-benzodiazole (1.70 g, 50%) as a yellow solid. MS ESI calculated for C 1172[M+H] +, 320.96, found 321.00; 1H NMR (300 MHz, CDCl 3) δ 7.91 (s, 1H), 7.22-7.18 (m, 1H), 3.41-3.36 (m, 1H), 1.31-1.02 (m, 4H).
      Step 17: 3-[2-(1-Cyclopropyl-4,6-difluoro-1,3-benzodiazol-5-yl)ethynyl]-1-[(3S,5R)-5-(methoxymethyl)-1-(prop-2-enoyl)pyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide
      To a stirred solution of 1-cyclopropyl-4,6-difluoro-5-iodo-1,3-benzodiazole (0.97 g, 3.02 mmol), 3-ethynyl-1-[(3S,5R)-5-(methoxymethyl)-1-(prop-2-enoyl)pyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide (1.00 g, 3.02 mmol), Pd(PPh 32Cl (0.21 g, 0.30 mmol) and CuI (0.11 g, 0.60 mmol) in DMF (15.00 mL) was added TEA (0.92 g, 9.05 mmol) dropwise at room temperature. The reaction mixture was degassed with argon for three times and stirred for 40 min at 90° C. The resulting mixture was cooled down to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford the crude product. Then the crude product was further purified by reverse phase flash with the following conditions: column: C18 silica gel; mobile phase: ACN in water (10 mmol/L NH 3HCO 3), 10% to 50% gradient in 30 min; detector: UV 254 nm. The fractions contained desired product were combined and concentrated to afford 3-[2-(1-cyclopropyl-4,6-difluoro-1,3-b enzodiazol-5-yl)ethynyl]-1-[(3S,5R)-5-(methoxymethyl)-1-(prop-2-enoyl)pyrrolidin-3-yl]-5-(methylamino)pyrazole-4-carboxamide (0.51 g, 33%) as a white solid. MS ESI calculated for C 262727[M+H] +, 524.21, found 524.35; 1H NMR (300 MHz, CDCl 3) δ 7.99 (s, 1H), 7.30-7.12 (m, 2H), 6.83 (brs, EH), 6.67-6.32 (m, 2H), 5.84-5.73 (m, 1H), 5.64-5.12 (m, 2H), 4.71-4.38 (m, 1H), 4.25-3.84 (m, 3H), 3.60-3.33 (m, 5H), 3.20-3.08 (m, 3H), 2.86-2.70 (m, 1H), 2.37-2.31 (m, 1H), 1.40-0.89 (m, 4H).

PATENT

WO2021247969 Kinnate Biopharma Inc EG78

WO2023107980 solid state forms, Kinnate Biopharma Inc

WO2023107979 FGFR kinase inhibitor, Kinnate Biopharma Inc

References

  1.  Franovic A, Mohan A, Uryu S, Wu Q, Jiang P, Miller N, et al. (February 2022). “Activity of KIN-3248, a next-generation pan-FGFR inhibitor, against acquired FGFR-gatekeeper and molecular-brake drug resistance mutations”. Journal of Clinical Oncology40 (4_suppl): 461. doi:10.1200/JCO.2022.40.4_suppl.461.
  2.  Harding JJ, Perez CA, Kato S, Sharma M, Garmezy B, Quah CS, et al. (February 2023). “First in human (FIH) phase 1/1b study evaluating KIN-3248, a next-generation, irreversible pan-FGFR inhibitor (FGFRi), in patients (pts) with advanced cholangiocarcinoma (CCA) and other solid tumors harboring FGFR2 and/or FGFR3 gene alterations”. Journal of Clinical Oncology41 (4_suppl): TPS637-TPS637. doi:10.1200/JCO.2023.41.4_suppl.TPS637S2CID 256257314.
  3.  Wang Z, Anderson KS (2022). “Therapeutic Targeting of FGFR Signaling in Head and Neck Cancer”Cancer Journal (Sudbury, Mass.)28 (5): 354–362. doi:10.1097/PPO.0000000000000615PMC 9523489PMID 36165723.
Identifiers
CAS Number2750709-91-0
PubChem CID162381323
UNIIW728TB393W
Chemical and physical data
FormulaC26H27F2N7O3
Molar mass523.545 g·mol−1
3D model (JSmol)Interactive image
SMILES
InChI

