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 2,841 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

Personal Links

Verified Services

View Full Profile →

Recent Posts

Ceftobiprole


C20H22N8O6S2

534.57

209467-52-7

(6R,7R)-7-[(2Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(N-hydroxyimino)acetamido]-8-oxo-3-{[(3E,3’R)-2-oxo-[1,3′-bipyrrolidin]-3-ylidene]methyl}-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid

  • BAL-9141
  • BAL-9141-000
  • BAL-9141000
  • BAL9141-000
  • RO 63-9141
  • RO-63-9141
  • RO-639141

ceftobiprole medocaril sodium

Ceftobiprole medocaril sodium (BAL5788 sodium) | Ceftobiprole Prodrug | MedChemExpress
Ceftobiprole medocaril sodium salt.png
Molecular Weight712.64
FormulaC26H25N8NaO11S2
CAS No.252188-71-9
AppearanceSolid
ColorOff-white to light yellow
SMILESO=C(C(N(C1=O)[C@@](SC2)([H])[C@@H]1NC(/C(C3=NSC(N)=N3)=N\O)=O)=C2/C=C(CCN4[C@@](CC5)([H])CN5C(OCC(OC6=O)=C(O6)C)=O)/C4=O)O[Na]

fda approved 4/3/2024, To treat certain bloodstream infections, bacterial skin and associated tissue infections, and community-acquired bacterial pneumonia
Press Release  zevtera

Ceftobiprole, sold under the brand name Zevtera among others, is a fifth-generation[5] cephalosporin antibacterial used for the treatment of hospital-acquired pneumonia (excluding ventilator-associated pneumonia) and community-acquired pneumonia. It is marketed by Basilea Pharmaceutica under the brand names Zevtera and Mabelio.[6][7][8][9][10][11] Like other cephalosporins, ceftobiprole exerts its antibacterial activity by binding to important penicillin-binding proteins and inhibiting their transpeptidase activity which is essential for the synthesis of bacterial cell walls. Ceftobiprole has high affinity for penicillin-binding protein 2a of methicillin-resistant Staphylococcus aureus strains and retains its activity against strains that express divergent mecA gene homologues (mecC or mecALGA251). Ceftobiprole also binds to penicillin-binding protein 2b in Streptococcus pneumoniae (penicillin-intermediate), to penicillin-binding protein 2x in Streptococcus pneumoniae (penicillin-resistant), and to penicillin-binding protein 5 in Enterococcus faecalis.[12]

Medical uses

In the US, ceftobiprole is indicated for the treatment of adults with Staphylococcus aureus bloodstream infections (bacteremia) including those with right-sided infective endocarditis;[4] adults with acute bacterial skin and skin structure infections;[4] and people with community-acquired bacterial pneumonia.[4]

Microbiology

Ceftobiprole has shown in vitro antimicrobial activity against a broad range of Gram-positive and Gram-negative pathogens. Among the Gram-positive pathogens, ceftobiprole has demonstrated good in vitro activity against methicillin-resistant Staphylococcus aureusmethicillin-susceptible Staphylococcus aureus and coagulase-negative staphylococci, as well as against strains of methicillin-resistant Staphylococcus aureus with reduced susceptibility to linezolid, daptomycin or vancomycin.[13] Ceftobiprole has also displayed potent activity against Streptococcus pneumoniae (including penicillin-sensitive, penicillin-resistant and ceftriaxone-resistant strains) and Enterococcus faecalis, but not against Enterococcus faecium. For Gram-negative pathogens, ceftobiprole has shown good in vitro activity against Haemophilus influenzae (including both ampicillin-susceptible and ampicillin-non-susceptible isolates), Pseudomonas aeruginosa and strains of Escherichia coliKlebsiella pneumoniae and Proteus mirabilis that do not produce extended-spectrum β-lactamases (ESBL). Like all other cephalosporins, ceftobiprole was inactive against strains that produce extended-spectrum β-lactamases.[14]

The efficacy of ceftobiprole has been demonstrated in two large randomized, double-blind, phase 3 clinical trials in patients with hospital-acquired and community-acquired pneumonia. Ceftobiprole was non-inferior to ceftazidime plus linezolid in the treatment of hospital-acquired pneumonia (excluding ventilator-acquired pneumonia) and non-inferior to ceftriaxone with or without linezolid in the treatment of community-acquired pneumonia.[15][16]

Pharmacology

Ceftobiprole medocaril

Ceftobiprole is the active moiety of the prodrug ceftobiprole medocaril and is available for intravenous treatment only. It is mainly excreted via the kidney.[17]

Society and culture

Legal status

500 mg powder

Ceftobiprole has been approved for the treatment of adults with hospital acquired pneumonia (excluding ventilator-acquired pneumonia) and community-acquired pneumonia in twelve European countries, Canada, and Switzerland.[18]

In February 2010, the Committee for Medicinal Products for Human Use of the European Medicines Agency adopted a negative opinion, recommending the refusal of the marketing authorization for the medicinal product Zeftera, intended for treatment of complicated skin and soft-tissue infections in adults. The company that applied for authorization is Janssen-Cilag International N.V. The applicant requested a re-examination of the opinion. After considering the grounds for this request, the CHMP re-examined the opinion, and confirmed the refusal of the marketing authorization in June 2010.[19]

syn

https://www.sciencedirect.com/topics/neuroscience/ceftobiprole

syn

WO2010136423

Processes for producing ceftobiprole medocaril are known per se. What the processes known from the prior art have in common is that, starting from 7-aminocephalosporanic acid, a large number of intermediates have to be produced, isolated and purified in order to obtain ceftobiprole medocaril of the general formula (1) in sufficient purity.

The compound of the general formula (1) is known per se and is described, for example, in WO 99/65920. It can be used for the treatment and prophylaxis of bacterial infectious diseases, especially infectious diseases caused by methicillin-resistant Staphylococcus Aureus strains.

WO 99/65920 describes, as the last step in the production process of ceftobiprole Medocaril, a reaction in which the Medocaril prodrug unit is introduced into a compound of the general formula (2).

STR1

The compound of the general formula (2) is also known per se and has been described, for example, in EP 0 849 269 A1. The compound of the general formula (2) is prepared according to EP 0 849 269 A1 starting from (2R,6R,7R)-te rt. B u toxyc abonylamin o-3-formyl-8-oxo-5-thia-1 -azabicyclo[4.2.0]oct-3-ene-2-carboxylic acid benzhydryl ester by Wittig reaction with (1 ‘-allyloxycarbonyl-2- oxo-[1,3’]bipyrrolidinyl-3-yl)-triphenylphosphonium bromide. The resulting Δ2 reaction product is reisomerized to the desired Δ3 isomer by sulfoxidation and subsequent reduction and then deprotected from the benzhydryl ester with trifluoroacetic acid. The acylation in position 7 occurs by reaction with (Z)-(5-amino-[1,2,4]-thiadiazol-3-yl)-trityloxyiminothioacetic acid S-benzothiazol-2-yl ester. The compound of the general formula (2) is then obtained by removing the protective groups.

In EP 1 067 131 A1 the formation of the ylide in toluene or a mixture of toluene and dichloromethane is tert by adding alkali. Butylate in tetrahydrofuran, which allows the base to be added as a solution. The reaction of the ylide with the corresponding aldehyde is described at a reaction temperature of -70 0 C.

EP 0 841 339 A1 relates to cephalosporin derivatives and processes for their production. WO 95/29182 also discloses intermediates for the production of cephalosporins.

WO 01/90111 describes a further production of ceftobiprole Medocaril in several stages starting from desacetyl-7-aminocephalosporanic acid by acylation with (Z)-(5-amino-[1,2,4]-thiadiazol-3-yl)- trityloxyiminothioacetic acid S-benzothiazol-2-yl ester in N,N-dimethylformamide, followed by in situ esterification with diphenyldiazomethane in dichloromethane to give the corresponding benzohydryl ester, which is precipitated and isolated by adding hexane. In the next step, this product is oxidized to the corresponding aldehyde using TEMPO/NaOCI in dichloromethane/water or with Braunstein in tetrahydrofuran/dichloromethane. The next reaction step involves the Wittig reaction to the 3-vinyl-substituted derivative, in which the reaction takes place in dichloromethane/toluene/tetrahydrofuran at -78°C. The crude product is stirred with ethanol and made from dichloromethane/tert. Butyl methyl ether recrystallized or purified chromatographically. According to the method disclosed in WO 01/901 11, the Wittig reaction is carried out at low temperatures of -80 to -70 0 C in a complex solvent mixture of dichloromethane, toluene and tetrahydrofuran. This leads to significant disadvantages when carrying out the reaction on a production scale, since regeneration of the process solvents is difficult.

