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

Mizagliflozin


Mizagliflozin

  • CAS 666843-10-3
  • 1X96A704XV
  • DSP-3235
  • KGA-3235

WeightAverage: 564.68
Monoisotopic: 564.315914393

Chemical FormulaC28H44N4O8

  • Dsp-3235 free base
  • GSK-1614235 free base
  • Kga-3235 free base

2,2-dimethyl-3-[3-[3-methyl-4-[[5-propan-2-yl-3-[(2S,3R,4S,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-1H-pyrazol-4-yl]methyl]phenoxy]propylamino]propanamide

Mizagliflozin is an SGLT1 inhibitor developed as a potential treatment for chronic constipation.[1][2] It progressed as far as Phase II trials in humans but was not approved for medical use, however it has since been investigated for other applications.[3][4]

MIZAGLIFLOZIN is a small molecule drug with a maximum clinical trial phase of II and has 1 investigational indication.

Mizagliflozin is under investigation in clinical trial NCT05721729 (Effect of Mizagliflozin Repeat Dosing on Adverse Events and Postprandial Glucose Excursions).

an SGLT1 inhibitor; structure in first source

  • OriginatorKissei Pharmaceutical
  • DeveloperKissei Pharmaceutical; Vogenx
  • ClassAmides; Antihypoglycaemics; Laxatives; Pyrazoles; Small molecules
  • Mechanism of ActionSodium-glucose transporter 1 inhibitors
  • Phase IIHypoglycaemia
  • Phase IGastroparesis
  • PreclinicalUnspecified
  • DiscontinuedConstipation
  • 18 Jun 2025Phase-I clinical trials in Gastroparesis in USA (PO) (Vogenx pipeline, June 2025)
  • 18 Jun 2025Preclinical trials in Undisclosed rare disease in USA (PO) (Vogenx pipeline, June 2025)
  • 01 Oct 2019Chemical structure information added

LIT

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

……

Legal status
Legal statusInvestigational
Identifiers
IUPAC name
CAS Number666843-10-3
PubChem CID10460535
ChemSpider8635948
UNII1X96A704XV
ChEMBLChEMBL5314923
Chemical and physical data
FormulaC28H44N4O8
Molar mass564.680 g·mol−1
3D model (JSmol)Interactive image
SMILES
InChI

References

  1.  Inoue T, Takemura M, Fushimi N, Fujimori Y, Onozato T, Kurooka T, et al. (July 2017). “Mizagliflozin, a novel selective SGLT1 inhibitor, exhibits potential in the amelioration of chronic constipation”. European Journal of Pharmacology806: 25–31. doi:10.1016/j.ejphar.2017.04.010PMID 28410751.
  2.  Fukudo S, Endo Y, Hongo M, Nakajima A, Abe T, Kobayashi H, et al. (September 2018). “Safety and efficacy of the sodium-glucose cotransporter 1 inhibitor mizagliflozin for functional constipation: a randomised, placebo-controlled, double-blind phase 2 trial”. The Lancet. Gastroenterology & Hepatology3 (9): 603–613. doi:10.1016/S2468-1253(18)30165-1PMID 30056028.
  3.  Ishida N, Saito M, Sato S, Tezuka Y, Sanbe A, Taira E, et al. (October 2021). “Mizagliflozin, a selective SGLT1 inhibitor, improves vascular cognitive impairment in a mouse model of small vessel disease”Pharmacology Research & Perspectives9 (5): e00869. doi:10.1002/prp2.869PMC 8480397PMID 34586752.
  4.  Tsunokake S, Iwabuchi E, Miki Y, Kanai A, Onodera Y, Sasano H, et al. (October 2023). “SGLT1 as an adverse prognostic factor in invasive ductal carcinoma of the breast”. Breast Cancer Research and Treatment201 (3): 499–513. doi:10.1007/s10549-023-07024-9PMID 37439959.

/////////666843-10-3, 1X96A704XV, DSP 3235, KGA 3235, Mizagliflozin, Dsp-3235 free base, GSK-1614235 free base, Kga-3235 free base

Ebselen


Ebselen

274.19 g/mol,
C13H9NOSe

2-phenyl-1,2-benzoselenazol-3-one

  • CAS 60940-34-3
  • 2-phenyl-1,2-benzoselenazol-3-one
  • 2-Phenyl-1,2-benzisoselenazol-3(2H)-one
  • Ebselene
  • PZ 51DR3305, and SPI-1005
  • 40X2P7DPGH

Ebselen is a benzoselenazole that is 1,2-benzoselenazol-3-one carrying an additional phenyl substituent at position 2. Acts as a mimic of glutathione peroxidase. It has a role as a neuroprotective agent, an apoptosis inducer, an anti-inflammatory drug, an antioxidant, a hepatoprotective agent, a genotoxin, a radical scavenger, an enzyme mimic, an EC 1.3.1.8 [acyl-CoA dehydrogenase (NADP(+))] inhibitor, an EC 1.8.1.12 (trypanothione-disulfide reductase) inhibitor, an EC 1.13.11.33 (arachidonate 15-lipoxygenase) inhibitor, an EC 1.13.11.34 (arachidonate 5-lipoxygenase) inhibitor, an EC 2.5.1.7 (UDP-N-acetylglucosamine 1-carboxyvinyltransferase) inhibitor, an EC 2.7.10.1 (receptor protein-tyrosine kinase) inhibitor, an EC 3.5.4.1 (cytosine deaminase) inhibitor, an EC 5.1.3.2 (UDP-glucose 4-epimerase) inhibitor, a ferroptosis inhibitor, an antifungal agent, an EC 3.4.22.69 (SARS coronavirus main proteinase) inhibitor, an anticoronaviral agent, an antibacterial agent, an antineoplastic agent and an EC 3.1.3.25 (inositolphosphate phosphatase) inhibitor.

Ebselen (also called PZ 51DR3305, and SPI-1005), is a synthetic organoselenium molecule under preliminary investigation as a drug candidate.[1] It belongs to the class of compounds related to benzene and its derivatives.[1] It is being developed by the Seattle biotechnology company, Sound Pharmaceuticals, Inc.[1] It has also been reported to target tubulin, blocking its polymerization.[2]

Ebselen has been investigated for the treatment and basic science of Meniere’s Disease, Type 2 Diabetes Mellitus, and Type 1 Diabetes Mellitus.
Ebselen has been entered into clinical trials as a lead compound intended for the potential treatment of various diseases.[3] Its most advanced clinical trial is a Phase III study in people with Meniere’s disease, completed in July 2024.[4]

In vitro, ebselen is a mimic of glutathione peroxidase and reacts with peroxynitrite.[5] It is purported to have antioxidant and anti-inflammatory properties.[1][5]

Synthesis

Generally, synthesis of the characteristic scaffold of ebselen, the benzoisoselenazolone ring system, can be achieved either through reaction of primary amines (RNH2) with 2-(chloroseleno)benzoyl chloride (Route I),[6] by ortho-lithiation of benzanilides followed by oxidative cyclization (Route II) mediated by cupric bromide (CuBr2),[7] or through the efficient Cu-catalyzed selenation / heterocyclization of o-halobenzamides, a methodology developed by Kumar et al.[8] (Route III).

SYN

Synthesis of ebselen from benzoic acid by ortholithiation of benzanilide SOCl 2 =Thionyl chloride, R-NH 2 =Substituted aryl mine, BuLi/THF=n-butyllithium/ tetrahydrofuran, CuBr 2 =Cupper bromide, CH 2 =CH- CH 2 -Br = Allyl bromide.  

SYN

New Chiral Ebselen Analogues with Antioxidant and Cytotoxic Potential

Molecules, March 2017, 22(3):492

DOI:10.3390/molecules22030492

SYN

https://pubs.acs.org/doi/10.1021/ol102027j

2-Phenylbenzo[d][1,2]selenazol-3(2H)-one (1) from 2-Iodo-N-phenylbenzamide (Typical
Procedure): Copper iodide (114 mg, 0.6 mmol) and 1,10-phenanthroline (108 mg, 0.6 mmol)
were added into DMF (3 mL) in a single neck flask. Resulted brownish solution was stirred for
15 min and then 2-iodo-N-phenylbenzamide1 (0.97 g, 3.0 mmol), selenium powder (0.29 g, 3.6
mmol), and potassium carbonate powder (0.65 g, 4.7 mmol) were added sequentially to same reaction flask. Brown colored reaction mixture was refluxed at 110oC using refluxing condenser
under nitrogen atmosphere. Progress of reaction was monitored by TLC. Reaction mixture was
refluxed for 8h. After this, reaction mixture poured over brine solution (60 mL) and stirred for 3
h. Product was precipitated as white solid which was collected by filtration over Buchner funnel,
product was washed with water (15 mL x 2), dried in air, dissolved in ethyl acetate, concentrated
over rotary evaporator, resulted brown solid which was purified by column chromatography
using hexane/ ethyl acetate (8:2) over silica gel. Yield 0.69 g (84%), mp 182-183 °C (180-181
°C).14,15 1H NMR (400 MHz, DMSO-d6)  8.09 (d, J = 8.0 Hz, 1H), 7.91 (d, J = 8.0 Hz, 1H),
7.71-7.62 (m, 3H), 7.51-7.43 (m, 3H), 7.28 (t, J = 8.0 Hz, 1H). 1H NMR (400 MHz, CDCl3) 
8.12 (d, 7.6 Hz, 1H), 7.68-7.62 (m, 4H), 7.52-7.41 (m, 3H), 7.29 (m, 1H). IR (plate): 3057, 2921,
1598, 1443, 1346, 1263, 1028 cm-1; ESMS m/z: 276 (M+H+).

2-Phenylbenzo[d][1,2]selenazol-3(2H)-one (1) from 2-Iodo-N-phenylbenzamide at 74 mmol
scale: Reaction was carried out at 74 mmol scale using 2-iodo-N-phenylbenzamide (24.00 g,
74.3 mmol), selenium powder (7.04 g, 89.1 mmol), CuI (2.83 g, 14.9 mmol), 1,10
phenanthroline (2.69 g, 14.9 mmol), and anhydrous potassium carbonate powder (15.40 g, 111.4
mmol) in DMF (50 mL) and procedure and workup followed are similar to 3.6 mmol scale
reaction. Yield 16.28 g (80%), Figure S1.
2-Phenylbenzo[d][1,2]selenazol-3(2H)-one (1) from 2-Bromo-N-phenylbenzamide: Ebselen 1
was prepared from 2-bromo-N-phenylbenzamide2 (1.00 g, 3.6 mmol), selenium powder (0.34 g,
4.3 mmol), K2CO3 powder (0.74 g, 5.4 mmol), CuI (137 mg, 0.7 mmol), and 1,10-phenanthroline
(130 mg, 0.7 mmol) in DMF (3 mL). Reaction mixture was refluxed for 16 h at 110oC. Progress of reaction was monitored by TLC. After completion of reaction, mixture was poured into brine
solution (60 mL) and the resulted white precipitate was washed with water (20 mL x 2), and
dried in air. Purification by column chromatography on silica gel using CH2Cl2 provided white
crystalline solid (0.77 g, 78%).
2-Phenylbenzo[d][1,2]selenazol-3(2H)-one (1) from 2-Chloro-N-phenylbenzamide: Reaction
was carried out at 4 mmol scale using 2-chloro-N-phenylbenzamide3 (1.00 g, 4.3 mmol), CuI
(172 mg, 0.9 mmol), 1,10-phenanthroline (162 mg, 0.9 mmol), selenium powder (0.41 g, 5.2
mmol), K2CO3 (0.89 g, 6.4 mmol) in DMF (4 mL). Reaction mixture was refluxed for 24 h at
110oC. Workup procedure is similar as followed for bromo substrate. Yield 0.55 g (47%).

History

The first patent for 2-phenyl-1,2-benzoselenazol-3(2H)-one was filed in 1980 and granted in 1982.[9]

Research

Ebselen is in preliminary clinical development for the potential treatment of hearing loss and depression, among other medical indications.[3][10]

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

……

References

  1.  “Ebselen”. DrugBank. 29 January 2025. Retrieved 4 February 2025.
  2.  Baksheeva VE, La Rocca R, Allegro D, Derviaux C, Pasquier E, Roche P, Morelli X, Devred F, Golovin AV, Tsvetkov PO (2025). “NanoDSF Screening for Anti-tubulin Agents Uncovers New Structure–Activity Insights”. Journal of Medicinal Chemistrydoi:10.1021/acs.jmedchem.5c01008.
  3.  “Ebselen pipeline”. Sound Pharmaceuticals, Inc. 2025. Retrieved 4 February 2025.
  4.  “SPI-1005 for the Treatment of Meniere’s Disease (STOPMD-3)”. ClinicalTrials.gov, US National Library of Medicine. 1 August 2024. Retrieved 4 February 2025.
  5.  Schewe T (October 1995). “Molecular actions of ebselen – an antiinflammatory antioxidant”. General Pharmacology26 (6): 1153–69. doi:10.1016/0306-3623(95)00003-JPMID 7590103.
  6.  Kamigata N, Iizuka H, Izuoka A, Kobayashi M (July 1986). “Photochemical Reaction of 2-Aryl-1, 2-benzisoselenazol-3 (2 H)-ones”Bulletin of the Chemical Society of Japan59 (7): 2179–83. doi:10.1246/bcsj.59.2179.
  7.  Engman L, Hallberg A (1989-06-01). “Expedient synthesis of ebselen and related compounds”. The Journal of Organic Chemistry54 (12): 2964–2966. doi:10.1021/jo00273a035ISSN 0022-3263.
  8.  Balkrishna SJ, Bhakuni BS, Chopra D, Kumar S (December 2010). “Cu-catalyzed efficient synthetic methodology for ebselen and related Se-N heterocycles”. Organic Letters12 (23): 5394–7. doi:10.1021/ol102027jPMID 21053969.
  9.  DE3027073A1, Etschenberg, Eugen Dr; Renson, Marcel Prof Dipl-Chem Jupille & Winkelmann, Johannes Dr 5000 Köln, “2-phenyl-1,2-benzisoselenazol-3(2h)-on enthaltende pharmazeutische praeparate und ihre verwendung”, issued 1982-02-18
  10.  “Ebselen search: list of clinical trials sponsored by Sound Pharmaceuticals”. ClinicalTrials.gov, US National Library of Medicine. 2025. Retrieved 4 February 2025.
Names
Preferred IUPAC name2-Phenyl-1,2-benzoselenazol-3(2H)-one
Identifiers
CAS Number60940-34-3 
3D model (JSmol)Interactive imageInteractive image
ChEBICHEBI:77543 
ChEMBLChEMBL51085 
ChemSpider3082 
ECHA InfoCard100.132.190 
PubChem CID3194
UNII40X2P7DPGH 
CompTox Dashboard (EPA)DTXSID7045150 
InChIcheckcheck
SMILES
Properties
Chemical formulaC13H9NOSe
Molar mass274.17666
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).
References
  1. Zhou Y, Zhang Y, Zhao D, Yu X, Shen X, Zhou Y, Wang S, Qiu Y, Chen Y, Zhu F: TTD: Therapeutic Target Database describing target druggability information. Nucleic Acids Res. 2024 Jan 5;52(D1):D1465-D1477. doi: 10.1093/nar/gkad751. [Article]