/////////Resigratinib, Pan-FGFR Inhibitor KIN-3248, KIN 3248, Pan-fibroblast Growth Factor Receptor Inhibitor KIN-3248, W728TB393W

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

Sebetralstat


Sebetralstat, KVD 900

CAS 1933514-13-6

491.5 g/mol

O5ZD2TU2B7

FDA 7/3/2025, Ekterly, To treat acute attacks of hereditary angioedema

N-[(3-fluoro-4-methoxypyridin-2-yl)methyl]-3-(methoxymethyl)-1-[[4-[(2-oxopyridin-1-yl)methyl]phenyl]methyl]pyrazole-4-carboxamide

Sebetralstat, sold under the brand name Ekterly, is a medication used for the treatment of hereditary angioedema.[1] Sebetralstat is a plasma kallikrein inhibitor.[1]

Sebetralstat was approved for medical use in the United States in July 2025.[1][2]

SYN

https://pubs.acs.org/doi/10.1021/acs.jmedchem.2c00921

aReagents and conditions: (a) 2-Hydroxypyridine (1.2 equiv), K2CO3 (3.0 equiv), acetone, 50 °C, 18 h, 78%; (b) methanesulfonyl chloride (1.3 equiv), Et3N, (1.4 equiv), dichloromethane, rt, 18h, 93%; (c) methyl 3-(methoxymethyl)-1H-pyrazole-4-carboxylate (0.83 equiv), K2CO3 (2.0 equiv), DMF, 60 °C, 18 h, 54%; (d) NaOH (3.0 equiv), THF-MeOH-H2O, rt, 18 h, 34%; (e) 22a (1.0 equiv), C-(3-fluoro-4-methoxy-pyridin-2-yl)-methylamine (1.0 equiv), HATU (1.1 equiv), Et3N (6.0 equiv), dichloromethane, rt, 4 h, 64%.

Synthesis of Sebetralstat

1-(4-Hydroxymethyl-benzyl)-1H-pyridin-2-one (19)

4-(Chloromethyl)benzyl alcohol 18 (5.0 g, 31.9 mmol) was added to a solution of potassium carbonate (13.2 g, 96 mmol) and 2-hydroxypyridine (3.6 g, 38.3 mmol) in acetone (250 mL). The reaction mixture was heated at 50 °C for 18 h and then concentrated in vacuo. The residue was partitioned between dichloromethane (300 mL) and water (300 mL). The organic layer was separated, and the aqueous layer was extracted with dichloromethane (2 × 300 mL). The combined organic layers were washed with brine (300 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash chromatography on silica (0–10% methanol in dichloromethane) to afford 19 (5.4 g, 25.1 mmol, 78% yield) as a white solid. MS (ESI) m/z 216.0 (M + H)+1H NMR (400 MHz, DMSO-d6) δ 7.76 (dd, J = 6.8, 2.1 Hz, 1H), 7.41 (ddd, J = 9.0, 6.6, 2.1 Hz, 1H), 7.34–7.21 (m, 4H), 6.41 (dd, J = 9.1, 1.3 Hz, 1H), 6.22 (td, J = 6.7, 1.4 Hz, 1H), 5.15 (t, J = 5.7 Hz, 1H), 5.07 (s, 2H), 4.46 (d, J = 5.7 Hz, 2H). 13C NMR (100 MHz, DMSO-d6) δ 161.4, 141.9, 140.0, 139.0, 135.7, 127.5, 126.6, 119.8, 105.4, 62.6, 50.8.