EXAMPLES

1. Example: (6R,7R)-7-Amino-3[E-(R)-1′-(5-tert-butyloxycarbonyl)-2-oxo-[I.S’lbipyrrolidinyl-S-ylidenemethyll-β- oxo-S-thia-i -aza-bicyclo^^.Oloct^-ene-2-carboxylic acid

5 , 1 4 g of 7-amino-3-formyl-ceph-3-em-4-carboxy I at was dissolved in 2 7 , 8 m bis(trimethylsilyl)acetamide and 50 ml propylene oxide. 16.8 g of (1 R/S,3’R)-(1′-tert-butyloxycarbonyl-2-oxo-[1,3′]bipyrrolidinyl-3-yl)-triphenylphosphonium bromide (EP1067131, WO02/14332) slowly added in portions. Stirring was continued at 1 ° C until the starting material had reacted and then the crystalline precipitate was added

Nitrogen atmosphere filtered off and washed with 50 ml cyclohexane/bis(tirmethylsilyl)acetamide 99.5/0.5. After drying under vacuum, the desired product was obtained in silylated form.

The material was dissolved in 100ml dichloromethane and at 0 0 C with 50ml 3%

NaHCC> 3 solution added. The phases were separated, the organic phase was washed with 30 ml of water and the combined water phases were adjusted to pH 3.5 with 3% H 3 PO 4 after activated carbon treatment. The crystalline precipitate was filtered, washed with water and dried under vacuum.

Auswaage: 6,09g

1H-nmr(DMSO-d6) δ 1.39(s,9H), 2.00(m, 2H), 2.8-3.2(m, 2H), 3.2-3.5(m,6H), 3.84(ABq, 2H, J= 18.2Hz), 4.57(m,1H), 4.82(d,1H, J=5.1Hz), 5.01 (d,1H, J=5.1Hz), 7.21 (m,1H)

13 C-nmr(DMSO-d 6 ) d 24.63, 26.11 , 28.09, 28.89, 41.54, 44.94, 45.31 , 47.98, 48.34, 51.27, 52.00, 58.98, 63.76, 79.95, 121.95 , 126.19, 126.28, 129.90, 134.21, 154.97 , 164.36, 169.05, 169.13

MS- ESI negative mode: 927.2(2M-H, 100%, 463.1(M-H, 25%)

H2O content: 2.2 %

IR (golden gate, cm“1): 2978, 1793, 1682, 1551, 1397, 1363, 1330

2. Beispiel: (6R,7R)-7-Trimethylsilylamino-3[E-(R)-1′-(5-tert.butyloxycarbonyl)-2- oxo-[1 , 3′]bipyrrolidinyl -3 -ylidenemethyll-δ-oxo-δ-thia-i-aza-bicyclo^^.0]oct- 2-ene-2-carbonsäure trimethylsilylester


10.28 g of 7-amino-3-formyl-ceph-3-em-4-carboxylate were dissolved in 55.6 ml of bis(trimethylsilyl)acetamide and 100 ml of propylene oxide. 33.6 g of (1R/S, 3’R)-(1′-tert. Butyloxycarbonyl-2-oxo-[1,3′]bipyrrolidinyl-3-yl)-triphenylphosphonium bromide (EP1067131, WO02.) were then added at 0 0 C /14332) slowly added in portions over 22 hours. The mixture was stirred at 1° C. until the starting material had reacted and then the reaction mixture was cooled to -20 ° C. The crystalline precipitate was filtered off under a nitrogen atmosphere and washed in portions with 180 ml of cyclohexane/bis(trimethylsilyl)acetamide 99.5/0.5. After drying under vacuum, the bissilylated

Product received.

Weight: 16.2g

1H-nmr(CDCI3) δ 0.04,0.10,0.12(3s, 9H), 0,34(s, 9H), 1,43 (s, 9H), 1.74 (br s, 1H),

1.9-2.2 (m, 2H), 2.8-3.0 (m,2H), 3.2-3.7 (m,8H), 4.7-4.95 (m, 3H), 7.43 (m, 1H)

3. Beispiel: Dicyclohexylammonium (6R,7R)-7-Amino-3[E-(R)-1′-(5-tert. butyloxycarbonyl)-2-oxo-[1 ,3′]bipyrrolidinyl-3-ylidenemethyl]-8-oxo-5-thia-1 – aza-bicyclo[4.2.0]oct-2-ene-2-carboxylat

1.0g (6R,7R)-7-Trimethylsilylamino-3[E-(R)-1′-(5-tert.butyloxycarbonyl)-2-oxo-[I.Slbipyrrolidinyl-S-ylidenemethyO-δ-oxo-δ -thia-i-aza-bicyclo^^.Oloct^-ene^- carboxylic acid trimethylsilyl esters were dissolved in 10ml dichloromethane and a solution of 300mg dicyclohexylamine in 1ml EtOH and 10ml ethyl acetate was added. The precipitate was filtered off, washed with ethyl acetate and dried in vacuo.

Auswaage: 0,9g 1H-nmr(D2O/DMSO-d6) δ 0.9-1 .3(m, 10H), 1 .30(s,9H), 1 .4-2.18m,12H), 2.7- 3.5(01,1 OH), 3.64(ABq, J= 17.2Hz, 2H), 4.5 (m,1 H) * , 4.58 (d,1 H, J=5.1 Hz); 4.88 (d,1 H, J=5.1 Hz), 7.07 (s,1 H)

* partly overlaid by D20 signal

MS- ESI negative mode: 927.2(2M-H, 100%), 463.1 (M-H, 25%)

IR (golden gate, cm “1): 2932, 2856, 1754, 1692, 1671 , 1630, 1569, 1394, 1329

4. Specifically: (6R, 7R )-7-[(Z)-2-(5-Amino-[1 ,2,4]thiadiazol-3-yl)-2-hydroxyimino- acetylamino]-8-oxo- 3-[(E)-(R)-2-oxo-[1 , 3′]bipyrrolidinyl-3-ylidenemethyl]-5-thia-1-aza-bicyclo[4.2.0]oct-2-jen-2 carbons Trifluoroacetate

4.1 Variant A:

3.0g (6R,7R)-7-Amino-3[E-(R)-1′-(5-tert-butyloxycarbonyl)-2-oxo-[1,3′]bipyrrolidinyl-3-ylidenemethyl]-8 -oxo-5-thia-1-aza-bicyclo[4.2.0]oct-2-ene-2-carboxylic acid in silylated form was dissolved in 150 ml of dichloromethane at 0°. 600 μl of DMF/water 5/1 and 1.8 ml of bis(trimethylsilyl) acetamide and 2.29 g of 2-trityloxyimino-2-(5-amino-1,2,4-thiadiazol-3-yl) acetic acid chloride were then added Hydrochoride (J. Antibiotics 37:557 – 571, 1984) was added in portions.

After 3 hours at 0°, the mixture was poured into 30 ml MeOH/120 ml water and the methylene chloride phase was separated off. The organic phase was concentrated to 66g and 25ml of trifluoroacetic acid was added. After 10 minutes, 1.5 ml triethylsilane and 10 ml water were added and the mixture was cooled to -15 ° C. The organic phase was separated off and again with 6 ml

Washed trifluoroacetic acid/water 1/1. The combined aqueous phases were diluted to 150 ml with water and filtered through an adsorber resin column with XAD-1600. After washing out the column with water, elution was carried out with water/acetonitrile 85/15. The product-containing fractions were concentrated in vacuo and allowed to stand at 0° for post-crystallization. The crystalline

Product was filtered off, washed with water and dried under vacuum.