////////Ebselen, Ebselene, PZ 51DR 3305, SPI 1005, PHASE 3, 40X2P7DPGH, Meniere’s Disease, Type 2 Diabetes Mellitus, Type 1 Diabetes Mellitus

Myself, NIPER-G and NDTL Collaborate to Synthesize ‘Methandienone LTM’ for Global Anti-Doping Efforts


A proud moment for me [ ANTHONY MELVIN CRASTO ] as Scientific Advisor at Niper-G Dept Pharma Ministry of Chemicals and Fertilizers Govt of India 🇮🇳

Congrats team Niper-G and team National Dope Testing Lab Govt of India 🇮🇳 Prof. (Dr.) P. L. Sahu and Myself being a part of it as Scientific Advisor Niper-G for few years in Medicinal chem dept. Launched by Union minister Mansukh Mandaviya on 4th sept 2025 in Delhi

will be distributed across the world via World Anti-Doping agency WADA

Methandienone LTM is high purity rare reference material 

Hope my interactions and guidance has given fruitful results. Thanks to Dr Murty for appointing me as advisor. Methandienone LTM will make India proud across the World 🌎

A great achievement for India 🇮🇳 as nation and advanced capability demonstration

https://www.pib.gov.in/PressReleasePage.aspx?PRID=2163812

https://lnkd.in/d9KWWsJ9

India Develops Rare Reference Material for Enhanced Anti-Doping Testing in Sports

NIPER Guwahati and NDTL Collaborate to Synthesize ‘Methandienone Long-Term Metabolite’ for Global Anti-Doping Efforts

//////////NIPER-G, NDTL, Methandienone LTM, Global Anti-Doping, INDIA

Sergliflozin Etabonate


 

Sergliflozin Etabonate

408504-26-7 Cas no

Ethyl [(2R,3S,4S,5R,6S)-3,4,5-trihydroxy-6-[2-[(4-methoxyphenyl)methyl]phenoxy]oxan-2-yl]methyl carbonate

2-(4-methoxybenzyl)phenyl 6-O-ethoxycarbonyl-beta-D-glucopyranoside
ethyl [(2R,3S,4S,5R,6S)-3,4,5-trihydroxy-6-[2-[(4-methoxyphenyl)methyl]phenoxy]tetrahydropyran-2-yl]methyl carbonate
ethyl [(2R,3S,4S,5R,6S)-3,4,5-trihydroxy-6-{2-[(4-methoxyphenyl)methyl]phenoxy}oxan-2-yl]methyl carbonate

PHASE 2……….TYPE 3 DIABETES AND OBESITY
A SGLT-2 inhibitor potentially for the treatment of type 2 diabetes and obesity.

GW-869682; GW-869682X; KGT-1251

  • etabonate de sergliflozine
  • etabonato de sergliflozina

 MW 448.4, C23H28O9

KISSEI INNOVATOR
GSK DEVELOPER

Sergliflozin Etabonate is a benzylphenol glucoside and selective sodium-glucose co-transporter subtype 2 (SGLT2) inhibitor with antihyperglycemic activity. Its prodrug form, sergliflozin etabonate, is orally available and is converted to sergiflozin upon absorption.

Sergliflozin etabonate (INN/USAN,[1][2] codenamed GW869682X) is an investigational anti-diabetic drug being developed by GlaxoSmithKline. It did not undergo further development after phase II

Sergliflozin inhibits subtype 2 of the sodium-glucose transport proteins (SGLT2), which is responsible for at least 90% of the glucose reabsorption in the kidney. Blocking this transporter causes blood glucose to be eliminated through the urine.[3][4]

Chemistry

Etabonate refers to the ethyl carbonate group. The remaining structure, which is the active substance, is called sergliflozin.

Sergliflozin

   [PDF] Design, Syntheses, and SAR Studies of Carbocyclic Analogues of …onlinelibrary.wiley.com974 × 740Search by imageDesign, Syntheses, and SAR Studies of Carbocyclic Analogues of Sergliflozin as Potent SodiumDependent Glucose Cotransporter 2 In

 Sergliflozin Etabonate is a benzylphenol glucoside and selective sodium-glucose co-transporter subtype 2 (SGLT2) inhibitor with antihyperglycemic activity. Its prodrug form, sergliflozin etabonate, is orally available and is converted to sergiflozin upon absorption.

Figure US20130096076A1-20130418-C00003

sergliflozin and prodrugs of sergliflozin, in particular sergliflozin etabonate, including hydrates and solvates thereof, and crystalline forms thereof. Methods for its manufacture are described in the patent applications EP 1344780 and EP 1489089 for example.

The compounds are described in EP 1 329 456 A1 and a crystalline form ofSergliflozin etabonate is described in EP 1 489 089 A1.

PATENT

US6872706B2

https://patentscope.wipo.int/search/en/detail.jsf?docId=US40677423&_cid=P20-MF4ZUQ-42384-1

PATENT

WO2001068660A1

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2001068660&_cid=P20-MF4ZXC-45172-1

SYN

Heterocycles 2016, 92, 1599

Our initial synthetic route of Serglifrozin etabonate (1) in early development consisted of six steps,
including synthesis of tetra-O-acetyl-D-glucopyranosyl trichloroacetimidate (9), as shown in Scheme 1
and Scheme 2 The first step is the coupling reaction of phenol (3) and 4-methoxybenzyl chloride (4) in the presence of
lithium hydroxide monohydrate (LiOH·H2O) to provide the aglycon 5 in a 30% yield following
chromatographic purification (Scheme 1). We prepared 9 separately by mono-deacetylation of
penta-O-acetyl-β-D-glucopyranose (7) with N,N-dimethylethylenediamine in THF followed by reaction of
the crude product of 8 with trichloroacetonitrile in the presence of potassium carbonate (K2CO3) in ethyl
acetate (EtOAc) (Scheme 2). Next, we carried out glycosylation of 5 with 9 in the presence of boron
trifluoride diethyl etherate (BF3·OEt2) in EtOAc to produce 6 in a 77% yield. The obtained 6 was
deacetylated with sodium methoxide (NaOMe) in MeOH to produce Serglifrozin (2) in a 73% yield, and
reaction of the isolated 2 with ethyl chloroformate in the presence of 2,6-lutidine in acetone provided 1 in
a 66% yield. The overall yield from 3 was 11%. While this route was capable of supplying small
amounts of 1, it suffered from several disadvantages.
The coupling reaction between 3 and 4 provided the aglycon 5 in low yield (30%); thus, chromatographic
purification was required to obtain highly pure 5. The trichloroimidation reaction of 8 is too hazardous
for large-scale manufacturing, because an excess amount of trichloroacetonitrile, a volatile and highly
toxic reagent, is required to obtain the trichloroacetimidate 9. Furthermore, 9 is too unstable to use
conveniently in large-scale manufacturing. Trichloroacetamide, a sublimation compound, is formed as a
by-product from the glycosylation of 5 with 9. Thus, the vacuum line and the vacuum pump of the
manufacturing equipment would be polluted by trichloroacetamide.
Because of these issues, this synthetic method is unsuitable for large-scale manufacturing. Therefore,
we investigated alternative processes for the preparation of 1, suitable for large-scale manufacturing. An improved synthetic method for 1 was achieved in a five-step procedure without purification of 6
(intermediate), as shown in Scheme 3.

The Friedel-Crafts acylation of anisole (10) with 2-methoxybenzoyl chloride (11) in the presence of
aluminum chloride (AlCl3) at 110 °C provided benzophenone (12), which was selectively demethylated
on the methoxy group at the 2-position. The crude product of 12 was crystallized from MeOH to
provide highly pure 12 in a 78% yield. Hydrogenation of 12 in EtOH with 0.3–0.4 MPa H2 at room
temperature in the presence of 10% Pd/C provided 5. The crude product of 5 was crystallized from
toluene/n-heptane to provide highly pure 5 in an 88% yield.
The key step of the synthesis was the formation of the O-glycosylated product 6. In the initial synthesis,
it was necessary to isolate 6 to remove trichloroacetamide. Consequently, 2 was provided in a 56%
yield from 5. To obtain 6 efficiently without using the trichloroacetimidate (9), we evaluated several
conditions for the direct O-glycosylation of 5 with 7. The results are summarized in Table 1. The
O-glycosylation of 5 with 7 (200 mol%) in the presence of boron trifluoride diethyl etherate (BF3·OEt2;
100 mol%) in dichloromethane (DCM) at room temperature provided the crude product of 6 with a good
yield (80%) and β-selectivity (94/6), and then the deacetylation of the crude product of 6 in the presence
of sodium methoxide (NaOMe) in MeOH proceeded almost quantitatively to provide 2 in a 71% isolated
yield from 5 (run 1). Using this method, it was not necessary to isolate 6 because the excess amount of
7 was converted to glucose and removed to the aqueous layers in the deacetylation step. Use of DCM is
undesirable for large-scale manufacturing because quenching of O-glycosylation with water is highly
exothermic and washing of the DCM layer with water is a complicated procedure. Additionally, it is
strongly desirable to avoid using DCM in a manufacturing process due to environmental issues. For the reasons mentioned above, we attempted to use toluene as an alternative solvent. The O-glycosylation in
the presence of BF3·OEt2 (100 mol%) in toluene at 30 °C did not proceed completely, and the yield of 6
was lower than run 1 (run 2). We concluded that the lower solubility of 7 in toluene, compared with
DCM, caused the low yield. Because it was difficult to increase the amount of toluene from the
perspective of manufacturing efficiency, we tried to improve its solubility by optimizing the reagent
equivalent. Fortunately, we found that an excess amount of BF3·OEt2 enhanced the solubility of 7 in
toluene, and using 300 mol% of BF3·OEt2 in toluene provided 6 in a good yield (80%), similar to that
when using DCM (run 3). In contrast, reducing the amount of 7 provided 6 in an insufficient yield, and
2 was consequently provided in a lower yield (60%) (run 4). To achieve higher β-selectivity and an
increased yield, triethylamine (Et3N) was added to the O-glycosylation of 5 with 7 in the presence of
BF3·OEt2, according to the method of Lee et al.
9 Addition of Et3N (30 mol%) at 30 °C resulted in both
higher yield (89%) and higher β-selectivity (97/3) to provide 2 with a 79% isolated yield (run 5).
Increasing the amount of Et3N to 60 mol% at 30 °C resulted in a lower yield (85%) of 6 compared with
run 5, and the yield of 2 decreased (74%) (run 6). Increasing the reaction temperature to 40 °C in the
presence of 60 mol% of Et3N achieved the best results for both high yield (90%) and high β-selectivity
(99/1) to provide 2 in an 80% yield (run 7).

6-O-Ethoxycarbonyl-2-[(4-methoxyphenyl)methyl]phenyl-β-D-glucopyranoside (1). Ethyl
chloroformate (407 mg, 3.75 mol) was added drop-wise to the mixture of 2 (1.13 g, 3.0 mmol) and
2,6-lutidine (563 mg, 5.25 mmol) in acetone (4 mL) while maintaining the temperature between 12 and
18 °C. The reaction mixture was stirred at 15 °C for 23 h. Water (5 mL) was added drop-wise while
maintaining the temperature below 30 °C, and EtOAc (10 mL) was then added to the mixture. The
biphasic solution was transferred to a separating funnel for phase separation. The aqueous layer was
extracted with EtOAc (5 mL). The EtOAc layers were combined, washed successively with an aqueous
solution of 10% citric acid (5 mL × 2), an aqueous solution of 10% NaCl (5 mL), an aqueous solution of
5% NaHCO3 (5 mL × 2), and an aqueous solution of 10% NaCl (5 mL). They were then dried over
Na2SO4 and the filtrate was concentrated under reduced pressure. EtOH was added to the residue, and
the weight was adjusted to 7.2 g. The mixture was heated to 65 °C to dissolve solids. The solution was
cooled to 55 °C and seeded with 1. The solution was aged for 1 h at 50 °C, during which time the
product began to crystallize. After the slurry was cooled to 25 °C, n-heptane (11 mL) was added
drop-wise to the slurry at 25 °C followed by stirring for 1 h at 25 °C. The slurry was cooled to 3 °C and
then stirred for 2 h at 3 °C. The slurry was filtered, and the wet cake was washed with a mixed solvent
of EtOH (1.5 mL) and n-heptane (3 mL). The precipitate was dried in vacuo at 70 °C to give 888 mg
(66% yield) of 1 as a white solid. [α]
20
D -43.5 (c 1.0, DMSO). IR (KBr) cm-1
: 3495, 1744, 1514, 1488,
1454, 1467, 1411, 1372, 1340, 1266. 1H-NMR (CDCl3) δ: 1.27 (3H, t, J=7.0 Hz), 2.00 (1H, d, J=1.6
Hz), 3.46–3.54 (3H, m), 3.56–3.61 (2H, m), 3.72 (1H, d, J=2.1 Hz), 3.75 (3H, s), 3.82 (1H, d, J=15.5 Hz),
4.03 (1H, d, J=15.5 Hz), 4.11–4.22 (2H, m), 4.42 (2H, d, J=3.8 Hz), 4.69 (1H, d, J=7.4 Hz), 6.79–6.83
(2H, m), 6.97–7.02 (2H, m), 7.04–7.07 (2H, m), 7.15–7.22 (2H, m). 13C-NMR (CDCl3) δ: 14.2 (q), 36.1
(t), 55.4 (q), 64.4 (t), 66.4 (t), 69.6 (d), 73.4 (d), 73.8 (d), 75.7 (d), 100.8 (d), 114.1 (d×2), 114.4 (d), 122.7
(d), 128.0 (d), 129.2 (d×2), 130.0 (s), 131.1 (d), 133.4 (s), 155.2 (s), 155.4 (s), 157.8 (s). HRMS (ESI)
m/z: 466.2070 [M+NH4]
+
(Calcd for C23H32NO9: 466.2072)