1-(4-Chloromethyl-benzyl)-1H-pyridin-2-one (20)

A reaction flask containing 1-(4-hydroxymethyl-benzyl)-1H-pyridin-2-one (19) (8.45 g, 39.3 mmol), dry dichloromethane (80 mL), and triethylamine (7.66 mL, 55.0 mmol) was cooled in an ice–water bath. Methanesulfonyl chloride (3.95 mL, 51.0 mmol) was added to the reaction at 0 °C, and ice–water bath cooling continued. After 15 min, the ice–water bath was removed and stirring continued at room temperature overnight. The reaction mixture was partitioned between dichloromethane (100 mL) and saturated aqueous ammonium chloride solution (100 mL). The aqueous layer was extracted with further dichloromethane (2 × 50 mL), and the combined organic layers were washed with brine (50 mL), dried over sodium sulfate, filtered, and concentrated to afford 20 (8.65 g, 36.6 mmol, 93% yield) as a pale yellow solid. MS (ESI) m/z 234.1 (M + H)+1H NMR (400 MHz, DMSO-d6) δ 7.79 (ddd, J = 6.8, 2.1, 0.7 Hz, 1H), 7.49–7.39 (m, 1H), 7.40 (d, J = 7.8 Hz, 2H), 7.28 (d, J = 8.4 Hz, 2H), 6.42 (ddd, J = 9.2, 1.3, 0.7 Hz, 1H), 6.24 (td, J = 6.7, 1.4 Hz, 1H), 5.09 (s, 2H), 4.73 (s, 2H). 13C NMR (101 MHz, DMSO-d6) δ 161.4, 140.1, 139.1, 137.6, 136.9, 129.0, 127.9, 119.9, 105.5, 50.8, 45.8.

Methyl 3-(Methoxymethyl)-1-(4-((2-oxopyridin-1(2H)-yl)methyl)benzyl)-1H-pyrazole-4-carboxylate (21a) and Methyl 5-(Methoxymethyl)-1-(4-((2-oxopyridin-1(2H)-yl)methyl)benzyl)-1H-pyrazole-4-carboxylate (21b)

Methyl 3-(methoxymethyl)-1H-pyrazole-4-carboxylate (2.11 g, 11.77 mmol; CAS No. 318496-66-1) was added to a solution of potassium carbonate (3.25 g, 23.54 mmol) and 1-(4-chloromethyl-benzyl)-1H-pyridin-2-one 20 (3.30 g, 14.12 mmol) in N,N-dimethylformamide (5 mL) and heated at 70 °C for 3 h. The reaction mixture was diluted with ethyl acetate (50 mL) and washed with brine (2 × 100 mL), and the organic layer was dried over magnesium sulfate, filtered, and concentrated in vacuo. The crude product was purified by flash chromatography (120 g column, 0–100% (10% ethanol in ethyl acetate) in isohexanes to afford two regioisomers: 21a (2.03 g, 5.47 mmol, 47% yield) as an off-white solid and 21b (350 mg, 0.92 mmol, 8% yield). 21a MS (ESI) m/z 368.1 (M + H)+1H NMR (400 MHz, DMSO-d6) δ 8.42 (s, 1H), 7.76 (dd, J = 6.8, 2.2 Hz, 1H), 7.41 (ddd, J = 8.9, 6.5, 2.1 Hz, 1H), 7.25 (d, J = 1.2 Hz, 4H), 6.40 (dt, J = 9.1, 1.0 Hz, 1H), 6.22 (td, J = 6.7, 1.4 Hz, 1H), 5.30 (s, 2H), 5.07 (s, 2H), 4.49 (s, 2H), 3.72 (s, 3H), 3.23 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 163.2, 161.8, 150.5, 140.6, 139.6, 137.6, 136.3, 135.6, 128.5, 128.4, 120.3, 111.8, 106.0, 66.0, 58.0, 55.1, 51.5, 51.2. 21b MS (ESI) m/z 368.1 (M + H)+1H NMR (400 MHz, DMSO-d6) δ 7.88 (s, 1H), 7.76 (dd, J = 6.8, 2.1 Hz, 1H), 7.41 (ddd, J = 8.9, 6.6, 2.1 Hz, 1H), 7.28–7.21 (m, 2H), 7.17 (d, J = 8.2 Hz, 2H), 6.43–6.36 (m, 1H), 6.22 (td, J = 6.7, 1.4 Hz, 1H), 5.35 (s, 2H), 5.06 (s, 2H), 4.78 (s, 2H), 3.75 (s, 3H), 3.25 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 163.4, 161.8, 142.4, 140.9, 140.5, 139.6, 137.4, 136.2, 128.3, 120.3, 112.8, 106.0, 61.7, 58.2, 53.0, 51.7, 51.2.