Auswaage: 2,66g

4.2 Variant B:

7.4g (6R,7R)-7-Amino-3[E-(R)-1′-(5-tert-butyloxycarbonyl)-2-oxo-[1,3′]bipyrrolidinyl-3-ylidenemethyl]-8 -oxo-5-thia-1-aza-bicyclo[4.2.0]oct-2-ene-2-carboxylic acid was dissolved at 0° in 781 ml of dichloromethane with the addition of 6.7 ml of triethylamine. 8.65 g of 2-trityloxyimino-2-(5-amino-1,2,4-thiadiazol-3-yl)-acetic acid chloride hydrochloride were then added in portions. After the starting material had reacted, the mixture was poured into 500 ml of water and the methylene chloride phase was separated off. The organic phase was dried over Na 2 SC> 4 and concentrated in vacuo.

The residue was dissolved in 148 ml of dichloromethane and 4.5 ml of triethylsilane and 74 ml of trifluoroacetic acid were added at room temperature. After 30 minutes, 222 ml of dichloromethane and 222 ml of water were added and the mixture was cooled to -20 0 C. The organic phase was separated off and washed again with a mixture of 37 ml of trifluoroacetic acid and 148 ml of water. The combined aqueous phases were diluted with water to 364 ml, filtered through an adsorber resin and eluted with acetonitrile/water 15/85.

The filtrate was concentrated to 35g on a Rotavapor, filtered and washed with water.

After drying in a vacuum, 4.5 g of the sample was obtained.

1H-nmr(DMSO-d6) δ 1.9-2.2(m,2H), 2.8-3.5(m, 8H), 3.85(Abq, 2H; J=18.3Hz), 4.63(m,1 H), 5.16(d, 2H, J=4.9Hz), 5.85(dd, 1 H, J1 = 4.9Hz, J2=8.4Hz), 7.23(s, 1 H),

8.06(s, 2H), 9.08 (br. s, 2H), 9.49(d, 2H, J=8.4Hz), 1 1.95 (s, 1 H)

5. Being typical: (6R.7R )-7-[(Z)-2-(5-Amino-[1 ,2,4]thiadiazol-3-yl)-2-hydroxyimino- acetylamino]- 8-oxo- . 3-[(E)-(R)-2-oxo-[1 ,3′]bipyrrolidinyl-3-ylidenemethyl]-5- thia-1 -aza-bicyclo[4.2.0]oct-2-jan-2 carbons

6.0g (6R,7R)-7-Amino-3[E-(R)-1′-(5-tert-butyloxycarbonyl)-2-oxo-[1,3′]bipyrrolidinyl-3-ylidenemethyl]-8 -oxo-5-thia-1-aza-bicyclo[4.2.0]oct-2-ene-2-carboxylic acid in silylated form was dissolved in 300 ml of dichloromethane at 0°. 1200 μl of DMF/water 5/1 and 8.1 ml of bis(tirmethylsilyl)acetamide were then added as well as 5.3g of 2-trityloxyimino-2-(5-amino-1,2,4-thiadiazol-3-yl)acetic acid chloride Hydrochoride (J. Antibiotics 37:557 – 571, 1984) added in portions. The mixture was then poured into 60 ml MeOH/240 ml water and the methylene chloride phase was separated off. The organic phase was concentrated to 48g and 1.5 ml of triethylsilane was added. After adding 50ml

Trifluoroacetic acid was stirred at room temperature for 60 min, 20 ml of water was added and the mixture was cooled to -15°C. The organic phase was separated off and washed again with 20 ml trifluoroacetic acid/water 1/1. The combined aqueous phases were diluted to 500 ml with water and treated with 2.0 g of activated carbon. After filtration, the solution was concentrated in vacuo.

The residue was diluted to 50 ml with water and adjusted to pH 6.9 with saturated NaHCO 3 solution. The mixture was stirred at 0 0 C for 2 hours, filtered and the precipitate washed with water.

Auswaage: 4,5g

1H-nmr(DMSO-d6/CF3COOD) δ 1.9-2.3(m,2H), 2.8-3.5(m,8H), 3.85(ABq, 2H, 18.7Hz), 4.61 (m,1 H), 5.16(d, 1 H,J=4.8Hz), 5.86(dd, 1 H,J1 =4.8Hz, J2=8.4Hz),

7.24(s,1 H), 8.05(br s, 2H), 8.93(s, 2H), 9.50(d,1 H,J=8.4Hz), 11.96(s, 1 H)

MS- ESI negative mode: 533.2(M-H, 10%)

6. Beispiel: Ceftobiprol Medocaril Na-SaIz

0, 5 3 g ( 6 R , 7 R )-7-[(Z)-2-(5-amino-[1,2,4]thiadiazol-3-yl)-2-hydroxyimino-acetylamino]-8- oxo-3-[(E)-(R)-2-oxo-[1,3′]bipyrrolidinyl-3-ylidenemethyl]-5-thia-1 – aza-bicyclo[4.2.0]oct-2-ene- 2 carboxylic acid were dissolved in 5 ml of dimethyl sulfoxide and 0.27 g of carbonic acid (5-methyl-2-oxo-[1,3]dioxol-4-ylmethyl)-4-nitrophenyl ester were added and stirred at room temperature. A solution of sodium ethyl hexanoate in 30 ml of acetone was added for precipitation. The precipitate was filtered and washed with acetone.

Auswaage: 0,6g

1H-nmr(DMSO-d6) δ 1.9-2.05(m, 2H), 2.10(s,3H), 2.7-3.1 (m,2H), 3.1-3.6(m,6H), 3.64(q, 2H; J=17.1 Hz), 4.56(m,1 H), 4.87(s,2H), 4.98(d,1 H,J=4.9Hz), 5.65(dd,1 H,J1 =4.9Hz, J2=8.4Hz), 7.34(s,1 H), 8.02(s,2H), 9.36(d,1 H,J=8.4Hz)

MS- ESI negative mode: 689.0(M-H, 100%)

Clinical data
Trade namesZevtera, Mabelio
Other namesRO0639141-000,[1] BAL9141,[2] ceftobiprole medocaril
AHFS/Drugs.comInternational Drug Names
License dataUS DailyMedCeftobiprole
Routes of
administration
Intravenous
Drug classCephalosporin antibacterial
ATC codeJ01DI01 (WHO)
Legal status
Legal statusAU: S4 (Prescription only)[3]UK: POM (Prescription only)US: ℞-only[4]In general: ℞ (Prescription only)
Identifiers
showIUPAC name
CAS Number209467-52-7 252188-71-9
PubChem CID6918430
DrugBankDB04918 
ChemSpider23350302 
UNII5T97333YZK
KEGGD08885 
ChEMBLChEMBL520642 
CompTox Dashboard (EPA)DTXSID40870229 
ECHA InfoCard100.129.666 
Chemical and physical data
FormulaC20H22N8O6S2
Molar mass534.57 g·mol−1
3D model (JSmol)Interactive image
hideSMILESC1CNC[C@@H]1N2CC/C(=C\C3=C(N4[C@@H]([C@@H](C4=O)NC(=O)/C(=N\O)/c5nc(sn5)N)SC3)C(=O)O)/C2=O
hideInChIInChI=1S/C20H22N8O6S2/c21-20-24-14(26-36-20)11(25-34)15(29)23-12-17(31)28-13(19(32)33)9(7-35-18(12)28)5-8-2-4-27(16(8)30)10-1-3-22-6-10/h5,10,12,18,22,34H,1-4,6-7H2,(H,23,29)(H,32,33)(H2,21,24,26)/b8-5+,25-11-/t10-,12-,18-/m1/s1 Key:VOAZJEPQLGBXGO-SDAWRPRTSA-N 
  (what is this?)  (verify)