6-O-Ethoxycarbonyl-2-[(4-methoxyphenyl)methyl]phenyl-β-D-glucopyranoside (1). Ethyl
chloroformate (21.6 g, 0.199 mol) was added drop-wise to the mixture of 2 (65.0 g, 0.173 mol),
2,6-lutidine (27.8 g, 0.259 mol) and pyridine (0.33 g, 4.2 mmol) in acetone (210 mL), maintaining the
temperature between -1 and 5 °C. The reaction mixture was stirred at 0 °C for 2 h. The reaction was
monitored by HPLC.15 Water (200 mL) was added drop-wise, maintaining the temperature below 30 °C,
and then EtOAc (220 mL) was added to the mixture. The biphasic solution was transferred to a
separating funnel for phase separation. The aqueous layer was extracted with EtOAc (140 mL). The
EtOAc layers were combined, washed successively with an aqueous solution of 10% citric acid (180 mL
× 2), an aqueous solution of 10% NaCl (66 g), an aqueous solution of 5% NaHCO3 (65 g × 2), and an aqueous solution of 10% NaCl (100 g), and then dried over Na2SO4 (65 g). After acetic acid (10 g,
0.167 mol) was added to the filtrate, the mixture was concentrated under reduced pressure. The residue
was dissolved in EtOH (660 mL) at 65 °C. The solution was concentrated under reduced pressure until
more than 330 mL distillate had been collected. EtOH was added to the residue, and the weight was
adjusted to 370 g. n-Heptane (120 mL) was added, and the resulting slurry was heated to 65 °C to
dissolve solids. The solution was cooled to 55 °C and seeded with 1. The solution was aged for 1 h at
50 °C, during which time the product began to crystallize. n-Heptane (480 mL) was added drop-wise to
the slurry, maintaining the temperature between 50 and 60 °C, and the slurry was stirred for 0.5 h at 55 °C.
The slurry was allowed to cool slowly over 2.5 h to 30 °C, then cooled to 3 °C, and then stirred for 1.5 h
at 3 °C. The slurry was filtered, and the wet cake was washed with a mixed solvent of EtOH (80 mL)
and n-heptane (180 mL). The precipitate was dried in vacuo at 70 °C to give 63.6 g (82% yield) of 1 as
a white solid.

REFERENCES (AND NOTES)

  1. W. N. Washburn, Expert Opin. Ther. Patents, 2009, 19, 1485.
  2. A. M. Pajor and E. M. Wright, J. Biol. Chem., 1992, 267, 3557.
  3. E. M. Wright, Am. J. Physiol. Renal Physiol., 2001, 280, F10.
  4. Y. Kanai, W. S. Lee, G. You, D. Brown, and M. A. Hediger, J. Clin. Invest., 1994, 93, 397.
  5. H. Fujikura, N. Fushimi, T. Nishimura, K. Tatani, and M. Isaji, PCT, WO 02/28872 (2002).
  6. H. Fujikura, N. Fushimi, T. Nishimura, K. Tatani, K. Katsuno, M. Hiratochi, Y. Tokutake, and M.
    Isaji, PCT, WO 01/688660 (2001).
  7. K. Katsuno, Y. Fujimori, Y. Takemura, M. Hiratochi, F. Itoh, Y. Komatsu, H. Fujikura, and M. Isaji,
    J. Pharmacol. Exp. Ther., 2007, 320, 323.
  8. M. Isaji, Curr. Opin. Investig. Drugs, 2007, 8, 285.
  9. S. Y. Lee, S. E. Rho, K. Y. Min, T. B. Kim, and H. K. Kim, J. Carbohydr. Chem., 2001, 20, 503.
  10. M. Yamaguchi, A. Horiguchi, A. Fukuda, and T. Minami, J. Chem. Soc., Perkin Trans. 1, 1990,
    1079.
  11. K. Ishihara, H. Kurihara, and H. Yamamoto, J. Org. Chem., 1993, 58, 3791.
  12. I. T. Akimova, A. V. Kaminsky, and V. I. Svistunova, Chem. Heterocycl. Compd., 2005, 41, 1374.
  13. B. N. Cook, S. Bhakta, T. Biegel, K. G. Bowman, J. I. Armstrong, S. Hemmerich, and C. R. Bertozzi,
    J. Am. Chem. Soc., 2000, 122, 8612.
  14. HPLC conditions: column, Inertsil ODS-3 (5 µm) 4.6 mm × 250 mm (GL Science Inc.); mobile
    phase, isocratic elution with acetonitrile / 0.02 M KH2PO4, pH 3 = 6/4; flow rate, 1.0 mL/min;
    column oven temperature, 40 °C; wave length, 225 nm; retention times, 5 = 16 min, α-anomer of 5 =18 min.
  15. HPLC conditions: column, Inertsil ODS-3 (5 µm) 4.6 mm × 250 mm (GL Science Inc.); mobile
    phase, gradient elution with 5 min 4/6 → 15 min 6/4 → 30 min 6/4 of acetonitrile/0.02 M KH2PO4,
    pH 3; flow rate, 1.0 mL/min; column oven temperature, 40 °C; wavelength, 225 nm; retention times,
    1 = 17 min, 2,6- and 4,6-bis-O-ethoxycarbonyl derivatives = 24 min, 3,6-bis-O-ethoxycarbonyl
    derivative = 25 min.

SYN

Synthesis 2024, 56, 906–943

Sergliflozin etabonate (16), also known as GW869682X, was developed collaboratively by GlaxoSmithKline and Kissei Pharmaceutical (Japan). Unfortunately, it did not pass phase III trials. It belongs to the class of sodium–glucose linked transporter 2 (SGLT2) inhibitors and acts as a prodrug of sergliflozin, with the ethyl carbonate group referred to as etabonate. When compared to phlorizin, sergliflozin etabonate demonstrated significantly higher activity against SGLT2 than SGLT1. The initial synthetic route for the preparation of sergliflozin was described and patented by Kissei Pharmaceutical Co., Ltd. This particular route for Oaryl-glycoside-type derivatives was registered in the United States under patent application number US6872706B2.73 The first reported synthesis of sergliflozin etabonate
(16), which involves six steps, can be found in the patents US6872706B2 73a and WO2001068660A1 (Scheme 48).73b Compound 271 was prepared in a high yield of 96% follow ing a literature procedure. The selective monodeacetylationof penta-O-acetyl-b-D-glucopyranose, compound 269, was
achieved using N,N-dimethylethylenediamine in THF, resulting in the formation of compound 270. Subsequently, a reaction with trichloroacetonitrile and potassium carbonate led to the synthesis of intermediate 271 in excellent yield. To prepare the aglycone intermediate 268, phenol (235) was condensed with 4-methoxybenzyl chloride (267) using LiOH under reflux conditions. Further,O-glycosyla
tion of compound 268 with 271 was accomplished using boron trifluoride–diethyl etherate (BF3·OEt2), yielding intermediate 272. Removal of the acetyl groups from intermediate 272 was carried out using NaOMe in methanol to obtain sergliflozin (16a) in a yield of 73%. Finally, sergliflozin etabonate (16) was obtained by reacting compound 16a with ethyl chloroformate and 2,6-lutidine, resulting in a yield of
66%. The overall yield of sergliflozin etabonate (16a) was calculated to be 11%. It is important to note that the trichloroimidation reaction used in the synthesis of trichloroacetimidate 271 is considered hazardous and is not recommended for commercial use due to the highly toxic reagent, trichloroaceto
nitrile. Additionally, the process poses challenges in effectively removing the unwanted by-product, trichloroacetamide, formed during the preparation.A recently published approach presents an alternative synthesis of sergliflozin etabonate (16) that avoids the use of a trichloroacetimidate intermediate (Scheme 49).74a The five-step synthesis of compound 16a commenced from
readily available anisole (273a). An efficient Friedel–Crafts reaction was performed on anisole (273a) using the acid chloride 273 in the presence of aluminum chloride in chlorobenzene, leading to formation of benzophenone 274. Notably, demethylation of 274 was also observed under these
conditions. Next, ketone group reduction was achieved us ing 10% Pd/C and ethanol under 0.3–0.4 MPa of H2, providing compound 268 in 88% yield and high purity. Subsequently, O-glycosylation of 268 with penta-acetylated com pound 269 was carried out using BF3·Et2O and triethylamine, resulting in the formation of 272 in 90% yield with a high b-selectivity (99:1).74b Deacetylation of compound 272 was performed using NaOMe in methanol, affording sergliflozin (16a) in 80% yield. Further reaction with
ethyl chloroformate in the presence of 2,6-lutidine resulted in sergliflozin etabonate (16). The overall yield of compound 16 was calculated to be 41%. This novel synthetic route offers a promising alternative to the traditional method and demonstrates improved efficiency in the preparation of sergliflozin etabonate (16)

(73) (a) Fujikura, H.; Fushimi, N. US6872706B2, 2005. (b) Fujikura, H.; Fushimi, N.; Nishimura, T.; Tatani, K.; Katsuno, K.; Hiratochi, M.; Tokutake, Y.; Isaji, M. WO2001068660A1, 2001.
(74) (a) Kobayashi, M.; Isawa, H.; Sonehara, J.; Kubota, M. Heterocycles 2016, 92, 1599. (b) Lee, Y. S.; Rho, S. E.; Min, K. Y.; Kim, T. B.; Kim, H. K. J. Carbohydr. Chem. 2001, 20, 503.

PatentSubmittedGranted
Progression Inhibitor For Disease Attributed To Abnormal Accumulation Of Liver Fat [US2008045466]2008-02-21
NOVEL SUBSTITUTED TETRAHYDRONAPHTHALENES, PROCESS FOR THE PREPARATION THEREOF AND THE USE THEREOF AS MEDICAMENTS [US2010249097]2010-09-30
(CARBOXYLALKYLENEPHENYL)PHENYLOXAMIDES, METHOD FOR THE PRODUCTION THEREOF AND USE OF SAME AS A MEDICAMENT [US2010261645]2010-10-14
(CYCLOPROPYLPHENYL)PHENYLOXAMIDES, METHOD FOR THE PRODUCTION THEREOF, AND USE OF SAME AS A MEDICAMENT [US8148375]2010-10-142012-04-03
Crystals of glucopyranosyloxybenzyl benzene derivative [US7371730]2005-06-022008-05-13
CERTAIN CRYSTALLINE DIPHENYLAZETIDINONE HYDRATES, PHARMACEUTICAL COMPOSITIONS THEREOF AND METHODS FOR THEIR USE [US8003636]2009-08-132011-08-23
NOVEL DIPHENYLAZETIDINONE SUBSTITUTED BY PIPERAZINE-1-SULFONIC ACID AND HAVING IMPROVED PHARMACOLOGICAL PROPERTIES [US2009264402]2009-10-22
Arylaminoaryl-alkyl-substituted imidazolidine-2,4-diones, process for preparing them, medicaments comprising these compounds, and their use [US7759366]2009-08-272010-07-20
Glucopyranosyloxybenzylbenzene derivatives and medicinal compositions containing the same [US2005065098]2005-03-24
Glucopyranosyloxybenzylbenzene derivatives and medicinal compositions containing the same [US6872706]2004-01-292005-03-29
PatentSubmittedGranted
PROGRESSION INHIBITOR FOR DISEASE ATTRIBUTED TO ABNORMAL ACCUMULATION OF LIVER FAT [US2009286751]2009-11-19
THERAPEUTIC USES OF SGLT2 INHIBITORS [US2011077212]2011-03-31
PHARMACEUTICAL COMPOSITION COMPRISING A SGLT2 INHIBITOR IN COMBINATION WITH A DPP-IV INHIBITOR [US2011098240]2011-04-28
Substituted imidazoline-2,4-diones, process for preparation thereof, medicaments comprising these compounds and use thereof [US2011112097]2011-05-12
Heterocycle-substituted imidazolidine-2,4-diones, process for preparation thereof, medicaments comprising them and use thereof [US2011046105]2011-02-24
Arylchalcogenoarylalkyl-substituted imidazolidine-2,4-diones, process for preparation thereof, medicaments comprising these compounds and use thereof [US2011046185]2011-02-24
Arylchalcogenoarylalkyl-substituted imidazolidine-2,4-diones, process for preparation thereof, medicaments comprising these compounds and use thereof [US2011053947]2011-03-03
Novel aromatic fluoroglycoside derivatives, pharmaceuticals comprising said compounds and the use thereof [US2011059910]2011-03-10
Novel phenyl-substituted imidazolidines, process for preparation thereof, medicaments comprising said compounds and use thereof [US2011178134]2011-07-21
HETEROCYCLIC COMPOUNDS, PROCESSES FOR THEIR PREPARATION, MEDICAMENTS COMPRISING THESE COMPOUNDS, AND THE USE THEREOF [US2011183998]2011-07-28
Systematic (IUPAC) name
2-(4-methoxybenzyl)phenyl 6-O-(ethoxycarbonyl)-β-D-glucopyranoside
Clinical data
Routes of
administration
Oral
Identifiers
CAS Number408504-26-7 
ATC codeNone
PubChemCID: 9824918
IUPHAR/BPS4587
ChemSpider21234810 
ChEMBLCHEMBL450044 
Chemical data
FormulaC23H28O9
Molecular mass448.463 g/mol

References

  1.  World Health Organization (2008). “International Nonproprietary Names for Pharmaceutical Substances (INN). Recommended International Nonproprietary Names: List 59” (PDF). WHO Drug Information22 (1): 66. Archived from the original (PDF) on February 19, 2009.
  2.  “Statement on a nonproprietary name adopted by the USAN council: Sergliflozin etabonate” (PDF). American Medical Association. Retrieved 2008-08-10.
  3.  Katsuno K, Fujimori Y, Takemura Y, et al. (January 2007). “Sergliflozin, a novel selective inhibitor of low-affinity sodium glucose cotransporter (SGLT2), validates the critical role of SGLT2 in renal glucose reabsorption and modulates plasma glucose level”J Pharmacol Exp Ther320 (1): 323–30. doi:10.1124/jpet.106.110296PMID 17050778S2CID 8306408.
  4.  “Prous Science: Molecule of the Month November 2007”. Archived from the original on 2007-11-05. Retrieved 2008-10-28.
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