3-(Methoxymethyl)-1-(4-((2-oxopyridin-1(2H)-yl)methyl)benzyl)-1H-pyrazole-4-carboxylic acid (22a)

To methyl 3-(methoxymethyl)-1-(4-((2-oxopyridin-1(2H)-yl)methyl)benzyl)-1H-pyrazole-4-carboxylate 21a (3.77 g, 10.26 mmol) in tetrahydrofuran (5 mL) and methanol (5 mL) was added 2 M aqueous sodium hydroxide solution (15.39 mL, 30.80 mmol), and the reaction mixture was stirred at room temperature overnight. The reaction was acidified with 1 M aqueous HCl solution (50 mL) and extracted with ethyl acetate (50 mL). The organic layer was washed with brine (50 mL), dried over magnesium sulfate, filtered, and concentrated in vacuo to afford 22a (1.22 g, 3.45 mmol, 34% yield) as a white solid. MS (ESI) m/z 354.2 (M + H)+1H NMR (400 MHz, DMSO-d6) δ 12.32 (s, 1H), 8.32 (s, 1H), 7.76 (ddd, J = 6.8, 2.1, 0.7 Hz, 1H), 7.41 (ddd, J = 8.9, 6.6, 2.1 Hz, 1H), 7.30–7.20 (m, 4H), 6.40 (ddd, J = 9.1, 1.4, 0.7 Hz, 1H), 6.22 (td, J = 6.7, 1.4 Hz, 1H), 5.29 (s, 2H), 5.07 (s, 2H), 4.50 (s, 2H), 3.22 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 164.3, 161.8, 150.5, 140.6, 139.6, 137.6, 136.4, 135.6, 128.5, 128.4, 120.3, 113.0, 106.0, 66.0, 58.0, 55.1, 51.2.

3-Methoxymethyl-1-[4-(2-oxo-2H-pyridin-1-ylmethyl)-benzyl]-1H-pyrazole-4-carboxylic Acid (3-Fluoro-4-methoxy-pyridin-2-ylmethyl)-amide (14w)

3-(Methoxymethyl)-1-(4-((2-oxopyridin-1(2H)-yl)methyl)benzyl)-1H-pyrazole-4-carboxylic acid 22a (75 mg, 0.212 mmol), C-(3-fluoro-4-methoxy-pyridin-2-yl)-methylamine (49 mg, 0.212 mmol; CAS No. 1256812-75-5), and HATU (89 mg, 0.233 mmol) were suspended in anhydrous dichloromethane (3 mL) to which triethylamine (177 μL, 1.270 mmol) was added, sonicated, and then left to stir at room temperature for 4 h. The solvent was removed under reduced pressure, and the resulting residue was quenched with saturated aqueous ammonium chloride solution (5 mL). An off-white solid resulted, which was sonicated, filtered under reduced pressure, washed with water, and dried in a vacuum oven at 40 °C overnight. The residue was purified by chromatography eluting with 1% NH3 in MeOH/dichloromethane to afford 14w as a white solid (67 mg, 64% yield). MS (ESI) m/z 492.0 (M + H)+1H NMR (400 MHz, DMSO-d6) δ: 8.42 (t, J = 5.4 Hz, 1H), 8.29–8.21 (m, 2H), 7.75 (ddd, J = 0.7, 2.1, 6.8 Hz, 1H), 7.41 (ddd, J = 2.1, 6.6, 8.9 Hz,1H), 7.28–7.17 (m, 5H), 6.39 (ddd, J = 0.7, 1.4, 9.2 Hz, 1H), 6.22 (td, J = 1.4, 6.7 Hz, 1H), 5.28 (s, 2H), 5.07 (s, 2H), 4.57–4.46 (m, 4H), 3.92 (s, 3H), 3.25 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 161.9, 161.3, 153.0 (JC–F = 8.7 Hz), 147.5, 146.8 (JC–F = 253.5 Hz), 146.0 (JC–F = 7.2 Hz), 145.2 (JC–F = 11.6 Hz), 140.1, 139.1, 137.1, 136.0, 133.2, 128.1, 127.9, 119.9, 116.3, 108.7, 105.5, 66.3, 57.5, 56.4, 54.6, 50.7, 38.3.