References

  1. ^ Hebeisen P, Heinze-Krauss I, Angehrn P, Hohl P, Page MG, Then RL (March 2001). “In vitro and in vivo properties of Ro 63-9141, a novel broad-spectrum cephalosporin with activity against methicillin-resistant staphylococci”Antimicrobial Agents and Chemotherapy45 (3): 825–836. doi:10.1128/AAC.45.3.825-836.2001PMC 90381PMID 11181368.
  2. ^ Jones RN, Deshpande LM, Mutnick AH, Biedenbach DJ (December 2002). “In vitro evaluation of BAL9141, a novel parenteral cephalosporin active against oxacillin-resistant staphylococci”The Journal of Antimicrobial Chemotherapy50 (6): 915–932. doi:10.1093/jac/dkf249PMID 12461013.
  3. ^ “Prescription medicines: registration of new chemical entities in Australia, 2015”Therapeutic Goods Administration (TGA). 21 June 2022. Archived from the original on 10 April 2023. Retrieved 10 April 2023.
  4. Jump up to:a b c d https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/218275s000lbl.pdf
  5. ^ Scheeren TW (1 January 2015). “Ceftobiprole medocaril in the treatment of hospital-acquired pneumonia”Future Microbiology10 (12): 1913–1928. doi:10.2217/fmb.15.115PMID 26573022.
  6. ^ “Basilea announces distribution agreement with Cardiome to commercialize antibiotic Zevtera/Mabelio (ceftobiprole) in Europe and Israel”Basilea (Press release). 12 September 2017. Retrieved 7 April 2024.
  7. ^ “Basilea to launch Zevtera/Mabelio (ceftobiprole medocaril) in Europe through a commercial services provider” (Press release). Basilea Pharmaceutica. Archived from the original on 31 March 2019. Retrieved 20 September 2016.
  8. ^ “Basilea announces launch of antibiotic Zevtera (ceftobiprole medocaril) in Germany”Basilea (Press release). 5 December 2014. Retrieved 7 April 2024.
  9. ^ “Swissmedic approves Basilea’s antibiotic Zevtera (ceftobiprole medocaril) for the treatment of pneumonia”Basilea (Press release). 22 December 2014. Retrieved 7 April 2024.
  10. ^ “Basilea signs exclusive distribution agreement for Zevtera (ceftobiprole medocaril) in the Middle East and North Africa with Hikma Pharmaceuticals LLC”Basilea (Press release). 15 October 2015. Retrieved 7 April 2024.
  11. ^ “Basilea announces that Health Canada approved Zevtera for the treatment of bacterial lung infections”Basilea (Press release). 12 October 2015. Retrieved 7 April 2024.
  12. ^ Syed YY (September 2014). “Ceftobiprole medocaril: a review of its use in patients with hospital- or community-acquired pneumonia”Drugs74 (13): 1523–1542. doi:10.1007/s40265-014-0273-xPMID 25117196S2CID 2925496.
  13. ^ Zhanel GG, Lam A, Schweizer F, Thomson K, Walkty A, Rubinstein E, et al. (2008). “Ceftobiprole: a review of a broad-spectrum and anti-MRSA cephalosporin”. American Journal of Clinical Dermatology9 (4): 245–254. doi:10.2165/00128071-200809040-00004PMID 18572975S2CID 24357533.
  14. ^ Farrell DJ, Flamm RK, Sader HS, Jones RN (July 2014). “Ceftobiprole activity against over 60,000 clinical bacterial pathogens isolated in Europe, Turkey, and Israel from 2005 to 2010”Antimicrobial Agents and Chemotherapy58 (7): 3882–3888. doi:10.1128/AAC.02465-14PMC 4068590PMID 24777091.
  15. ^ Farrell DJ, Flamm RK, Sader HS, Jones RN (April 2014). “Activity of ceftobiprole against methicillin-resistant Staphylococcus aureus strains with reduced susceptibility to daptomycin, linezolid or vancomycin, and strains with defined SCCmec types”. International Journal of Antimicrobial Agents43 (4): 323–327. doi:10.1016/j.ijantimicag.2013.11.005PMID 24411474.
  16. ^ Nicholson SC, Welte T, File TM, Strauss RS, Michiels B, Kaul P, et al. (March 2012). “A randomised, double-blind trial comparing ceftobiprole medocaril with ceftriaxone with or without linezolid for the treatment of patients with community-acquired pneumonia requiring hospitalisation”. International Journal of Antimicrobial Agents39 (3): 240–246. doi:10.1016/j.ijantimicag.2011.11.005PMID 22230331.
  17. ^ Awad SS, Rodriguez AH, Chuang YC, Marjanek Z, Pareigis AJ, Reis G, et al. (July 2014). “A phase 3 randomized double-blind comparison of ceftobiprole medocaril versus ceftazidime plus linezolid for the treatment of hospital-acquired pneumonia”Clinical Infectious Diseases59 (1): 51–61. doi:10.1093/cid/ciu219PMC 4305133PMID 24723282.
  18. ^ “Zevtera 500 mg powder for concentrate for solution for infusion – Summary of Product Characteristics (SmPC)”(emc). 5 April 2023. Retrieved 1 June 2023.
  19. ^ “Zeftera (previously Zevtera) EPAR”European Medicines Agency (EMA). 18 February 2010. Retrieved 6 April 2024. Text was copied from this source which is copyright European Medicines Agency. Reproduction is authorized provided the source is acknowledged.

External links

  • Clinical trial number NCT03138733 for “Ceftobiprole in the Treatment of Patients With Staphylococcus Aureus Bacteremia” at ClinicalTrials.gov
  • Clinical trial number NCT03137173 for “Ceftobiprole in the Treatment of Patients With Acute Bacterial Skin and Skin Structure Infections” at ClinicalTrials.gov
  • Clinical trial number NCT00326287 for “Ceftobiprole in the Treatment of Patients With Community-Acquired Pneumonia” at ClinicalTrials.gov
  • Clinical trial number NCT03439124 for “Ceftobiprole in the Treatment of Pediatric Patients With Pneumonia” at ClinicalTrials.gov

////////Ceftobiprole, BAL-9141, BAL-9141-000, BAL-9141000, BAL9141-000, RO 63-9141, RO-63-9141, RO-639141, fda 2024, zevtera, approvals 2024, Ceftobiprole medocaril sodium salt

[H][C@@]1(NC(=O)C(=N/O)\C2=NSC(N)=N2)C(=O)N2C(C(O)=O)=C(CS[C@]12[H])\C=C1/CCN(C1=O)[C@]1([H])CCNC1

DANICOPAN


Danicopan.png

Danicopan

USFDA 3/29/2024, To treat extravascular hemolysis with paroxysmal nocturnal hemoglobinuria, Voydeya

C26H23BrFN7O3

580.418

(2S,4R)-1-[2-[3-acetyl-5-(2-methylpyrimidin-5-yl)indazol-1-yl]acetyl]-N-(6-bromopyridin-2-yl)-4-fluoropyrrolidine-2-carboxamide

  • ACH 0144471
  • ACH-4471
  • ACH0144471
  • ALXN 2040
  • ALXN-2040
  • ALXN2040

Danicopan, sold under the brand name Voydeya, is a medication used for the treatment of paroxysmal nocturnal hemoglobinuria.[2] It is a complement inhibitor which reversibly binds to factor D to prevent alternative pathway-mediated hemolysis and deposition of complement C3 proteins on red blood cells.[2]

Danicopan was approved for medical use in Japan in January 2024, and in the United States in March 2024.[3][4]

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired hematologic disease characterized by hemolysis, thrombophilia, and bone marrow dysfunction.1,7 Both hemolysis and thrombophilia are mediated primarily by the complement system.1 Standard therapy for PNH involves the use of complement C5 inhibitors (e.g. eculizumabravulizumab) which are effective in mitigating complement-mediated intravascular hemolysis and thromboembolism.1 Unfortunately, complement C5 inhibition does not address C3-mediated extravascular hemolysis, which occurs earlier in the complement cascade within the alternative pathway.1,5

Danicopan is a small molecule complement factor D inhibitor that selectively blocks the alternative pathway, thereby working to address extravascular hemolysis when used in conjunction with C5 inhibitors.3 It was first approved in January 2024 in Japan for patients with PNH,2,6 shortly after which the EMA adopted a positive opinion and recommended granting it marketing authorization.2 It was subsequently approved by the FDA in March 2024.4

SYN

WO2015130795

SYN

https://patents.google.com/patent/US9796741

 

SYN

PAT

https://patents.google.com/patent/US20230094124A1/en

https://patents.google.com/patent/US20230094124A1/en

 