……

////////// etabonate, Sergliflozin etabonate, Sergliflozin, PHASE 3, GW869682X, GSK, KISSEI, GW-869682; GW-869682X; KGT-1251
CCOC(=O)OCC1C(C(C(C(O1)OC2=CC=CC=C2CC3=CC=C(C=C3)OC)O)O)O
CCOC(=O)OCC1C(C(C(C(O1)Oc2ccccc2Cc3ccc(cc3)OC)O)O)O

Donidalorsen


Donidalorsen

CAS 2304692-48-4

분자량 Mw8672.64
화학식MfC296H435N83O151P20S15

ISIS 721744, ISIS-721744

FDA 8/21/2025, Dawnzera, To prevent attacks of hereditary angioedema

DNA, D((2′-O-(2-METHOXYETHYL))M5RU-SP-(2′-O-(2-METHOXYETHYL))RG-SP-(2′-O-(2-METHOXYETHYL))M5RC-(2′-O-(2-METHOXYETHYL))RA-(2′-O-(2-METHOXYETHYL))RA-SP-G-SP-T-SP-M5C-SP-T-SP-M5C-SP-T-SP-T-SP-G-SP-G-SP-M5C-SP-(2′-O-(2-METHOXYETHYL))RA-(2′-O-(2-METHOXYETHYL)

IngredientUNIICAS.
Donidalorsen sodiumY30VEG5PH12304701-45-7

Donidalorsen, sold under the brand name Dawnzera, is a medication used to prevent attacks of hereditary angioedema.[1] Donidalorsen is a prekallikrein-directed antisense oligonucleotide.[1] It is given by injection under the skin (subcutaneous).[1]

Donidalorsen was approved for medical use in the United States in August 2025.[2]

Donidalorsen is under investigation in clinical trial NCT05392114 to assess the long-term safety and efficacy of donidalorsen in the prophylactic treatment of hereditary angioedema (HAE)

Donidalorsen is an antisense oligonucleotide designed to reduce the production of prekallikrein (PKK). PKK plays an important role in the activation of inflammatory mediators associated with acute attacks of Hereditary angioedema (HAE).

https://ir.ionis.com/news-releases/news-release-details/dawnzeratm-donidalorsen-approved-us-first-and-only-rna-targeted

AWNZERA™ (donidalorsen) approved in the U.S. as first and only RNA-targeted prophylactic treatment for hereditary angioedema

August 21, 2025

View PDF

– DAWNZERA demonstrated significant and sustained HAE attack rate reduction and long-term disease control 

– Offers longest dosing option for HAE, with dosing every 4 or 8 weeks 

– Compelling profile supported by recently published switch data 

– Ionis’ second independent launch in just nine months, with potential for two additional launches next year 

– Ionis to host webcast today at 12:15pm ET 

CARLSBAD, Calif.–(BUSINESS WIRE)–Aug. 21, 2025– Ionis Pharmaceuticals, Inc. (Nasdaq: IONS) announced today that the U.S. Food and Drug Administration (FDA) has approved DAWNZERA™ (donidalorsen) for prophylaxis to prevent attacks of hereditary angioedema (HAE) in adult and pediatric patients 12 years of age and older. DAWNZERA is the first and only RNA-targeted medicine approved for HAE, designed to target plasma prekallikrein (PKK), a key protein that activates inflammatory mediators associated with acute attacks of HAE. DAWNZERA 80mg is self-administered via subcutaneous autoinjector once every four (Q4W) or eight weeks (Q8W).

This press release features multimedia. View the full release here: https://www.businesswire.com/news/home/20250818615141/en/

DAWNZERA (donidalorsen) logo

DAWNZERA (donidalorsen) logo

HAE is a rare and potentially life-threatening genetic condition that involves recurrent attacks of severe swelling (angioedema) in various parts of the body, including the hands, feet, genitals, stomach, face and/or throat. HAE is estimated to affect approximately 7,000 people in the U.S.

“DAWNZERA represents a significant advance for people living with HAE who need improved treatment options. With strong and durable efficacy, convenient administration and the longest dosing option available, we believe DAWNZERA will be the prophylactic treatment of choice for many people living with HAE. Importantly, the recently published switch data empowers patients and physicians with a roadmap for switching to DAWNZERA from other prophylactic therapies,” said Brett P. Monia, Ph.D., chief executive officer, Ionis. “At Ionis, we are dedicated to turning groundbreaking science into life-changing medicines. With the early success of our first independent launch of TRYNGOLZA® for familial chylomicronemia syndrome (FCS), and now with DAWNZERA, our second independent medicine approved in less than nine months, we are proudly delivering on that vision. To the patients, families, advocacy partners and investigators who helped make this moment a reality, we express our deepest gratitude.”

The approval of DAWNZERA was based on positive results from the Phase 3 global, multicenter, randomized, double-blind, placebo-controlled OASIS-HAE study in patients with HAE. The study met its primary endpoint, with DAWNZERA Q4W significantly reducing monthly HAE attack rate by 81% compared to placebo over 24 weeks. Mean attack rate reduction increased to 87% when measured from the second dose, a key secondary endpoint. Additionally, DAWNZERA Q4W reduced moderate-to-severe HAE attacks by ~90% over 24 weeks when measured from the second dose.

These results are bolstered by the ongoing OASISplus open-label extension (OLE) study, in which DAWNZERA Q8W had a similar effect as Q4W over time. DAWNZERA demonstrated 94% total mean attack rate reduction from baseline across both dosing groups after one year in the OLE.

The OASISplus study also includes a switch cohort evaluating DAWNZERA Q4W in patients previously treated with lanadelumab, C1-esterase inhibitor or berotralstat for at least 12 weeks. Switching to DAWNZERA reduced mean HAE attack rate by 62% from prior prophylactic treatment over 16 weeks, with no mean increase in breakthrough attacks observed during the switch. A total of 84% of patients surveyed preferred DAWNZERA over their prior prophylactic treatment, citing better disease control, less time to administer and less injection site pain or reactions.

Across clinical studies, DAWNZERA demonstrated a favorable safety and tolerability profile. The most common adverse reactions (incidence ≥ 5%) were injection site reactions, upper respiratory tract infection, urinary tract infection and abdominal discomfort.

“As the first FDA-approved RNA-targeted therapy for HAE, DAWNZERA represents a welcome advance in therapeutic options for preventing attacks. Today’s approval gives people living with HAE and their physicians another important choice for aligning treatment with individual needs,” said Anthony J. Castaldo, CEO & chairman of the board, U.S. Hereditary Angioedema Association (HAEA) and Hereditary Angioedema International (HAEi).

“People living with HAE manage this condition for all their lives, and many continue to face unpredictable, painful and dangerous breakthrough attacks even with current treatments. Durable efficacy is essential in maintaining long-term disease control,” said Marc Riedl, M.D., M.S., clinical director, U.S. HAEA Angioedema Center; University of California, San Diego; OASIS-HAE and OASISplus trial investigator. “DAWNZERA is positioned to help meet patient needs, providing substantial and sustained reduction of HAE attacks, continued improvement over time and reduced burden of treatment.”

DAWNZERA will be available in the U.S. in the coming days.

Ionis is committed to helping people access the medicines they are prescribed and will offer a suite of services designed to meet the unique needs of the HAE community through Ionis Every Step™. As part of Ionis Every Step, patients and healthcare providers will have access to a wide range of support and resources including dedicated support from a Patient Education Manager, assistance with the insurance approval process, information on affordability programs, access to the DAWNZERA Direct digital companion and other ongoing services and resources to help patients stay on track. Visit DAWNZERA.com for more information.

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

……

References

  1.  “DAWNZERA (donidalorsen) injection, for subcutaneous use” (PDF). Highlights of Prescribing Information. Ionis Pharmaceuticals, Inc.
  2.  “Dawnzera (donidalorsen) approved in the U.S. as first and only RNA-targeted prophylactic treatment for hereditary angioedema” (Press release). Ionis Pharmaceuticals, Inc. 21 August 2025. Retrieved 22 August 2025 – via Business Wire.
  3.  “Donidalorsen: An Investigational RNA-targeted Medicine” (PDF). Ionis Pharmaceuticals, Inc.
  4.  Farkas H, Balla Z (March 2024). “Kallikrein inhibitors for angioedema: the progress of preclinical and early phase studies”. Expert Opinion on Investigational Drugs33 (3): 191–200. doi:10.1080/13543784.2024.2320700PMID 38366937.
  5.  “Dawnzera: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 22 August 2025.
  6.  World Health Organization (2021). “International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 86”. WHO Drug Information35 (3). hdl:10665/346562.

Further reading

  • Raja A, Shuja MH, Raja S, Qammar A, Kumar S, Khurram L, et al. (December 2024). “Efficacy and safety of Donidalorsen in Hereditary Angioedema with C1 inhibitor deficiency: a systematic review and a meta analysis”. Archives of Dermatological Research317 (1): 110. doi:10.1007/s00403-024-03652-3PMID 39666085.
  • Clinical trial number NCT05139810 for “OASIS-HAE: A Study to Evaluate the Safety and Efficacy of Donidalorsen (ISIS 721744 or IONIS-PKK-LRx) in Participants With Hereditary Angioedema (HAE)” at ClinicalTrials.gov
Clinical data
Trade namesDawnzera
Other namesISIS 721744, ISIS-721744
AHFS/Drugs.comDawnzera
License dataUS DailyMedDonidalorsen
Routes of
administration
Subcutaneous
ATC codeNone
Legal status
Legal statusUS: ℞-only[1]
Identifiers
CAS Number2304692-48-42304701-45-7
DrugBankDB18751DBSALT003520
UNIIZD4D8M32TLY30VEG5PH1

//////////Donidalorsen, FDA 2025, APPROVALS 2025, Dawnzera, ISIS-721744 FREE ACID, ISIS 721744

Baxdrostat


Baxdrostat

  • NF3P9Z8J5Y
  • CIN-107
  • RO6836191
  • 363.5 g/mol

WeightAverage: 363.461
Monoisotopic: 363.194677057

Chemical FormulaC22H25N3O2

N-[(8R)-4-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)-5,6,7,8-tetrahydroisoquinolin-8-yl]propanamide

Baxdrostat is an investigational drug that is being evaluated for the treatment of hypertension.[1] It is an aldosterone synthase inhibitor.[2][3]

Baxdrostat is under investigation in clinical trial NCT06344104 (A Phase III Study to Investigate the Efficacy and Safety of Baxdrostat in Asian Participants With Uncontrolled Hypertension on Two or More Medications Including Participants With Resistant Hypertension).

LIT

US9353081,

https://patentscope.wipo.int/search/en/detail.jsf?docId=US76841362&_cid=P21-MEZ3MG-55484-1

Example 3-1

(+)-(R)—N-(4-(1-Methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-5,6,7,8-tetrahydroisoquinolin-8-yl)propionamide

   In analogy to the procedures described for the preparation of intermediate A-2 [E] and for the preparation of intermediate B-1, Suzuki reaction of (+)-(R)-4-bromo-5,6,7,8-tetrahydroisoquinolin-8-amine (intermediate B-3b) with 1-methyl-6-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-3,4-dihydro-1H-quinolin-2-one (intermediate A-1) gave (R)-6-(8-amino-5,6,7,8-tetrahydroisoquinolin-4-yl)-1-methyl-3,4-dihydroquinolin-2(1H)-one and after subsequent reaction with propionyl chloride the title compound as colorless solid. MS: 364.2 (M+H +).

Pat

CN 117247371 

https://patentscope.wipo.int/search/en/detail.jsf?docId=CN418385740&_cid=P12-MEZHY3-66430-1