PATENT

US10364238,

https://patentscope.wipo.int/search/en/detail.jsf?docId=US232820883&_cid=P22-MCYEU3-92408-1

Example 41

3-Fluoro-4-methoxy-pyridine-2-carbonitrile

      To a large microwave vial, cyanocopper (1.304 g, 14.56 mmol) was added to a solution of 2-bromo-3-fluoro-4-methoxypyridine (1 g, 4.85 mmol) in DMF (5 mL). The reaction vial was sealed and heated to 100° C. for 16 hrs. The reaction mixture was diluted with water (20 mL) and EtOAc (20 mL). The thick suspension was sonicated and required additional water (40 mL) and EtOAc (2×50 mL) with sonication to break-up the solid precipitated. The combined layers were filtered through a plug of celite and the organic layer isolated, washed with brine (50 mL), dried over magnesium sulfate, filtered and the solvent removed under reduced pressure to give a pale green solid identified as the desired compound 3-fluoro-4-methoxy-pyridine-2-carbonitrile (100 mg, 0.578 mmol, 12% yield)

(3-Fluoro-4-methoxy-pyridin-2-ylmethyl)-carbamic acid tert-butyl ester

      3-Fluoro-4-methoxy-pyridine-2-carbonitrile (100 mg, 0.578 mmol) was dissolved in anhydrous methanol (10 mL, 247 mmol) and nickel chloride hexahydrate (14 mg, 0.058 mmol) was added followed by di-tert-butyl dicarbonate (255 mg, 1.157 mmol). The resulting pale green solution was cooled in an ice-salt bath to −5° C. and then sodium borohydride (153 mg, 4.05 mmol) was added portionwise maintaining the reaction temperature ˜0° C. The deep brown solution was left to stir at 0° C. and slowly allowed to warm to rt and then left to stir at rt for 3 hrs. The reaction mixture was evaporated to dryness at 40° C. to afford a black residue which was diluted with DCM (10 mL) and washed with sodium hydrogen carbonate (10 mL). An emulsion formed so the organics were separated via a phase separating cartridge and concentrated. The crude liquid was purified by chromatography eluting with EtOAc/iso-Hexane to afford the title compound, (3-fluoro-4-methoxy-pyridin-2-ylmethyl)-carbamic acid tert-butyl ester as a clear yellow oil (108 mg, 62% yield)
      [MH] +=257

C-(3-Fluoro-4-methoxy-pyridin-2-yl)-methylamine hydrochloride salt

      (3-Fluoro-4-methoxy-pyridin-2-ylmethyl)-carbamic acid tert-butyl ester (108 mg, 0.358 mmol) was taken up in iso-propyl alcohol (1 mL) and then HCl (6N in iso-propyl alcohol) (1 mL, 0.578 mmol) was added at rt and left to stir at 40° C. for 2 hours. The reaction mixture was concentrated under reduced pressure and then triturated with ether, sonicated and then decanted to give a cream coloured solid (75 mg, 55% yield) identified as C-(3-fluoro-4-methoxy-pyridin-2-yl)-methylamine hydrochloride salt.
      [MH] +=157

3-Methoxymethyl-1-[4-(2-oxo-2H-pyridin-1-ylmethyl)-benzyl]-1H-pyrazole-4-carboxylic acid (3-fluoro-4-methoxy-pyridin-2-ylmethyl)-amide