  • Step 1: Synthesis of tert-Butyl (2S,4R)-2-((6-bromopyridin-2-yl)carbamoyl) fluoropyrrolidine-1-carboxylate (3): N-Boc-trans-4-Fluoro-L-proline (50.8 kg) was added to DCM (1000 L) in a glass-lined reactor under an atmosphere of nitrogen. The reaction mixture was cooled to 0±5° C. and N-methylimidazole (44.7 kg) was added while maintaining the temperature at 0±5° C. Methanesulfonyl chloride (29.97 kg) was slowly added to the reaction mixture followed by the addition of 2-amino-6-bromopyridine (2). The reaction temperature was warmed to room temperature and stirred for 12 h. The reaction was monitored by HPLC. After completion of the reaction water (2,000 kg) was added, the reaction was stirred and the DCM layer separated. The aqueous layer was once more extracted with DCM (1000 L). The combined DCM layer was washed in succession with dilute HCl, aqueous NaHCOand brine. The DCM extract was evaporated to dryness and tert-butyl (2S,4R)-2-((6-bromopyridin-2-yl)carbamoyl)-4-fluoropyrrolidine-1-carboxylate (3) was isolated using DCM heptane mixture and dried. Yield, 71.76 Kg (84.86%))
  • [0404]
    Step 2: Synthesis of (2S,4R)-N-(6-Bromopyridin-2-yl)-4-fluoropyrrolidine-2-carboxamide (4): To a solution 4M HCl/Dioxane (168 kg) was added intermediate 3 (40 kg) at 25±5° C. under an atmosphere of nitrogen and the reaction was stirred for 1 h. The reaction was monitored by HPLC and after completion, the reaction was diluted with DCM (800 L) and washed with aqueous NaHCO3. The DCM layer was separated and concentrated. The product, 2S,4R)-N-(6-bromopyridin-2-yl)-4-fluoropyrrolidine-2-carboxamide, (4), was isolated using DCM/heptane and dried. Yield, 25.81 kg, 87%.
  • [0405]
    Step 3: Synthesis of tert-Butyl 2-(3-acetyl-5-bromo-1H-indazol-1-yl)acetate (6): 1-(5-Bromo-1H-indazol-yl)ethan-1-one (5, 30 kg) was added to a reactor containing DMF (210 L) under an atmosphere of nitrogen followed by potassium carbonate (4.05 kg). Tert-butyl bromoacetate (3.42 kg) was added to the reaction mixture with stirring and maintaining the temperature at 30±10° C. After addition was complete, the reaction mixture was heated at 50±5° C. for 1 h. After the reaction was complete the reaction mixture was cooled to 25±5° C. and diluted with water (630 L). The precipitated solid was filtered, washed with water (90 L) and dried. Yield, 43.13 kg, 97.13%.
  • [0406]
    Step 4: Synthesis of tert-Butyl 2-(3-acetyl-5-(2-methylpyrimidin-5-yl)-1H-indazol-1-yl)acetate (9): Bispinnacolato diboron (14.67 kg) was added to a solution of 4-bromo methylpyrimidine (7, 10 kg) in dioxane (206 kg) under an atmosphere of nitrogen followed by the addition of potassium acetate (17 kg). The reaction mixture was degassed using nitrogen. Pd(dppf)Cl(0.94 kg) was added and the reaction mixture heated to 90±5° C. until the pyrimidine was consumed. The reaction mixture was cooled to 25±5° C. and intermediate 6 (16.33 kg) was added followed by potassium carbonate (20.7 kg) and water (16.33 kg) and the reaction was degassed using nitrogen. The reaction was again heated to 90±5° C. until completion. The reaction mixture was cooled to 25±5° C. and diluted with ethyl acetate (269 kg) and water (150 kg) maintaining the temp at 10±5° C. Activated charcoal (1 kg) was added to the mixture with stirring and then filtered through a bed of celite. The ethyl acetate layer was separated, washed with 5% aqueous sodium chloride followed by 5% L-Cysteine solution to remove palladium related impurities. The ethyl acetate layer was evaporated to dryness. The product (9) was isolated from MTBE/heptane. Yield, 11.8 kg, 56%.
  • [0407]
    Step 5: Synthesis of 2-(3-Acetyl-5-(2-methylpyrimidin-5-yl)-1H-indazol-1-yl)acetic acid (10): To a stirred solution of intermediate 9 (50 kg) in DCM (465 kg) at 15±5° C. was added TFA (374.5 kg) while maintaining the said temperature. The reaction was warmed to 35±5° C. and stirring continued until completion of the reaction. DCM and TFA were distilled off under reduced pressure. The residue was dissolved in DCM (kg) and stirred with aqueous sodium bicarbonate. The biphasic mixture was acidified with concentrated HCl and the pH was adjusted to 2-3. The precipitated solid was filtered, washed with water and dried. Yield, 42.4 kg, quantitative.
  • [0408]
    Step 6: Synthesis of Compound 1: To a solution of intermediate 9 (42 kg) in DMF (277 kg) was added intermediate 4 (38.7 kg) and the reaction was cooled to 10±5° C. Coupling agent TBTU (56.7 kg) was added to the reaction mixture followed by the addition of DIPEA (86.5 kg) while maintaining the reaction temperature at 10±5° C. The reaction was warmed to 25±+5° C. and stirred until complete. The reaction mixture was diluted with ethyl acetate (1344 kg) and washed with water twice. (The reaction may be washed with aq. K2COif fluorine related impurities are present.) Anhydrous sodium sulfate was added to silica gel and added to the ethyl acetate layer and filtered. The ethyl acetate layer was passed over a column of silica gel (40 kg) and the pure fractions were collected. The fractions were treated with activated charcoal and then filtered over celite. The palladium content was checked, and if above 10 ppm, the ethyl acetate layer was treated with palladium scavenging resin (SilabondThiol®). The ethyl acetate was evaporated to dryness under vacuum and the residue was crystallized from IPA (crystalline seed may be added) and heptane to afford Compound 1 Form II. Yield, 60 kg, 78%.

Society and culture

Legal status

In February 2024, the Committee for Medicinal Products for Human Use of the EMA adopted a positive opinion, recommending the granting of a marketing authorization for the medicinal product Voydeya, intended as add-on therapy to ravulizumab or eculizumab for the treatment of residual hemolytic anemia in adults with paroxysmal nocturnal hemoglobinuria (PNH).[2][5] The applicant for this medicinal product is Alexion Europe.[2]

Names

Danicopan is the international nonproprietary name.[6]

Clinical data
Trade namesVoydeya
Other namesACH-4471
Routes of
administration
By mouth
Drug classComplement factor D inhibitor
ATC codeL04AJ09 (WHO)
Legal status
Legal statusUS: ℞-only[1]In general: ℞ (Prescription only)
Identifiers
showIUPAC name
CAS Number1903768-17-1
DrugBankDB15401
ChemSpider75531295
UNIIJM8C1SFX0U
KEGGD11641
ChEMBLChEMBL4250860
ECHA InfoCard100.398.865 
Chemical and physical data
FormulaC6H3BrFN7O3
Molar mass320.038 g·mol−1
3D model (JSmol)Interactive image
hideSMILESCC(=O)C1=NN(CC(=O)N2C[C@H](F)C[C@H]2C(=O)NC2=CC=CC(Br)=N2)C2=C1C=C(C=C2)C1=CN=C(C)N=C1
hideInChIInChI=1S/C26H23BrFN7O3/c1-14(36)25-19-8-16(17-10-29-15(2)30-11-17)6-7-20(19)35(33-25)13-24(37)34-12-18(28)9-21(34)26(38)32-23-5-3-4-22(27)31-23/h3-8,10-11,18,21H,9,12-13H2,1-2H3,(H,31,32,38)/t18-,21+/m1/s1Key:PIBARDGJJAGJAJ-NQIIRXRSSA-N

References

  1. ^ “Novel Drug Approvals for 2024”U.S. Food and Drug Administration. 1 April 2024. Retrieved 2 April 2024.
  2. Jump up to:a b c d “Voydeya EPAR”European Medicines Agency. 22 February 2024. Archived from the original on 23 February 2024. Retrieved 24 February 2024. Text was copied from this source which is copyright European Medicines Agency. Reproduction is authorized provided the source is acknowledged.
  3. ^ “Voydeya (danicopan) granted first-ever regulatory approval in Japan for adults with PNH to be used in combination with C5 inhibitor therapy”AstraZeneca (Press release). 19 January 2024. Archived from the original on 24 February 2024. Retrieved 24 February 2024.
  4. ^ Research Cf (4 April 2024). “Novel Drug Approvals for 2024”FDA.
  5. ^ “First oral treatment against residual hemolytic anemia in patients with paroxysmal nocturnal hemoglobinuria”European Medicines Agency (EMA) (Press release). 23 February 2024. Retrieved 24 February 2024.
  6. ^ World Health Organization (2019). “International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 81”. WHO Drug Information33 (1). hdl:10665/330896.