Example 1
        
        Step A
        Dissolve 4-bromo-6,7-dihydroisoquinolin-8(5H)-one (1.56 g, 6.9 mmol) and (S)-tert-butylsulfenamide (2.51 g, 20.7 mmol) in 20 mL of tetrahydrofuran. Add ethyl titanate (10.08 mL, 48.28 mmol). Heat to 65°C and stir for 48 hours. Cool to room temperature, add ethyl acetate and water, stir for 15 minutes, and remove the resulting solid by filtration. Separate the liquids, dry the organic phase over anhydrous sodium sulfate, filter, and evaporate to dryness under reduced pressure to obtain the crude product (S,Z)-N-(4-bromo-6,7-dihydroisoquinolin-8(5H)-tert-butylsulfenimide), which is used directly in the next step.
        Step B
        Compound (S,Z)-N-(4-bromo-6,7-dihydroisoquinoline-8(5H)-tert-butylsulfonyl imide) (1.98 g, 6 mmol) was dissolved in 15 mL of tetrahydrofuran and cooled to -45°C. Sodium borohydride (0.34 g, 9.0 mmol) was added, and the mixture was allowed to return to room temperature and stirred for 18 hours. The mixture was quenched with ice water and extracted with dichloromethane. The resulting organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to obtain compound (S)-N-(4-bromo-6,7-dihydroisoquinoline-8(5H))-tert-butylsulfonyl imide (755 mg, 38% yield). LC/MS (ESI): m/z = 331.2 [M+H] + .
        Step C
        To a mixture of (S)-N-(4-bromo-6,7-dihydroisoquinoline-8(5H))-tert-butylsulfonimide (0.66 g, 2 mmol), pinacol diboronate (1.05 g, 2.1 mmol), and AcOK (0.578 g, 6 mmol) in toluene (10 mL) was added Pd(dppf)Cl 2 (0.144 g, 0.2 mmol). The mixture was degassed and stirred at 130 ° C for 3 hours. The reaction mixture was filtered and concentrated to give a residue. EtOAc (15 mL) and water (10 mL) were added to the residue. The organic phase was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 ) and eluted with 30-40% ethyl acetate in petroleum ether to afford (S)-N-tert-butylsulfonamido-6,7-dihydroisoquinolin-8(5H)-4-boronic acid pinacol ester (0.45 g, 60% yield). LC/MS (ESI): m/z = 378.3 [M+H] + .
        Step D
        To a reaction flask, add 6-bromo-1-methyl-3,4-dihydroquinolin-2(1H)-one (0.29 g, 1.2 mmol), (S)-N-tert-butylsulfonamido-6,7-dihydroisoquinolin-8(5H)-4-boronic acid pinacol ester (0.42 g, 1.26 mmol), bistriphenylphosphine palladium dichloride (84 mg, 0.12 mmol), cuprous iodide (38 mg, 0.2 mmol), triethylamine (1.01 g, 10.0 mmol), and 15 mL of N,N-dimethylformamide. The atmosphere was purged with nitrogen three times and the reaction was stirred at 90°C overnight. After cooling to room temperature, the reaction mixture was diluted with ethyl acetate and water, and extracted with ethyl acetate. The resulting organic phase was washed with water and saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to afford (S)-2-methyl-N-((R)-4-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-5,6,7,8-tetrahydroisoquinolin-8-yl)tert-butylsulfonimide (0.37 g, 74% yield) as a yellow solid. LC/MS (ESI): m/z = 411.5 [M+H] + .
        Step E
        Compound (S)-2-methyl-N-((R)-4-(1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-5,6,7,8-tetrahydroisoquinolin-8-yl)tert-butylsulfonimide (0.33 g, 0.80 mmol) was dissolved in 1 mL of dichloromethane, and 1 mL of trifluoroacetic acid was added. The mixture was stirred and reacted for 1 hour. The reaction solution was concentrated under reduced pressure. The residue was purified by reverse preparative column chromatography to obtain compound (R)-6-(8-amino-5,6,7,8-tetrahydroisoquinolin-4-yl)-1-methyl-3,4-dihydroquinolin-2(1H)-one (0.24 g, 97% yield). LC/MS (ESI): m/z = 307.1 [M+H] + .
        Step F
        To a reaction flask, add (R)-6-(8-amino-5,6,7,8-tetrahydroisoquinolin-4-yl)-1-methyl-3,4-dihydroquinolin-2(1H)-one (100 mg, 0.33 mmol), triethylamine (51 mg, 0.5 mmol), and 4 ml of tetrahydrofuran. After cooling in an ice-water bath, slowly add a solution of propionyl chloride (46.25 mg, 0.5 mmol) in 0.5 ml of tetrahydrofuran dropwise. Stirring is continued for 4 hours after addition. The reaction mixture is quenched with methanol and evaporated to dryness under reduced pressure. The residue is purified by column chromatography to obtain the target compound, Baxdrostat (46 mg, 38% yield). LC/MS(ESI):m/z=363.1[M+H]+.H NMR(400MHz, CDCl3)ppm 1.22(t,3H)1.79(s,3H)2.07(s,1H)2.28(q,2H)2.43-2.68(m,2H)2.71(t,2H)2.82-3.12(m,2H) 3.40(s,3H)5.34(d,1H)5.78(d,1H)7.05(d,1H)7.09(s,1H)7.17(d,1H)8.28(s,1H)8.49(s,1H)
        Example 2
        

        Step A
        Compound (S)-N-(4-bromo-6,7-dihydroisoquinolin-8(5H))-tert-butylsulfonylimide (1.65 g, 5 mmol) was dissolved in 20 mL of dichloromethane, and 20 mL of trifluoroacetic acid was added. The mixture was stirred and reacted for 1 hour. The reaction solution was concentrated under reduced pressure. The residue was purified by reverse-phase preparative column chromatography to obtain compound (R)-4-bromo-5,6,7,8-tetrahydroisoquinolin-8-amine (1.07 g, 94% yield). LC/MS (ESI): m/z = 226.0 [M+H] + .
        Step B
        To a mixture of (R)-4-bromo-5,6,7,8-tetrahydroisoquinolin-8-amine (0.86 g, 3.8 mmol), pinacol diboron (2 g, 4 mmol), AcOK (1.10 g, 11.4 mmol) in toluene (10 mL) was added Pd(dppf)Cl 2 (0.27 g, 0.38 mmol). The mixture was degassed and stirred at 130 ° C for 3 hours. The reaction mixture was filtered and concentrated to give a residue. EtOAc (10 mL) and water (10 mL) were added to the residue. The organic phase was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 ) and eluted with 30-40% ethyl acetate in petroleum ether to afford (R)-8-amino-5,6,7,8-tetrahydroisoquinoline-4-boronic acid pinacol ester (0.68 g, 65% yield). LC/MS (ESI): m/z = 274.1 [M+H] + .
        Step C
        To a reaction flask, add 6-bromo-1-methyl-3,4-dihydroquinolin-2(1H)-one (0.72 g, 3.0 mmol), (R)-8-amino-5,6,7,8-tetrahydroisoquinolin-4-boronic acid pinacol ester (0.99 g, 3.6 mmol), bistriphenylphosphine palladium dichloride (210 mg, 0.3 mmol), and potassium phosphate monohydrate (204 mg, 0.9 mmol). Dissolve the mixture in dioxane and water (9:1, 30 mL). Replace the atmosphere with nitrogen three times and allow the mixture to react overnight at 90°C with stirring. Cool to room temperature, dilute the reaction solution with ethyl acetate and water, and extract with ethyl acetate. The resulting organic phase is then washed with water and saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to obtain (R)-6-(8-amino-5,6,7,8-tetrahydroisoquinolin-4-yl)-1-methyl-3,4-dihydroquinolin-2(1H)-one (0.81 g, 88% yield). LC/MS (ESI): m/z = 307.1 [M+H] + . The target compound, Baxdrostat, was then prepared using a method similar to the last step in Example 1.
        Example 3
        
        Step A
        4-Bromo-6,7-dihydroisoquinolin-8(5H)-one (1.88 g, 6.9 mmol) and (S)-tert-butylsulfenamide (2.51 g, 20.7 mmol) were dissolved in 20 mL of tetrahydrofuran. Ethyl titanate (10.08 mL, 48.28 mmol) was added and the mixture was heated to 65°C with stirring for 48 hours. After cooling to room temperature, ethyl acetate and water were added and stirred for 15 minutes. The resulting solid was removed by filtration. The organic phase was separated and dried over anhydrous sodium sulfate, filtered, and evaporated to dryness under reduced pressure to obtain the crude product (S,Z)-N-(4-bromo-6,7-dihydroisoquinolin-8(5H)-tert-butylsulfenimide), which was used directly in the next step. LC/MS (ESI): m/z = 376.2 [M+H] + .
        Step B
        Compound (S,Z)-N-(4-iodo-6,7-dihydroisoquinoline-8(5H)-tert-butylsulfonyl imide) (2.26 g, 6 mmol) was dissolved in 15 mL of tetrahydrofuran and cooled to -45°C. Sodium borohydride (0.36 g, 9.0 mmol) was added, and the mixture was allowed to return to room temperature and stirred for 18 hours. The mixture was quenched with ice water and extracted with dichloromethane. The resulting organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to obtain compound (S)-N-(4-iodo-6,7-dihydroisoquinoline-8(5H))-tert-butylsulfonyl imide (1.04 g, 46% yield). LC/MS (ESI): m/z = 378.0 [M+H] + .
        Step C
        To a mixture of (S)-N-(4-iodo-6,7-dihydroisoquinoline-8(5H))-tert-butylsulfonimide (0.76 g, 2 mmol), pinacol diboronate (1.05 g, 2.1 mmol), and AcOK (0.578 g, 6 mmol) in toluene (10 mL) was added Pd(dppf)Cl 2 (0.144 g, 0.2 mmol). The mixture was degassed and stirred at 130 ° C for 3 hours. The reaction mixture was filtered and concentrated to give a residue. EtOAc (15 mL) and water (10 mL) were added to the residue. The organic phase was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 ) and eluted with 30-40% ethyl acetate in petroleum ether to afford (S)-N-tert-butylsulfonamido-6,7-dihydroisoquinolin-8(5H)-4-boronic acid pinacol ester (0.51 g, 68% yield). LC/MS (ESI): m/z = 378.2 [M+H] + .
        The next three steps were carried out in the same manner as in Example 1 to prepare the target compound Baxdrostat.

LIT

https://medicalxpress.com/news/2025-08-stubborn-high-blood-pressure-experimental.html

A new treatment has been shown to significantly lower blood pressure in people whose levels stay dangerously high, despite taking several existing medicines, according to the results of a Phase III clinical trial led by a UCL Professor. Globally, around 1.3 billion people have high blood pressure (hypertension), and in around half of cases the condition is uncontrolled or treatment resistant. These individuals face a much greater risk of heart attack, stroke, kidney disease, and early death. In the UK the number of people with hypertension is around 14 million.

The international BaxHTN trial, led by Professor Bryan Williams (UCL Institute of Cardiovascular Science), assessed the new drug baxdrostat—which is taken as a tablet—with participation from nearly 800 patients across 214 clinics worldwide.

Results were presented at the European Society of Cardiology (ESC) Congress 2025 in Madrid and were simultaneously published in the New England Journal of Medicine.

The trial results showed that, after 12 weeks, patients taking baxdrostat (1 mg or 2 mg once daily in pill form) saw their blood pressure fall by around 9-10 mmHg more than placebo—a reduction large enough to cut cardiovascular risk. About four in 10 patients reached healthy blood pressure levels, compared with fewer than two in 10 on placebo.

Principal Investigator, Professor Williams, who is presenting the results at ESC, said, “Achieving a nearly 10 mmHg reduction in systolic blood pressure with baxdrostat in the BaxHTN Phase III trial is exciting, as this level of reduction is linked to substantially lower risk of heart attack, stroke, heart failure and kidney disease.”

How baxdrostat works

Blood pressure is strongly influenced by a hormone called aldosterone, which helps the kidneys regulate salt and water balance.

Some people produce too much aldosterone, causing the body to hold onto salt and water. This aldosterone dysregulation pushes blood pressure up and makes it very difficult to control.

Addressing aldosterone dysregulation has been a key effort in research over many decades, but it has been so far difficult to achieve.

Baxdrostat works by blocking aldosterone production, directly addressing this driver of high blood pressure (hypertension).

Professor Williams, Chair of Medicine at UCL, said, “These findings are an important advance in treatment and in our understanding of the cause of difficult-to-control blood pressure.

“Around half of people treated for hypertension do not have it controlled, however this is a conservative estimate and the number is likely higher, especially as the target blood pressure we try to reach is now much lower than it was previously.

“In patients with uncontrolled or resistant hypertension, the addition of baxdrostat 1mg or 2mg once daily to background antihypertensive therapy led to clinically meaningful reductions in systolic blood pressure, which persisted for up to 32 weeks with no unanticipated safety findings.

“This suggests that aldosterone is playing an important role in causing difficult to control blood pressure in millions of patients and offers hope for more effective treatment in the future.”

Historically, higher-income Western countries were reported to have far higher levels of hypertension. However, largely due to changing diets (adding less salt to food), the numbers of people living with the condition is now far higher in Eastern and lower-income countries. More than half of those affected live in Asia, including 226 million people in China and 199 million in India.

Professor Williams added, “The results suggest that this drug could potentially help up to half a billion people globally—and as many as 10 million people in the UK alone, especially at the new target level for optimal blood pressure control.”

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

……

Identifiers
IUPAC name
CAS Number1428652-17-8
PubChem CID71535962
IUPHAR/BPS12362
ChemSpider76804781
UNIINF3P9Z8J5Y
KEGGD12789
ChEMBLChEMBL4113975
Chemical and physical data
FormulaC22H25N3O2
Molar mass363.461 g·mol−1
3D model (JSmol)Interactive image
SMILES
InChI

PATENTS

References

  1.  “Baxdrostat – CinCor Pharma”AdisInsight. Springer Nature Switzerland AG.
  2.  Dogra S, Shah S, Gitzel L, Pusukur B, Sood A, Vyas AV, Gupta R (July 2023). “Baxdrostat: A Novel Aldosterone Synthase Inhibitor for Treatment Resistant Hypertension”. Current Problems in Cardiology48 (11): 101918. doi:10.1016/j.cpcardiol.2023.101918PMID 37399857S2CID 259320969.
  3.  Awosika A, Cho Y, Bose U, Omole AE, Adabanya U (October 2023). “Evaluating phase II results of Baxdrostat, an aldosterone synthase inhibitor for hypertension”. Expert Opinion on Investigational Drugs32 (11): 985–995. doi:10.1080/13543784.2023.2276755PMID 37883217S2CID 264517675.

/////Baxdrostat, PHASE 3, NF3P9Z8J5Y, CIN 107, RO 6836191,

Linaprazan


Linaprazan

CHINA 2024, APPROVALS 2024, AstraZeneca, CINCLUS, GERD, linaprazan glurate, for the treatment of moderate to severe GERD,

8-[(2,6-dimethylphenyl)methylamino]-N-(2-hydroxyethyl)-2,3-dimethylimidazo[1,2-a]pyridine-6-carboxamide

Chemical structure of linaprazan glurate CAS No.: 1228559-81-6 , X842

Molecular formulaC26H32N4O5
Molecular weight480.556086540222
Accurate quality480.237

5-[2-[[8-[(2,6-dimethylphenyl)methylamino]-2,3-dimethylimidazo[1,2-a]pyridine-6-carbonyl]amino]ethoxy]-5-oxopentanoic acid

  • OriginatorAstraZeneca
  • DeveloperCinclus Pharma; Jiangsu Sinorda Biomedicine Co., Ltd; Shanghai Pharmaceutical Group
  • Class2 ring heterocyclic compounds; Amines; Aminopyridines; Anti-inflammatories; Antibacterials; Antiulcers; Glutarates; Imidazoles; Pentanoic acids; Pyridines; Small molecules; Toluenes
  • Mechanism of ActionPotassium-competitive acid blockers
  • RegisteredReflux oesophagitis
  • Phase IIDuodenal ulcer; Erosive oesophagitis; Helicobacter infections
  • Phase IGastro-oesophageal reflux
  • 28 Aug 2025No recent reports of development identified for phase-I development in Gastro-oesophageal-reflux(In volunteers) in Sweden (PO, Tablet)
  • 29 Jun 2025Cinclus Pharma Holding plans a phase III trial for Gastro-oesophageal-reflux in the US, Bulgaria, Czech Republic, Georgia, Germany, Hungary, Poland (PO) (NCT07037875)
  • 13 Jun 2025Cinclus Pharma secures EMA and FDA pediatric study waivers for linaprazan glurate in H. pylori infection

Linaprazan is a lipophilic, weak base with potassium-competitive acid blocking (P-CAB) activity. Linaprazan concentrates highly in the gastric parietal cell canaliculus and on entering this acidic environment is instantly protonated and binds competitively and reversibly to the potassium binding site of the proton pump hydrogen-potassium adenosine triphosphatase (H+/K+ ATPase), thereby inhibiting the pump’s activity and the parietal cell secretion of H+ ions into the gastric lumen, the final step in gastric acid production.