      3-(Methoxymethyl)-1-(4-((2-oxopyridin-1(2H)-yl)methyl)benzyl)-1H-pyrazole-4-carboxylic acid (75 mg, 0.212 mmol), C-(3-Fluoro-4-methoxy-pyridin-2-yl)-methylamine hydrochloride salt (49 mg, 0.212 mmol) and HATU (89 mg, 0.233 mmol) were suspended in anhydrous DCM (3 mL) to which triethylamine (177 μL, 1.270 mmol) was added, sonicated and then left to stir at rt for 4 hours. The solvent was removed under reduced pressure and the resulting residue was quenched with ammonium chloride solution (5 mL). An off white solid resulted which was sonicated, filtered under reduced pressure washed with water and then placed in the vac oven at 40° C. overnight. The crude material was purified by chromatography eluting with (1% ammonia-methanol)/DCM to afford the 3-methoxymethyl-1-[4-(2-oxo-2H-pyridin-1-ylmethyl)-benzyl]-1H-pyrazole-4-carboxylic acid (3-fluoro-4-methoxy-pyridin-2-ylmethyl)-amide as a white solid (67 mg, 64% yield)
      [MH] +=492
      NMR (d 6-DMSO) δ: 3.25 (3H, s), 3.92 (3H, s), 4.46-4.57 (4H, m), 5.07 (2H, s), 5.28 (2H, s), 6.22 (1H, td, J=1.4, 6.7 Hz), 6.39 (1H, ddd, J=0.7, 1.4, 9.2 Hz), 7.17-7.28 (5H, m), 7.41 (1H, ddd, J=2.1, 6.6, 8.9 Hz), 7.75 (1H, ddd, J=0.7, 2.1, 6.8 Hz), 8.21-8.29 (2H, m), 8.42 (1H, t, J=5.4 Hz)

Medical uses

Sebetralstat is indicated for the treatment of acute attacks of hereditary angioedema.[1]

Pharmacology

Sebetralstat is a plasma kallikrein inhibitor that contains the unusual 2-pyridone heterocycle.[3]

Society and culture

Sebetralstat was approved for medical use in the United States in July 2025.[1] The US Food and Drug Administration granted the application for sebetralstat orphan drug designation.[4]

Names

Sebetralstat is the international nonproprietary name.[5]

Sebetralstat is sold under the brand name Ekterly.[1]

References

  1. Jump up to:a b c d e f g “Ekterly- sebetralstat tablet”DailyMed. 7 July 2025. Retrieved 9 July 2025.
  2. ^ “KalVista Pharmaceuticals Announces FDA Approval of Ekterly (sebetralstat), First and Only Oral On-demand Treatment for Hereditary Angioedema” (Press release). Kalvista. 7 July 2025. Retrieved 9 July 2025 – via Business Wire.
  3. ^ Davie RL, Edwards HJ, Evans DM, Hodgson ST, Stocks MJ, Smith AJ, et al. (October 2022). “Sebetralstat (KVD900): A Potent and Selective Small Molecule Plasma Kallikrein Inhibitor Featuring a Novel P1 Group as a Potential Oral On-Demand Treatment for Hereditary Angioedema”Journal of Medicinal Chemistry65 (20): 13629–13644. doi:10.1021/acs.jmedchem.2c00921PMC 9620001PMID 36251573.
  4. ^ “Sebetralstat Orphan Drug Designations and Approvals”U.S. Food and Drug Administration (FDA). Retrieved 9 July 2025.
  5. ^ World Health Organization (2022). “International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 87”. WHO Drug Information36 (1). hdl:10665/352794.
Clinical data
Trade namesEkterly
Other namesKVD-900, KVD900
License dataUS DailyMedSebetralstat
Routes of
administration
By mouth
ATC codeB06AC08 (WHO)
Legal status
Legal statusUS: ℞-only[1]
Identifiers
showIUPAC name
CAS Number1933514-13-6
PubChem CID121365142
IUPHAR/BPS11947
DrugBankDB18305
ChemSpider115006749
UNIIO5ZD2TU2B7
KEGGD12396
ChEMBLChEMBL5095248
Chemical and physical data
FormulaC26H26FN5O4
Molar mass491.523 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI

//////////Sebetralstat, FDA 2025, APPROVALS 2025, Ekterly, angioedema, KVD 900, O5ZD2TU2B7

str1

AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

This image has an empty alt attribute; its file name is research.jpg

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

Follow New Drug Approvals on WordPress.com

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

Join 37.9K other subscribers

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

DISCLAIMER

I , Dr A.M.Crasto is writing this blog to share the knowledge/views, after reading Scientific Journals/Articles/News Articles/Wikipedia. My views/comments are based on the results /conclusions by the authors(researchers). I do mention either the link or reference of the article(s) in my blog and hope those interested can read for details. I am briefly summarising the remarks or conclusions of the authors (researchers). If one believe that their intellectual property right /copyright is infringed by any content on this blog, please contact or leave message at below email address amcrasto@gmail.com. It will be removed ASAP