Further reading

  • Lee JW, Griffin M, Kim JS, Lee Lee LW, Piatek C, Nishimura JI, et al. (December 2023). “Addition of danicopan to ravulizumab or eculizumab in patients with paroxysmal nocturnal haemoglobinuria and clinically significant extravascular haemolysis (ALPHA): a double-blind, randomised, phase 3 trial”. The Lancet. Haematology10 (12): e955–e965. doi:10.1016/S2352-3026(23)00315-0PMID 38030318.

External links

//////////fda 2024, Voydeya, danicopan, approvals 2024, ACH-4471, ACH 4471, ACH 0144471, ACH-4471, ACH0144471, ALXN 2040, ALXN-2040, ALXN2040

INAXAPLIN


INAXAPLIN
Inaxaplin.png

INAXAPLIN, 2446816-88-0

  • 2446816-88-0
  • S2SJ2RVZ6Y
  • UNII-S2SJ2RVZ6Y
  • C21H18F3N3O3

3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]-N-[(3S,4R)-4-hydroxy-2-oxopyrrolidin-3-yl]propanamide

Inaxaplin (VX-147) is a small-molecule apolipoprotein L1 inhibitor developed by Vertex Pharmaceuticals for APOL1-mediated kidney disease. In preliminary studies the drug has shown promise in treating people with kidney disease and multiple gain of function mutations on the APOL1 gene.[1][2]

    FSGS is a disease of the podocyte (glomerular visceral epithelial cells) responsible for proteinuria and progressive decline in kidney function. NDKD is a disease characterized by hypertension and progressive decline in kidney function. Human genetics support a causal role for the G1 and G2 APOL1 variants in inducing kidney disease. Individuals with 2 APOL1 risk alleles are at increased risk of developing primary (idiopathic) FSGS, human immunodeficiency virus (HIV)-associated FSGS, and NDKD. Currently, FSGS and NDKD are managed with symptomatic treatment (including blood pressure control using blockers of the renin angiotensin system), and patients with FSGS and heavy proteinuria may be offered high dose steroids. Corticosteroids induce remission in a minority of patients and are associated with numerous side effects. These patients, in particular individuals of recent sub-Saharan African ancestry with 2 APOL1 risk alleles, experience rapid disease progression leading to end-stage renal disease (ESRD). Thus, there is an unmet medical need for treatment for FSGS and NDKD.

PATENT

US20230271945, Compound 2

Compound 2

3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]-N-[(3S,4R)-4-hydroxy-2-oxo-pyrrolidin-3-yl]propanamide (2)

Synthesis of 3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]-N-[(3S,4R)-4-hydroxy-2-oxo-pyrrolidin-3-yl]propanamide (2)

      Method G: Amide Coupling with CDMT. A 2 L 3-neck RB flask with magnetic stirrer, temperature probe and nitrogen inlet was charged with 3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]propanoic acid S12 (90.5 g, 283.5 mmol) and (3S,4R)-3-amino-4-hydroxy-pyrrolidin-2-one S2 (39.9 g, 343.6 mmol) in DMF (1.65 L), and stirred for 15 minutes. CDMT (61.1 g, 348 mmol) was added. The mixture was then cooled to −2° C. on an ice bath. N-methylmorpholine was added (131 mL, 1.2 mol) dropwise over 20 minutes and the mixture was heated at 30° C. overnight. The reaction mixture was added into approx. 4.5 L of ice water, and extracted with EtOAc (1.2 L×4). The combined organic layers, were washed with 1.2 L of 1 M HCl (×3) and then water (1.2 L) and brine (1.2 L). The combined organic layers were dried over Na 2SO 4, filtered and concentrated. The mixture was washed through a silica gel plug (1.8 L of silica gel), first eluting with 25% EtOAc in dichloromethane (8 L) to remove impurities, followed by hot EtOAc (8 L), to elute the product. The EtOAc filtrate was concentrated in vacuo. TBME was then added (400 mL), and the mixture allowed to stirred for overnight. Filtration of the resulting solid afforded the product as a white solid. 62 g, 52%) 1H NMR (300 MHz, CD 3OD) δ 7.70-7.58 (m, 2H), 7.29-7.13 (m, 3H), 6.73 (ddd, J=11.1, 9.6, 2.2 Hz, 1H), 4.34 (td, J=7.6, 6.8 Hz, 1H), 4.21 (d, J=7.8 Hz, 1H), 3.56 (dd, J=9.9, 7.6 Hz, 1H), 3.20-3.04 (m, 3H), 2.65-2.53 (m, 2H). LCMS m/z 418.2 [M+H] +. Optical rotation: [α] D20.7=−14.01 (c=1.0, 10 mg in 1 mL of MeOH).

Alternative Procedure for Synthesis of Compound (2)

Step 1. Synthesis of Methyl (E)-3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]prop-2-enoate (C51)

      A solution of 5,7-difluoro-2-(4-fluorophenyl)-1H-indole C50 (100 g, 1.0 equiv) in dichloromethane (850 mL, 8.5 vol) was agitated at 22° C. Methyl 3,3-dimethoxypropionate (63 mL, 1.1 equiv) was charged followed by trifluoroacetic acid (96 mL, 3.1 equiv), which was rinsed forward with dichloromethane (25 mL, 0.25 vol). The batch was heated to 38° C. and stirred at that temperature. After 4 h, the batch was cooled to 22° C. and charged with n-heptane (200 mL, 2 vol). The mixture was stirred for no less than 1 h at 22° C. The slurry was filtered, and the reactor and the filter cake were washed with n-heptane (1×2 vol (200 mL) and 1×3 vol (300 mL)). The resulting solid was dried under vacuum with nitrogen bleed at 45° C. to afford the product C51 (127.7 g, 95% yield).

Step 2. Synthesis of Methyl 3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]propanoate (C52)

      To a hydrogenator was charged methyl (E)-3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]prop-2-enoate C51 (100.4 g, 1.0 equiv) followed by Pd(OH) 2/C (0.014 equiv). The vessel was sealed and three vacuum/purge cycles with N2 were performed. 2-MeTHF (2000 mL, 20 vol) was charged using residual vacuum and the resulting mixture was stirred at 22° C. The vessel was sealed and three vacuum/purge cycles with N2 were performed followed by one vacuum purge cycle with hydrogen (H 2). The temperature was adjusted to 22° C., and the vessel pressurized with 20 psi H 2. The mixture was agitated at 22° C. for 4 h. Three vacuum/purge cycles with nitrogen N2 were performed. The batch was filtered through a pad of Hyflo® and the filter cake was rinsed with 2-MeTHF (2×300 mL, 2×3 vol). The combined filtrates were placed under vacuum and distilled at ≤45.0° C. to 2.0 to 3.0 total volumes. The batch temperature was adjusted to 22° C. and the vessel was charge with n-heptane (1000 mL, 10 vol) over at least 1 h. A vacuum was applied and the filtrate distilled at 45.0° C. to 3.5 to 4.5 total volumes. The slurry was cooled to 22° C. and allowed to stir for no less than 1 h. The slurry was filtered and the filter cake was washed with n-heptane (1×1 vol (100 mL) and 1×0.5 vol (50 mL)). The solids were dried under vacuum with nitrogen bleed at 45° C. to afford the product C52 (91.9 g, 91% yield).