Linaprazan is an experimental drug for the treatment of gastroesophageal reflux disease (GERD). Unlike the proton-pump inhibitors (PPIs) which are typically used to treat GERD, linaprazan is a potassium-competitive acid blocker (P-CAB).[1][2] Linaprazan was developed by AstraZeneca, but it was not successful in clinical trials.[3]

The drug was then licensed to Cinclus Pharma,[4] which is now investigating linaprazan glurate, a prodrug of linaprazan which is expected to have a longer biological half-life than linaprazan itself.[4]

Linaprazan glurate inhibits exogenously or endogenously stimulated gastric acid secretion. Linaprazan glurate exhibits several favorable properties, such as rapid onset of action, high in vivo potency, and/or prolonged duration of action. Linaprazan glurate is useful in the research of gastrointestinal inflammatory diseases and peptic ulcer disease (disclosed in patent WO2010063876A1).

SYN

WO2010063876

https://patentscope.wipo.int/search/en/WO2010063876

Examples

Example 1

Preparation of 5- {2-[( {8-[(2,6-dimethylbenzyl)amino]-2,3-dimethylimidazo[ 1 ,2-a]pyridin-6-yl}carbonyl)amino]ethoxy}-5-oxopentanoic acid

2,3-dimethyl-8-(2,6-dimethylbenzylamino)-N-hydroxyethyl-imidazo[l,2-a]pyridi-ne-6-carboxamide (obtained using the process according to WO02/20523) (2.0 g,

5.46 mmol) and glutaric anhydride (0.95 g, 8.33 mmol) was added to DMF (10 ml). The mixture was heated to 80 0C and stirred 16 h at this temperature.

Acetone (20 ml) was added to the reaction mixture whereby the product started to crystallize. The mixture was cooled to room temperature. After 4 h the product was filtered off and washed with acetone (20 ml). 2.25 g (86%) of the title compound was obtained. The structure of the compound was confirmed with 1H- NMR spectrum.

1H-NMR (300 MHz, DMSO): δ 1.73 (m, 2H), 2.2-2.4 (m, 16H), 3.52 (m,2H), 4.18 (t, 2H), 4.36 (d, 2H), 4.99 (t, IH), 6.67 (s, IH), 7.0-7.2 (m, 3H), 8.04 (s, IH), 8.56 (t, IH), 12.10 (bs, IH).

SYN

US6900324B2.

https://patentscope.wipo.int/search/en/detail.jsf?docId=US40374322&_cid=P12-MEXO1E-18626-1

Example 1.16

       Synthesis of 8-[(216-dimethylbenzyl)amino]-N-(2-hydroxyethyl)-2,3-dimethylimidazo[1,2-a]pyridine-6-carboxamide
       A reactor was charged with isopropyl 8-[(2,6-dimethylbenzyl)amino]-2,3-dimethylimidazo[1,2-a]pyridine-6-carboxylate (11.30 kg, 1 equiv., 27.02 mol) and THF (45 L), ethanolamine (18.97 kg, 11 equiv., 309.2 mol) was added at about 20° C. The suspension was heated to about 100° C. Some solvent was distilled off and then THF (35 L) was added and the distillation was continued. The procedure of adding THF and distilling it off was repeated until complete conversion. To the suspension ethanol (140L) was added and the suspension was heated to reflux. To obtain a clear solution additional ethanol (13L) was added. The hot solution was filtered and then cooled. The white solid was filtered off, washed with ethanol and dried to yield the product as a white powder. (8271 g).
       2. Preparation of Starting Materials

Example 2.1

       Synthesis of 6-amino-5-nitro-nicotinamide
       100 g of 6-hydroxy-5-nitro-nicotinic acid (0.54 mol; HPLC>98% area) was suspended in toluene (750 mL). DMF (1 mL, 0.013 mol, 0.024 equiv.) was added and the mixture was heated to 110° C. (inner temperature). Thionylchloride (99 mL, 2.5 equiv.) was added over 120 min. Heating was continued for 4 h at 110° C. The reaction mixture was concentrated to half the volume (400 mL of solvent were distilled off), and toluene (400 mL) was added.
       This procedure was repeated once again (410 mL of toluene were distilled off and fresh toluene (410 mL) was added again). The solution was then cooled to 20° C. and slowly added to aqueous ammonia (25%, 440 mL, 12 equiv.) over 40 min. Precipitation started immediately. During the addition the temperature was maintained below 15° C. After the addition had been completed the reaction mixture was allowed to warm up to room temperature and stirring was continued for 16 h. The solid was filtered off, washed with water (500 mL), ethanol (250 mL), TBME (250 mL) and dried (50-10 mbar, 40° C. bath temperature, 16 h) to yield 91.3 g of the title compound (0.501 mol, 87%).

Example 2.2

       Synthesis of 5,6-diamino-nicotinamide
       44.5 g of 6-amino-5-nitro-nicotinamide (0.24 mol; HPLC: 93% area) were suspended in methanol/water 1:1 (500 mL), 5.0 g of catalyst [Pd(4%)-Ru(1%)/C paste (62% H2O type: 485; Johnson Matthey); type: 485; Johnson Matthey] was added. Hydrogenation was carried out at 5 bar and 30° C. for 5 h. After completion the catalyst was filtered off and washed with methanol/water 1/1 (50 mL). 480 mL of the solvent was distilled off. The resulting suspension was cooled to 20° C. and filtered off. The solid was washed with methanol (20 mL) and TBME (30 mL). After drying (200-10 mbar; 40° C. bath temperature, 16 h) 27.3 g of the title compound (0.18 mol, 73%) were obtained.

Example 2.3

       Synthesis of 5,6-diamino-nicotinamide
       42.3 g of 6-amino-5-nitro-nicotinamide (0.23 mol, HPLC: 93% area) was suspended in methanol/water 1:1 (500 mL). 5.2 g of catalyst [Pd(5%)/C (57.8% H2O); type: 39, Johnson Matthey] was added. Hydrogenation was carried out at 5 bar and 30° C. for 4 h. After completion the catalyst was filtered off and washed with methanol/water 1/1 (100 mL). 550 mL of the solvent was distilled off. The resulting suspension was cooled to 20° C. and filtered off. The solid was washed with methanol (20 mL) and TBME (30 mL). After drying (200-10 mbar; 40° C. bath temperature, 16 b) 28.5 g of the title compound (0.18 mol, 78%) was obtained

SYN

European Journal of Medicinal Chemistry 291 (2025) 117643

Linaprazan is a potassium-competitive acid blocker (P-CAB) initially developed by AstraZeneca between 2001 and 2005 for treating gastroesophageal reflux disease (GERD). Subsequently, Cinclus Pharma ac
quired the rights to linaprazan and developed linaprazan glurate. In 2024, the NMPA approved linaprazan glurate for the treatment of moderate to severe GERD, marking Cinclus Pharma’s first marketing approval in China. Linaprazan glurate is a P-CAB that inhibits gastric acid secretion by reversibly blocking the potassium-binding site of the gastric H+/K +-ATPase enzyme, leading to rapid and sustained acid suppression [94]. Clinical efficacy was demonstrated in Phase III trials NCT04567810), showing superior acid suppression and symptom relief compared to PPIs in GERD patients. Regarding toxicity, linaprazan was generally well tolerated in clinical studies. However, some issues were
noted, such as elevated liver transaminases in a few patients, which were addressed in the development of linaprazan glurate by achieving lower peak plasma concentrations (Cmax) to minimize liver load 95,96]. The synthetic route of Linaprazan, shown in Scheme 22 [97], initiates with condensative Cyclization between Lina-001 and Lina-002 to yield Lina-003. This intermediate undergoes nucleophilic substitution with Lina-004 under basic conditions to generate Lina-005. Final thermolytic amidation of Lina-005 at 100 DEG CENT affords Linaprazan

[95] C. Scarpignato, R.H. Hunt, Potassium-competitive acid blockers: current clinical use and future developments, Curr. Gastroenterol. Rep. 26 (2024) 273–293.
[96] J.F. Willart, M. Durand, L.E. Briggner, A. Marx, F. Dan`ede, M. Descamps, Solid-state amorphization of linaprazan by mechanical milling and evidence of polymorphism, J Pharm Sci 102 (2013) 2214–2220.
[97] B. Elman, S. Erback, E. Thiemermann, Process for Preparing a Substituted Imidazopyridine Compound, 2002. US6900324B2.

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

……

References

  1.  Rawla P, Sunkara T, Ofosu A, Gaduputi V (December 2018). “Potassium-competitive acid blockers – are they the next generation of proton pump inhibitors?”World Journal of Gastrointestinal Pharmacology and Therapeutics9 (7): 63–68. doi:10.4292/wjgpt.v9.i7.63PMC 6305499PMID 30595950.
  2.  “Linaprazan”Inxight Drugs. National Center for Advancing Translational Sciences.
  3.  Tong A (4 March 2020). “Can reformulation of an AstraZeneca castoff rival Takeda’s new heartburn drug? Here’s a $26M bet on yes”endpts.com.
  4.  “Linaprazan glurate”. Cinclus Pharma.
Clinical data
Other namesAZD-0865
Legal status
Legal statusInvestigational
Identifiers
IUPAC name
CAS Number248919-64-4
PubChem CID9951066
UNIIE0OU4SC8DP
Chemical and physical data
FormulaC21H26N4O2
Molar mass

////////////Linaprazan, CHINA 2024, APPROVALS 2024, AstraZeneca, CINCLUS, GERD, linaprazan glurate, moderate to severe GERD, 248919-64-4, AZD 0865, E0OU4SC8DP, DTXSID90870279, X 842

Pradefovir


Pradefovir

WeightAverage: 423.79
Monoisotopic: 423.0863188

Chemical FormulaC17H19ClN5O4P

9-[2-[[(2R,4S)-4-(3-chlorophenyl)-2-oxo-1,3,2λ5-dioxaphosphinan-2-yl]methoxy]ethyl]purin-6-amine

2R,4S-(+)-9-(2-(4-(3-chlorophenyl)-2-oxo-1,3,2-dioxaphosphorinan-2-yl)methoxyethyl)adenine

HEPATITIS B VIRUS, APPROVALS 2024, CHINA 2024, Xi’an Xintong Pharmaceutical Research Co, Xinshumu

Pradefovir Mesylate

CAS No. : 625095-61-6, Remofovir mesylate

분자량519.90
화학식C18H23ClN5O7PS

Pradefovir is a cyclodiester antiviral prodrug with specific activity against hepatitis B virus (HBV). Pradefovir is specifically metabolized in the liver by hepatic enzymes, mainly by CYP4503A4, to adefovir. In turn, adefovir is phosphorylated by cellular kinases to its activated form adevofir diphosphate. By competing with the natural substrate dATP, the diphosphate form is incorporated into viral DNA and inhibits RNA-dependent DNA polymerase. This causes DNA chain termination and eventually results in an inhibition of HBV replication.

PAT

Syn

J. Med. Chem. 51 (2008) 666–676

SYN

https://pubs.acs.org/doi/10.1021/jm7012216

Syn

European Journal of Medicinal Chemistry 291 (2025) 117643

Pradefovir, developed by Xi’an Xintong Pharmaceutical Research Co., Ltd., is a liver-targeted nucleotide analog prodrug designed for the treatment of chronic hepatitis B virus (HBV) infection. It was approved
by the NMPA in 2024, under the brand name Xinshumu, for the treat ment of adult patients with chronic hepatitis B. Pradefovir utilizes HepDirect liver-targeting technology, allowing it to remain stable in the
gastrointestinal tract and bloodstream. It is specifically metabolized into its active form in the liver by the enzyme CYP3A4, leading to high hepatic concentrations and low systemic exposure. This targeted activation enhances antiviral efficacy while minimizing potential side effects on other organs. The clinical efficacy of pradefovir was demonstrated in a Phase III randomized, double-blind, positive-controlled trial (NCT04543565) involving patients with chronic hepatitis B. Participants were randomized to receive pradefovir or tenofovir disoproxil fumarate (TDF) in a 2:1 ratio, with a treatment duration of 96 weeks
followed by a 48-week open-label extension. Interim analysis conducted after 48 weeks showed that pradefovir achieved comparable reductions in HBV DNA levels to TDF, with a favorable safety profile. Regarding safety, pradefovir exhibited a favorable profile [90]. The total occurrence rate of adverse events was similar in both the pradefovir and TDF groups. Nevertheless, the incidence of drug-related adverse events was notably lower in the pradefovir group. Significantly, compared to the typical concerns associated with nucleotide analogs, pradefovir showed a diminished influence on renal function and bone mineral density. This is a crucial aspect considering the known side-effects of nucleotide an alogs in clinical applications, where issues related to kidney function and bone health often pose challenges. Pradefovir, in contrast, appears to have a more favorable safety profile in these regards, which could potentially offer significant advantages in long-term treatment scenarios
[91]. Additionally, it had minimal effects on lipid profiles, suggesting a lower potential risk for cardiovascular events during long-term therapy.
The approval of pradefovir mesylate offers a new therapeutic option for adult patients with chronic hepatitis B, providing effective antiviral activity with an improved safety profile, particularly concerning renal and bone health [92,93].
The synthetic route of Pradefovir Mesylate is shown in Scheme 21 [93]. The route commences with a stereoselective reduction of Prad-001 employing ( )-DIP-Cl, affording Prad-002. Subsequent acid-catalyzed cyclodehydration between the hydroxyl groups of Prad-002 and Prad-003 generates Pradefovir, followed by mesylate salt formation to complete the synthesis.