Step 3. Synthesis of 3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]propanoic Acid (S12)

      A mixture of methyl 3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]propanoate C52 (80.0 g, 1.0 equiv) and 2-MeTHF (480 mL, 6 vol) was agitated at 22° C. and treated with methanol (72 mL, 0.9 vol). A solution of KOH (27.1 g, 2.0 equiv) in water (107 mL, 1.3 vol) was charged over approximately 20 min. The resulting mixture was heated to an internal temperature of 35° C. and stirred for 3 h. The temperature was adjusted to 22° C. A vacuum was applied and the mixture was distilled at ≤45° C. to 3.0 total volumes. The internal temperature was adjusted to 12° C. The mixture was then charged with water (64 mL, 0.8 vol) and 2-MeTHF (304 mL, 3.8 vol). 6N HCl (75 mL, 0.9 vol) was slowly charged into the mixture with vigorous agitation until the batch attained a pH<3. The internal temperature was adjusted to 22° C., and the biphasic mixture was stirred for no less than 0.5 h. The stirring was stopped and the phases were allowed to separate for no less than 0.5 h. The lower aqueous phase was removed. Water (160 mL, 2 vol) was charged to the reactor at 22° C., and the biphasic mixture stirred for no less than 0.5 h. The stirring was stopped, and the phases allowed separated over no less than 0.5 h. The lower aqueous phase was removed and the batch was filtered through a pad of Hyflo®. The reactor and filter cake were rinsed with 2-MeTHF (160 mL, 2 vol). A vacuum was applied and the combined filtrates distilled at ≤40.0° C. to 2-3 total volumes. The vessel was charged with n-heptane (160 mL, 2 vol), a vacuum was applied and the filtrate distilled at 40.0° C. to 2 total volumes (this step was repeated one additional time). The mixture was then charged with additional n-heptane (144 mL, 1.8 vol). The internal temperature was adjusted to 40° C. and stirred for no less than 2 h. The internal temperature was adjusted to 22° C. over a minimum of 5 h and stirred for no less than 16 hours. The slurry was filtered. The filter cake was washed with n-heptane (3×40 mL, 3×0.5 vol). The solids were dried under vacuum with nitrogen bleed at 45° C. to afford product S12 (72.6 g, 95% yield).

Step 4. Synthesis of 3-[5,7-difluoro-2-(4-fluorophenyl)-1H-indol-3-yl]-N-[(3S,4R)-4-hydroxy-2-oxo-pyrrolidin-3-yl]propanamide (2)

      A mixture of S12 (50.0 g, 1.0 equiv), (3S,4R)-3-amino-4-hydroxypyrrolidin-2-one hydrochloride S2 (25.1 g, 1.05 equiv), and CDMT (30.3 g, 1.1 equiv) in DMF (250 mL, 5 vol) was agitated and cooled to 0° C. The reactor was charged with NMM (60 mL, 3.5 equiv) over no less than 1 h, while maintaining the internal temperature at ≤5° C. The batch was stirred at −5° C. for no less than 1 h. The batch was warmed to 22° C. over at least 1 h and stirred at 22° C. for 16 h. The batch was cooled to 0° C. Water (250 mL, 5 vol) was charged, while keeping the internal temperature <20° C. The mixture was charged with a 90/10 mixture of EtOAc/IPA (1000 mL, 20 vol). 6N HCl (40 mL, 0.8 vol) was then charged, while maintaining an internal temperature <10° C., until a pH˜1-3 was achieved. The internal temperature was adjusted to 22° C. and the biphasic mixture stirred for no less than 0.5 h. Stirring was stopped and the phases allowed to separate for no less than 0.5 h. The lower aqueous phase was removed. The aqueous layer was back extracted with a 90/10 mixture of EtOAc/IPA (2×250 mL, 2×5 vol) at 22° C. The combined organic phases from extractions were washed with water (5×500 mL, 5×10 vol) at 22° C., by mixing for no less than 0.5 h and settling for no less than 0.5 h for each wash. The batch was polish filtered. A vacuum was applied and the organic phase distilled at <50° C. to 9.5-10.5 total volumes. The mixture was charged with EtOAc (500 mL, 10 vol), vacuum was applied and the organic phase distilled at <50° C. to 9.5-10.5 total volumes (this step was repeated one more time). The mixture was charged with EtOAc (300 mL, 6 vol) and n-heptane (200 mL, 4 vol). The resulting slurry was heated to 50° C. and stirred for no less than 17 h. The mixture was then cooled to 22° C. over 2 h, and stirred for no less than 1 h. The slurry was filtered. The filter cake was washed with 1:1 EtOAc/n-heptane (2×150 mL, 2×3 vol). The solids were dried under vacuum with nitrogen bleed at ≤45° C. to afford Compound 2 (52.6 g, 80% yield).

Re-Crystallization of Compound 2

      Compound 2 (37.6 g, 1.0 equiv) was charged to a reactor followed by a 3:1 mixture of IPA/water (240 mL, 6.4 vol). The slurry was heated to an internal temperature of 75° C. The batch was cooled to an internal temperature of 55° C. and stirred at that temperature for at least 0.5 h. The batch was seeded with 0.5 wt % of a previously generated batch of Compound 2, as a suspension in a mixture of 3:1 IPA/water (4 mL, 0.1 vol). The mixture was stirred at 55° C. for no less than 1.5 h. Water (218 mL, 5.8 vol) was added over minimum period of 5 h while maintaining the temperature at 55° C. The slurry was cooled to 22° C. over no less than 5 h and stirred for no less than 2 h. The slurry was filtered. The filter cake was washed with 2:3 IPA/water (2×114 mL, 2×3 vol). The solids were dried under vacuum with nitrogen bleed at ≤45° C. to afford Compound 2 (34.5 g, 92% yield).

Form A of Compound 2

      12.3 kg of Compound 2 was charged to the reactor follow by a 3:1 mixture of 2-propanol/water. Agitation was initiated and the mixture was heated to 75° C. to achieve complete dissolution. The mixture was cooled to 55° C. over 1 hour and agitated at that temperature for 30 minutes. Agitation was continued for 1.5 hours. Water (5.8 vol) was charged over 5 h at 55° C., after which the mixture was cooled to 22° C. over 6 hours. The mixture was agitated at 22° C. for 2 hours then filtered under vacuum. The resulting wet cake was washed with a 3:1 mixture of 2-propanol/water (2.74 vol×2) and pulled dry under vacuum. The wet cake was further dried under vacuum with nitrogen bleed at 45° C. to yield 11.2 kg of Form A.

Hydrate Form A of Compound 2

      200 mg of Compound 2 was charged with 10 mL of water. The slurry was cooled to 5° C. and allowed to stir. Hydrate A was observed after 3 days of stirring.

PATENT

US20230271945, Compound 88

PATENT

US20230271945, Compound 89

str1
Flag Counter

AS ON DEC2021 3,491,869 VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@gmail.com

//////////

Clinical data
Other namesVX-147
Legal status
Legal statusInvestigational
Identifiers
showIUPAC name
CAS Number2446816-88-0
PubChem CID147289591
ChemSpider115009344
UNIIS2SJ2RVZ6Y
ChEMBLChEMBL5083170
Chemical and physical data
FormulaC21H18F3N3O3
Molar mass417.388 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI

References

  1. ^ Gbadegesin, Rasheed; Lane, Brandon (August 2023). “Inaxaplin for the treatment of APOL1-associated kidney disease”Nature Reviews Nephrology19 (8): 479–480. doi:10.1038/s41581-023-00721-0PMC 10461697PMID 37106136.
  2. ^ Egbuna, Ogo; Zimmerman, Brandon; Manos, George; Fortier, Anne; Chirieac, Madalina C.; Dakin, Leslie A.; Friedman, David J.; Bramham, Kate; Campbell, Kirk; Knebelmann, Bertrand; Barisoni, Laura; Falk, Ronald J.; Gipson, Debbie S.; Lipkowitz, Michael S.; Ojo, Akinlolu; Bunnage, Mark E.; Pollak, Martin R.; Altshuler, David; Chertow, Glenn M. (16 March 2023). “Inaxaplin for Proteinuric Kidney Disease in Persons with Two APOL1 Variants”. New England Journal of Medicine388 (11): 969–979. doi:10.1056/NEJMoa2202396PMID 36920755S2CID 257534251.