90-93

[90] Y. Gao, F. Kong, X. Song, J. Shang, L. Yao, J. Xia, Y. Peng, W. Liu, H. Gong, M. Mu,
H. Cui, T. Han, W. Chen, X. Wu, Y. Yang, X. Yan, Z. Jin, P. Wang, Q. Zhu, L. Chen,
C. Zhao, D. Zhang, W. Jin, D. Wang, X. Wen, C. Liu, J. Jia, Q. Mao, Y. Ding, X. Jin,
Z. Zhang, Q. Mao, G. Li, J. Niu, Pradefovir treatment in patients with chronic
hepatitis B: week 24 results from a multicenter, double-blind, randomized,
noninferiority, phase 2 trial, Clin. Infect. Dis. 74 (2022) 1925–1932.
[91] Y. Ding, H. Zhang, X. Li, C. Li, G. Chen, H. Chen, M. Wu, J. Niu, Safety,
pharmacokinetics and pharmacogenetics of a single ascending dose of pradefovir, a
novel liver-targeting, anti-hepatitis B virus drug, in healthy Chinese subjects,
Hepatol Int 11 (2017) 390–400.
[92] H. Zhang, M. Wu, X. Zhu, C. Li, X. Li, W. Jin, D. Zhang, H. Chen, C. Liu, Y. Ding,
J. Niu, J. Liu, Safety, efficacy, and pharmacokinetics of pradefovir for the
treatment of chronic hepatitis B infection, Antiviral Res 174 (2020) 104693.
[93] K.R. Reddy, M.C. Matelich, B.G. Ugarkar, J.E. G´omez-Galeno, J. DaRe, K. Ollis,
Z. Sun, W. Craigo, T.J. Colby, J.M. Fujitaki, S.H. Boyer, P.D. van Poelje, M.D. Erion,
Pradefovir: a prodrug that targets adefovir to the liver for the treatment of hepatitis
B, J. Med. Chem. 51 (2008) 666–676.

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

……

//////////Pradefovir, HEPATITIS B VIRUS, APPROVALS 2024, CHINA 2024, Xi’an Xintong Pharmaceutical Research Co, Xinshumu, Pradefovir Mesylate, Remofovir, 625095-60-5, GZE85Q9Q61, DTXSID10870372, MB 6866, MB-06866, ICN2001-3

Bevifibatide


Bevifibatide

Cas 710312-77-9

817.9 g/mol, C34H47N11O9S2

2-[(3S,6S,12S,20R,23S)-20-carbamoyl-12-[3-(diaminomethylideneamino)propyl]-3-(1H-indol-3-ylmethyl)-2,5,8,11,14,22-hexaoxo-17,18-dithia-1,4,7,10,13,21-hexazabicyclo[21.3.0]hexacosan-6-yl]acetic acid

APPROVALS 2025, CHINA 2025, Bio-Thera Solutions, Beitaning RegisteredAcute coronary syndromes

BAT-2094 | batifiban | Beitaning | Betagrin® | Compound I [CN101085809A]

  • OriginatorBio-Thera Solutions
  • ClassAntiplatelets; Cardiovascular therapies; Cyclic peptides
  • Mechanism of ActionIntegrin alphaVbeta3 antagonists; Platelet glycoprotein GPIIb-IIIa complex anatgonists
  • 02 Dec 2024Zhujiang Hospital plans a phase II BCAIS-I trial for Acute ischemic stroke in China (IV) (NCT06712004)
  • 07 Aug 2024Chemical structure information updated
  • 28 Jun 2024Registered for Acute coronary syndromes in China (IV) – First global approval
  • Correctin
  • 7AKM76YKN5

Bevifibatide is a synthetic cyclic heptapeptide, and its synthesis involves several stages of peptide chemistry. The primary methods used for producing peptides of this nature are solid-phase peptide synthesis (SPPS), followed by cleavage, purification, and cyclization. 

Bevifibatide is a cyclic peptide with the Peptide sequence Arg-Gly-Asp-MeAsp-Phg-Val-Nal. 

 Bevifibatide (Bio-Thera Solutions) is a synthetic cyclic heptapeptide that functions as a αIIbβ3 and αvβ3 integrin receptor antagonist [1]. It was designed to inhibit platelet aggregation as an antiplatelet cardiovascular therapy.

SYN

CN101085809

https://patentscope.wipo.int/search/en/detail.jsf?docId=CN83278873&_cid=P21-MEUT2B-21989-1

Example 1: Fmoc solid phase synthesis
        1: Synthesis of Fmoc-Cys(Trt)-HN-Rink Amide AM resin
        (1) Fmoc-Rink Amide AM resin (produced by Tianjin Nankai Hecheng Technology Co., Ltd., substitution degree 0.59 mmol/g, 8.4746 g) was added to the solid phase reaction column, washed three times with DMF, and swelled with DCM for 30 minutes.
        (2) The solution was drained and Fmoc-protection was removed with 20% piperidine in DMF at room temperature for 20 minutes.
        (3) The solution was drained, the resin was washed five times with DMF, and the solution was drained.
        (4) Fmoc-Cys(Trt)-OH (2.9285 g), HOBt (0.6755 g), and DIPCDI (0.8 ml) were dissolved in DMF (20 ml) and DCM (20 ml) and pre-reacted in an ice bath for 20 minutes.
        (5) Add the above reaction solution to the solid phase reaction column, N 2 Stir with air flow to ensure full contact and reaction with the resin, and react at room temperature (31°C) for 2 hours.
        (6) The solution was drained, and the resin was washed three times with DMF and once with DCM. Acetic anhydride (10 ml), pyridine (8 ml), and N 2 Stir with air flow to ensure full contact and reaction with the resin, and react at room temperature (31°C) for 10 hours.
        (7) The solution was drained, and the resin was washed three times with DMF, three times with DCM, and three times with methanol. The resin was then dried under vacuum to obtain Fmoc-Cys(Trt)-HN-Rink Amide AM resin (10.2578 g). The degree of substitution was measured to be 0.5086 mmol/g.
        2: Synthesis of Fmoc-Pro-Cys(Trt)-HN-Rink Amide AM resin
        (1) 10.2578 g of Fmoc-Cys(Trt)-Rink Amide AM resin (substitution degree: 0.5086 mmol/g) was added to the solid phase reaction column, washed three times with DMF, and swelled with DCM for 30 minutes.
        (2) The solution was drained and Fmoc-protection was removed with 20% piperidine in DMF at room temperature for 20 minutes.
        (3) The solution was drained, the resin was washed five times with DMF, and the solution was drained.
        (4) Fmoc-Pro-OH (5.061 g), HOBt (3.04 g), and DIPCDI (7.5 ml) were dissolved in DMF (20 ml) and DCM (20 ml) and pre-reacted in an ice bath for 20 minutes.
        (5) Add the above reaction solution to the solid phase reaction column, N 2 Stir with air flow to ensure full contact and reaction with the resin. React at room temperature (30°C) for 2 hours, and monitor the reaction progress with Kaiser test.
        (6) The solution was drained and the resin was washed three times with DMF to obtain Fmoc-Pro-Cys(Trt)-HN-Rink Amide AM resin.
        3: Synthesis of Mpr-X-Gly-Asp(OtBu)-Trp(Boc)-Pro-Cys(Trt)-HN-Rink Amide AM resin, where X is Arg(Pbf), Har(Pbf) or Lys(Boc)
        The reaction steps for coupling each protected amino acid are the same as 2, except that the protected amino acids to be coupled are: Fmoc-Trp(Boc)-OH (7.899 g); Fmoc-Asp(OtBu)-OH (6.172 g); Fmoc-Gly-OH (4.460 g); Fmoc-X-OH (9.732 g); Mpr (1.592 g).
        4: Linear crude peptide Mpr-X-Gly-Asp-Trp-Pro-Cys-NH 2 Preparation
        (1) The resin obtained in step 3 was washed three times with DMF, three times with DCM, and three times with methanol, and then dried under vacuum to obtain 21.182 g of Mpr-X-Gly-Asp(OtBu)-Trp(Boc)-Pro-Cys(Trt)-HN-Rink Amide AM resin.
        (2) The obtained resin was placed in a round-bottom flask and TFA (180 ml), H 2 A mixed solution of O (10 ml) and TIS (10 ml) was introduced into 2 , stir electromagnetically in an ice bath for 10 minutes, remove the ice bath, and react at room temperature (29°C) for 2 hours.
        (3) After the reaction is completed, the solution is filtered, and the resin is washed twice with TFA. The filtrates are combined and ice-cold ether (2 L) is added to the filtrate. A white precipitate is precipitated, and the precipitate is collected by centrifugation and fully dried in vacuo.
        (4) The dried white precipitate (4.237 g) was collected to obtain the crude linear peptide Mpr-X-Gly-Asp-Trp-Pro-Cys-NH 2 , sealed and stored at -20℃.
        5: Linear crude peptide Mpr-X-Gly-Asp-Trp-Pro-Cys-NH 2 Cyclization
        The crude linear peptide of batifiban and its analogues obtained in Example 4 was dissolved in water, and 1 mmol/ml of I 2 The mixture was stirred for 30 minutes at room temperature, and the cyclization reaction was followed by analytical HPLC until completion, thereby obtaining Mpr-X-Gly-Asp-Trp-Pro-Cys-NH 2 (Disulfide bridge,Mpr1-Cys7)。

SYN

European Journal of Medicinal Chemistry 291 (2025) 117643

Bevifibatide, developed by Bio-Thera Solutions, is a synthetic peptide that functions as a glycoprotein IIb/IIIa (GP IIb/IIIa) receptor antagonist. It is marketed under the brand name Beitaning. In 2024, the
NMPA approved Bevifibatide citrate injection for use in patients with acute coronary syndrome undergoing percutaneous coronary intervention (PCI), including coronary stent implantation, to reduce the risk of acute occlusion, in-stent thrombosis, no-reflow, and slow flow phenomena. Bevifibatide exerts its therapeutic effects by specifically binding to the GP IIb/IIIa receptors on platelets, thereby inhibiting the binding of fibrinogen, von Willebrand factor, and other adhesive ligands to these receptors [80]. This inhibition prevents platelet aggregation, reducing the risk of thrombotic complications during and after PCI procedures. The clinical efficacy of Bevifibatide was demonstrated in a multicenter Phase III trial involving patients with acute coronary syndrome undergoing PCI (NCT04567890). The study achieved its primary efficacy endpoint, with the composite endpoint event rate at 30 days post-procedure being significantly lower in the Bevifibatide group (4.06%) compared to the control group (6.56 %), indicating superior antithrombotic efficacy. Regarding toxicity, Bevifibatide was generally well-tolerated. The most common adverse events included bleeding complications, which are consistent with the pharmacological action of GP IIb/IIIa inhibitors. These events were manageable with appropriate clinical interventions, and the overall safety profile was comparable to other agents in its class. The approval of Bevifibatide provides a new therapeutic option for patients undergoing PCI, aiming to enhance procedural safety by mitigating thrombotic risks associated with such
interventions [81].
The synthetic route of Bevifibatide, shown in Scheme 19, comprises sequential amidation reactions: Bevi-001 reacts with Bevi-002 to form Bevi-003, which undergoes deprotection and subsequent coupling with
Bevi-004 to generate Bevi-005 [82]. This intermediate undergoes consecutive amidation steps with Bevi-006 and Bevi-008, producing Bevi-007 and Bevi-009 respectively. Bevi-009 then reacts with Bevi-010
to form Bevi-011, followed by coupling with Bevi-012 to yield Bevi-013. Subsequent amidation with Bevi-014 produces Bevi-015, which undergoes TFA-mediated deprotection to give Bevi-016. The final synthesis involves oxidation of the sulfhydryl group in Bevi-016 followed by iodine-mediated coupling to afford Bevifibatide.

80-82

[80] G. Tonin, J. Klen, Eptifibatide, an older therapeutic peptide with new indications:
from clinical pharmacology to everyday clinical practice, Int. J. Mol. Sci. 24 (2023)5446.
[81] H. Patel, I. Lunn, S. Hameed, M. Khan, F.M. Siddiqui, A. Borhani, A. Majid, S.M. Bell, M. Wasay, Treatment of cerebral venous thrombosis: a review, Curr. Med.Res. Opin. 40 (2024) 2223–2236.
[82] S. Tan, Y. Yang, Y. Li, Synthesis of N2-(3-mercapto-1-oxopropyl)-L-arginylglycyl-L-α-aspartyl-L
α-tryptophyl-Lα-prolyl-L Its Analogues, 2007 CN101085809A.

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

……

1. Zhao X, Yuan L, Gong Z, Li M, Yuan Y, Geng J. (2025)
New drugs approved by the NMPA in 2024: Synthesis and clinical applications.
Eur J Med Chem291: 117643. [PMID:40262297]

////////Bevifibatide, APPROVALS 2025, CHINA 2025, Bio-Thera Solutions, Beitaning, BAT 2094, batifiban, 710312-77-9, Correctin, 7AKM76YKN5

Tegileridine


Tegileridine

  • YFJS8L4TGU
  • CAS 2095345-66-5
  • (9R)-N-((1S,4S)-4-Ethoxy-1,2,3,4-tetrahydro-1-naphthalenyl)-9-(2-pyridinyl)-6-oxaspiro(4.5)decane-9-ethanamine
  • 434.6 g/mol

WeightAverage: 434.624
Monoisotopic: 434.293328472

Chemical FormulaC28H38N2O2

(1S,4S)-4-ethoxy-N-[2-[(9R)-9-pyridin-2-yl-6-oxaspiro[4.5]decan-9-yl]ethyl]-1,2,3,4-tetrahydronaphthalen-1-amine

Tegileridine fumarate

CAS#2245827-85-2 (fumarate)

Chemical Formula: C32H42N2O6

Exact Mass: 550.3000

Molecular Weight: 550.70

CHINA 2025, APPROVALS 2025, AISUTE, Jiangsu Hengrui

Tegileridine is under investigation in clinical trial NCT06458400 (To Evaluate the Efficacy and Safety of Tegileridine and Oliceridine Injections in the Treatment of Postoperative Pain).

Tegileridine is a drug which acts as a μ-opioid receptor agonist. It is closely related to compounds such as oliceridineTRV734, and SHR9352, and shares a similar profile as a biased agonist selective for activation of the G-protein signalling pathway over β-arrestin 2 recruitment.[1]

In January 2024, tegileridine was approved in China for the treatment of moderate to severe pain after abdominal surgery.[2]

SYN

CN107001347

https://patentscope.wipo.int/search/en/detail.jsf?docId=CN203399246&_cid=P20-METU4Y-21400-1

SYN

WO 2017/063509

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017063509&_cid=P20-METU6J-22458-1

[0183]Examples 1 and 2 

[0184](S)-1-Ethyl-N-(2-((R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl)ethyl)-1,2,3,4-tetrahydroquinolin-1-amine 1 

[0185](R)-1-ethyl-N-(2-((R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl)ethyl)-1,2,3,4-tetrahydroquinolin-1-amine 2 

[0186]

[0187](R)-2-(9-(pyridin-2-yl)-6-oxaspiro[4,5]decane-9-yl)acetaldehyde 1a (294 mg, 1.135 mmol, prepared by the method disclosed in patent application “WO2012129495”) and 1-ethyl-1,2,3,4-tetrahydroquinolin-4-amine 1b (200 mg, 1.135 mmol, prepared by the method disclosed in patent application “WO2014078454”) were dissolved in 15 mL of dichloromethane, stirred for 1 hour, and sodium triacetoxyborohydride (1.203 g, 5.675 mmol) was added and stirred for 16 hours. 20 mL of water was added, and the mixture was extracted with dichloromethane (20 mL×3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by high performance liquid chromatography to obtain the title product, 1-ethyl-N-(2-((R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl)ethyl)-1,2,3,4-tetrahydroquinolin-1-amine. Chiral preparation was performed (separation conditions: chiral preparative column Superchiral S-AD (Chiralway), 2 cm ID*25 cm, 5 um; mobile phase: CO 

2 :methanol:diethanolamine=75:25:0.05, flow rate: 50 g/min). The corresponding fractions were collected and concentrated under reduced pressure to give the title products 1 (98 mg, brown oil) and 2 (95 mg, yellow solid). 

[0188]Example 1: 

[0189]MS m/z(ESI):420.3[M+1]; 

[0190]Chiral HPLC analysis: retention time 4.028 minutes, chiral purity: 99.7% (chromatographic column: Superchiral S-AD (Chiralway), 0.46 cm ID*15 cm, 5 μm; mobile phase: CO2: methanol: diethanolamine = 75:25:0.05 (v/v/v)) 

[0191]

1H NMR(400MHz,DMSO-d 6)δ8.54(s,1H),7.72(s,1H),7.45(d,1H),7.20(s,1H),6.95(s,1H),6.78(d,1H),6.52(d,1H),6.37(s,1H),3.60(br,2H),3.18-3.43(m,3H),2.99(m,1H),2.33-2.45(m,3H),1.77-1.99(m,3H),1.19-1.60(m,12H),1.00-1.06(m,4H),0.63(m,1H).

[0192]Example 2: 

[0193]MS m/z(ESI):420.3[M+1]; 

[0194]Chiral HPLC analysis: retention time 3.725 minutes, chiral purity: 99.8% (chromatographic column: Superchiral S-AD (Chiralway), 0.46 cm ID*15 cm, 5 μm; mobile phase: CO2: methanol: diethanolamine = 75:25:0.05 (v/v/v)) 

[0195]

1H NMR(400MHz,DMSO-d 6)δ8.53(s,1H),7.72(s,1H),7.46(d,1H),7.20(s,1H),6.97(s,1H),6.85(d,1H),6.54(d,1H),6.40(s,1H),3.61(br,2H),3.17-3.25(m,3H),3.00-3.01(m,1H),2.33-2.46(m,3H),1.78-1.97(m,3H),1.24-1.65(m,12H),1.01-1.06(m,4H),0.61(m,1H).

SYN

US11111236

https://patentscope.wipo.int/search/en/detail.jsf?docId=US306969245&_cid=P20-METUA8-25189-1

Embodiment 1: Preparation of (1S,4S)-4-ethoxy-N-(2-((R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl)ethyl)-1,2,3,4-tetrahydronaphthalen-1-amine

Step One: Synthesis of Intermediate (D-1)

      The compound represented by the formula (E1) (25 g), potassium hydroxide (22.4 g) and ethylene glycol (150 mL) were mixed and the resulting mixture was stirred at 150° C. for 16 hours, and then the reaction was stopped. The reaction solution was cooled to room temperature, diluted with water (150 mL) and extracted with dichloromethane (150 mL×2). The aqueous phase was adjusted to pH=6-7 with 3M hydrochloric acid and extracted with dichloromethane (200 mL×4). The combined organic phase was washed with saturated sodium chloride solution (200 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give the product (26.1 g, pale yellow oil) with a yield of 97.4% and a HPLC purity of 92%.

Step Two: Synthesis of Intermediate (D2-1)

      The compound represented by the formula (D-1) (28 g) was dissolved in anhydrous ethanol (100 mL) and the temperature was raised to 50° C. The resolving agent S-phenylethylamine (6.2 g) was dissolved in anhydrous ethanol (100 mL) and the resulting S-phenylethylamine solution was added dropwise into the above solution at 50° C. The mixture was heated to reflux and stirred for 2 hours. Then the mixture was allowed to cool to 10° C. naturally, and solid was precipitated. The mixture was filtered, and the filter cake was washed to give the product (13 g, solid) with an enantiomeric excess (ee) value of 96.7%;
      recrystallization: the obtained 13 g solid product was added to anhydrous ethanol (80 mL), heated to reflux and stirred for 6 hours. Then the mixture was naturally cooled to 10° C. and solid was precipitated. The mixture was filtered, the filter cake was washed and dried to give the product (10.6 g) with an ee value of 99.0%.

Step Three: Synthesis of Intermediate (D2)

      KOH (2.18 g) was dissolved in water (120 mL), and then the compound represented by formula (D2-1) was dissolved in the solution. The mixture was extracted with dichloromethane (100 mL×3). The aqueous phase was adjusted to pH=6-7 with IN HCl solution and then extracted with dichloromethane (150 mL×3). The combined organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give the product (7 g) with a yield of 50% and an ee value of 99.4%.
      MS m/z (ESI): 276.71 [M+H] +, 298.68 [M+Na] +.
       1H NMR (400 MHz, CDCl 3) δ 8.50-8.51 (m, 1H), 7.73-7.77 (m, 1H), 7.51-7.53 (d, 1H), 7.21-7.24 (m, 1H), 3.73-3.84 (m, 2H), 2.78-2.81 (d, 1H), 2.58-2.63 (m, 1H), 2.53-2.56 (d, 1H), 2.39-2.43 (m, 1H), 1.98-2.02 (d, 1H), 1.87-1.94 (m, 1H), 1.76-1.80 (m, 1H), 1.61-1.65 (m, 1H), 1.39-1.58 (m, 4H), 1.14-1.19 (m, 1H), (m, 1H), (m, 1H).

Step Four: Synthesis of Intermediate (C2)

      Dichloromethane (8.5 kg) was added to a reaction flask, and then the raw material (R)-2-(9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl)acetic acid (350 g), N,O-dimethylhydroxylamine hydrochloride (148.8 g), EDCI (292.3 g) and DMAP (15.5 g) were added under stirring. After the resulting mixture was stirred for 15-25 minutes, DIPEA (492.4 g) was added. Then the mixture was stirred under argon protection at room temperature for 16-18 hours. A saturated ammonium chloride aqueous solution (2.8 kg) was added to the reaction solution, and the resulting mixture was stirred for 5-10 minutes and partitioned. The organic phase was washed with saturated ammonium chloride aqueous solution (2.8 kg×2) and saturated brine (2.7 kg), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated to dryness under reduced pressure and then dichloromethane (2.5 kg) was added. The mixture was concentrated to dryness under reduced pressure to give an oil (372.03 g) with a yield of 92.0%.
      MS m/z (ESI): 319.1 [M+H] +, 341.3 [M+Na] +.
       1H NMR (400 MHz, CDCl 3) δ 8.50-8.51 (m, 1H), 7.66-7.71 (m, 1H), 7.43-7.45 (d, 1H), 7.15-7.18 (m, 1H), 3.63-3.66 (m, 2H), 3.47 (s, 3H), 2.86-2.88 (d, 3H), 2.62-2.65 (d, 1H), 2.50-2.57 (m, 1H), 2.36-2.39 (d, 1H), 1.96-2.00 (d, 1H), 1.80-1.86 (m, 1H), 1.68-1.72 (m, 1H), 1.48-1.55 (m, 1H), 1.31-1.46 (m, 4H), 1.03-1.07 (m, 1H), 0.63-0.71 (m, 1H).

Step Five: Synthesis of Intermediate (B2)

      The compound represented by formula (C2) (334.4 g) was dissolved in toluene (2.2 kg) in a reaction flask. The solution was cooled to −45° C. to −35° C. and purged with argon, and then red aluminum (348.76 g) was added dropwise while maintaining the temperature between −45° C. to −35° C. After completion of the addition, the reaction solution was stirred at −45° C. to −35° C. for 3-4 hours, and then 10% citric acid aqueous solution (1 kg) was added to the reaction solution at −45° C. to −35° C. Then concentrated hydrochloric acid solution was added to adjust the pH to 2-3, followed by addition of ethyl acetate (1.8 kg). The mixture was stirred and allowed to stand to partition. The aqueous phase was adjusted to pH=11-13 with 5N sodium hydroxide solution, and then extracted with dichloromethane (3.3 kg×2). The combined dichloromethane phase was washed with saturated sodium chloride solution (2.7 kg), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure, and then dichloromethane (3.3 kg) was added. The mixture was concentrated again under reduced pressure to give a pale red oil, which was directly used in the next step.

Step Six: Synthesis of the Compound Represented by Formula (III)

      The above oil was added to a reaction flask, followed by addition of dichloromethane (8.5 kg) and the compound represented by formula (A1) (134.56 g). The resulting mixture was stirred for 2-3 hours, followed by addition of sodium triacetoxyborohydride (373.86 g). The mixture was stirred at room temperature for 16-18 hours, followed by addition of saturated sodium carbonate solution (2.66 kg). Then the mixture was adjusted to pH=11-13 by addition of 5N sodium hydroxide aqueous solution and partitioned. The organic phase was washed with saturated ammonium chloride aqueous solution (2.83 kg) and saturated sodium chloride aqueous solution (2.74 kg), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated to dryness under reduced pressure, and then acetonitrile (120 g) was added. The mixture was stirred at room temperature for 16-18 hours to crystallize, and then filtered. The filter cake was dried to give the product (206.87 g) with a yield of 68.0%.
      MS m/z (ESI): 435.3 [M+H] +.
       1H NMR (400 MHz, CDCl 3) δ 9.74 (d, 1H), 9.58 (d, 1H), 8.94 (d, 1H), 8.37 (d, 1H), 7.94 (d, 1H), 7.67 (d, 1H), 7.52 (d, 1H), 7.47 (t, 1H), 4.46-4.49 (m, 1H), 4.30-4.33 (m, 1H), 3.84-3.87 (m, 1H), 3.66-3.70 (m, 2H), 3.53-3.56 (m, 2H), 2.82-2.85 (d, 2H), 2.67 (s, 2H), 2.39-2.41 (m, 4H), 2.30-2.33 (m, 4H), 1.85 (s, 2H), 1.48-1.52 (m, 6H), 1.27 (m, 3H).

SYN

US20200054594A1

SYN

Tegileridine fumarate, developed by Jiangsu Hengrui Pharmaceuti
cals Co., Ltd., is a novel small-molecule analgesic that functions as a
complete opioid receptor agonist with relative selectivity for -opioid
receptors (MOR). It is marketed under the brand name Aisute. In 2024,
the NMPA approved Tegileridine fumarate injection for the treatment of moderate to severe pain following abdominal surgery. Tegileridine ex
erts its analgesic effects by activating MOR, leading to inhibition of
adenylate cyclase activity, decreased intracellular cAMP levels, and
subsequent modulation of ion channel conductance. This results in hy
perpolarization of neuronal membranes and reduced neuronal excit
ability, effectively alleviating pain. The clinical efficacy of Tegileridine
was evaluated in a Phase III randomized, double-blind, placebo-
controlled trial (NCT05012516) involving patients experiencing mod
erate to severe pain after abdominal surgery. The research indicated that
Tegileridine offered substantial alleviation of pain in contrast to the
placebo. It manifested a quick-acting property, and its analgesic effects
endured throughout the period of observation. In terms of toxicity,
Tegileridine was typically well-tolerated by the subjects. The most
frequently encountered adverse reactions were nausea, vomiting, and
dizziness, all of which were of mild to moderate intensity. Importantly,
Tegileridine exhibited a favorable safety profile with a lower incidence
of gastrointestinal adverse reactions compared to traditional MOR ag
onists, potentially offering an improved therapeutic window for post
operative pain management. The approval of Tegileridine provides a
new treatment option for patients suffering from moderate to severe
postoperative pain, particularly following abdominal surgeries,
addressing a significant clinical need in pain management [72,73].
The synthesis of Tegileridine fumarate, illustrated in Scheme 17,
begins with nucleophilic substitution reaction involving Tegi-001 to
yield Tegi-002 [74]. Tegi-002 is subsequently acidified to produce
Tegi-003. Finally, Tegi-003 undergoes reductive amination with
Tegi-004 to synthesize Tegileridine.

[72] S. Dhillon, Correction: tegileridine: first approval, Drugs 84 (2024) 1011.
[73] S. Dhillon, Tegileridine: first approval, Drugs 84 (2024) 717–720.
[74] X. Li, B. Feng, Y. Chen, T. Liu, F. He, M. He, W. Tao, P. Sun, Oxa Spiro Derivative
Useful in Treatment of Pain and pain-related Disease and Its Preparation, 2017.
CN107001347A.

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

……

References

  1.  WO 2017/063509, “Oxa spiro derivative, preparation method therefor, and applications thereof in medicines”, published 10 April 2018, assigned to Jiangsu Hengrui Medicine Company and Shanghai Hengrui Pharmaceutical Company Ltd .
  2.  Dhillon S (June 2024). “Tegileridine: First Approval”. Drugs84 (6): 717–720. doi:10.1007/s40265-024-02033-4PMID 38771484.
Clinical data
Trade names艾苏特
Legal status
Legal statusRx in China
Identifiers
IUPAC name
CAS Number2095345-66-5
PubChem CID129049969
UNIIYFJS8L4TGU
Chemical and physical data
FormulaC28H38N2O2
Molar mass434.624 g·mol−1
3D model (JSmol)Interactive image
SMILES
InChI

////////////Tegileridine, CHINA 2025, APPROVALS 2025, AISUTE, Jiangsu Hengrui, YFJS8L4TGU, 2095345-66-5, Tegileridine FUMARATE

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