………///////INAXAPLIN

Golcadomide (CC-99282)


File:Golcadomide.svg
Golcadomide.png

Golcadomide (CC-99282)

Molecular Weight535.57
FormulaC28H30FN5O5
CAS No.2379572-34-4

(S)-2-(2,6-Dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline-1,3-dione

2-[(3S)-2,6-dioxopiperidin-3-yl]-4-[[2-fluoro-4-[(3-morpholin-4-ylazetidin-1-yl)methyl]phenyl]methylamino]isoindole-1,3-dione

  • 2-[(3S)-2,6-Dioxo-3-piperidinyl]-4-[[[2-fluoro-4-[[3-(4-morpholinyl)-1-azetidinyl]methyl]phenyl]methyl]amino]-1H-isoindole-1,3(2H)-dione (ACI)
  • (S)-2-(2,6-Dioxopiperidin-3-yl)-4-[[2-fluoro-4-[(3-morpholinoazetidin-1-yl)methyl]benzyl]amino]isoindoline-1,3-dione
  • CC 1007548
  • CC 99282
  • CC-99282
  • CC1007548
  • Golcadomide
  • WHO 12305

Golcadomide HCl, 2639939-36-7

Chemical Name: CC-99282 Hydrochloride; 2-[(3S)-2,6-Dioxopiperidin-3-yl]-4-[[2-fluoro-4-[(3-morpholin-4-ylazetidin-1-yl)methyl]phenyl]methylamino]isoindole-1,3-dione hydrochloride

Novel potent and orally active cereblon (CRBN) E3 ligase modulator

Ref: https://www.sciencedirect.com/science/article/abs/pii/S0268960X22000686

Golcadomide is a modulator of the E3 ubiquitin ligase complex containing cereblon (CRL4-CRBN E3 ubiquitin ligase), with potential immunomodulating and antineoplastic activities. Upon administration, golcadomide specifically binds to cereblon (CRBN), thereby affecting the ubiquitin E3 ligase activity, and targeting certain substrate proteins for ubiquitination. This induces proteasome-mediated degradation of certain transcription factors, some of which are transcriptional repressors in T-cells. This leads to modulation of the immune system, including activation of T-lymphocytes, and downregulation of the activity of other proteins, some of which play key roles in the proliferation of certain cancer cell types. CRBN, the substrate recognition component of the CRL4-CRBN E3 ubiquitin ligase complex, plays a key role in the ubiquitination of certain proteins.

PATENT

US20220324855

Example 1: Synthesis of (S)-2-(2,6-Dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline-1,3-dione (Compound 1)

(MOL)(CDX)
      (S)-2-(2,6-Dioxopiperidin-3-yl)-4-((2-fluoro-4-(hydroxymethyl)benzyl)amino)isoindoline-1,3-dione: A suspension of (S)-4-amino-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (5.00 g, 18.3 mmol) and 2-fluoro-4-(hydroxymethyl)benzaldehyde (2.82 g, 18.30 mmol) in 2:1 dioxane-MeOH (75 mL) was cooled to 0° C. and B 1014 (4.92 g, 40.3 mmol) was added in small portions over 5 minutes. The reaction flask was fitted with a septum and needle vent (pressure) and vigorously stirred for 10 minutes. The mixture was allowed to reach ambient temperature and stirred for 3 hours. The mixture was concentrated and the residue purified by silica gel chromatography (0-10% MeOH-DCM) to provide (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-(hydroxymethyl)benzyl)amino)isoindoline-1,3-dione as a yellow solid (4.23 g, 56%). LCMS (ESI) m/z 411.8 [M+H] +.
      (S)-4-((4-(Chloromethyl)-2-fluorobenzyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione: A solution of (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-(hydroxymethyl)benzyl)amino)isoindoline-1,3-dione (0.727 g, 1.77 mmol) in dry NMP (6 mL) was cooled to 0° C. and methane sulfonyl chloride (0.275 mL, 3.35 mmol) and DIEA (0.617 mL, 3.53 mmol) were added sequentially. The reaction mixture was allowed to reach ambient temperature and was stirred for 18 hours. The reaction mixture was slowly added to H 2O (60 mL) cooled to 0° C. with vigorous mixing. The resulting suspension was filtered and the collected solid was washed with H 2O and Et 2O. The solid was dissolved in EtOAc and the solution dried with MgSO 4, filtered and concentrated to provide (S)-4-((4-(chloromethyl)-2-fluorobenzyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione as a yellow solid (0.600 g, 79%). LCMS (ESI) m/z 430.0 [M+H] +.
      (S)-2-(2,6-Dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline-1,3-dione: To a solution of (S)-4-((4-(chloromethyl)-2-fluorobenzyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (300 mg, 0.698 mmol) in dry DMSO (1.0 mL) was added 4-(azetidin-3-yl)morpholine hydrochloride (125 mg, 0.698 mmol) and DIEA (0.122 mL, 0.698 mmol). The reaction mixture was stirred at ambient temperature for 18 hours and was diluted with DMSO (1 mL). The solution was purified by chiral reverse-phase chromatography to give (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline-1,3-dione (89 mg, 24%, 97% ee). LCMS (ESI) m/z 536.2 [M+H] +.

PATENT

WO2022271557

Example 1: Synthesis of (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3- morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline -1,3-dione

[00242] Synthesis of Ethyl 4-nitro-1,3-dioxo-isoindoline-2-carboxylate (Compound 10): To a solution of 4-nitroisoindoline-1,3-dione (Compound 11, 440 g, 2.29 mol) and TEA (262 g, 2.59 mol, 359 mL) in dry DMF (2.2 L) was cooled to 0 °C and ethyl chloroformate (313 g, 2.89 mol, 275 mL) was added dropwise over 5 minutes. The reaction mixture was stirred at 22 °C for 10 hours. The mixture was slowly added to chilled water (10 L) and the resulting suspension stirred for 5 minutes. The suspension was filtered and the filter cake was washed with water (1 L). The solid was dissolved with ethyl acetate (5 L) and the organic phase was washed with aqueous HCl (1 M, 1 L), water (2 L) and brine (2 L). The organic phase was dried over sodium sulfate , filtered and concentrated to give Compound 10 (360 g, 59%) as a white solid. 1 H NMR (400 MHz CDCl 3 ) δ ppm 8.24 (d, J = 7.6 Hz, 1H), 8.19 (d, J = 8.4 Hz, 1H), 8.06-8.02 (m, 1H), 4.49 (q, J = 7.2 Hz, 2H), 1.44 (t, J = 6.8 Hz, 3H).

[00243] Synthesis of tert-Butyl (4S)-5-amino-4-(4-nitro-1,3-dioxo-isoindolin-2-yl)-5-oxo-pentanoate (Compound 6): To a solution of Compound 10 (165 g, 625 mmol) and DIEA (113 g, 874 mmol, 153 mL) in dry THF (1700 mL) was added tert-butyl (4S)-4,5-diamino-5-oxo-pentanoate hydrochloride ( 149 g, 625 mmol) and heated at reflux for 10 hours. The reaction mixture was concentrated under reduced pressure. The resulting residue was diluted with methyl tert-butyl ether (5 L) and stirred at 20 °C for 1 hour. The suspension was filtered and the filter cake was dissolved with DCM (4 L). The organic phase was washed with water (1.5 L x 3), brine (1.5 L) and dried over sodium sulfate . The organic phase was filtered and concentrated under reduced pressure to give a light yellow oil. The oil was diluted with hexane / ethyl acetate (10/1, 2 L) and stirred until a light yellow suspension formed. The suspension was filtered and the filter cake was triturated and concentrated in vacuum to give Compound 6 (175 g, 74%) as a light yellow solid. 1 H NMR (400 MHz CDCl3) δ ppm 8.12 (d, J = 8.0 Hz, 2H), 7.94 (t, J = 8.0 Hz, 1H), 6.48 (s, 1H), 5.99 (s, 1H), 4.84- 4.80 (m, 1H), 2.49-2.44 (m, 2H), 2.32-2.27 (m, 2H), 1.38 (s, 9H).

[00244] Synthesis of tert-Butyl (S)-5-amino-4-(4-amino-1,3-dioxoisoindolin-2-yl)-5-oxopentanoate (Compound 5): To a suspension of Compound 6 (170.0 g, 450.5 mmol, 1.00 eq) in DMA (1.00 L) was added palladium on carbon (50.0 g, 10% purity) under nitrogen. The suspension was degassed under vacuum and purged with hydrogen gas several times. The mixture was stirred under hydrogen gas (50 psi) at 25 °C for 16 hours. The mixture was filtered and the filtrate was poured into cooled water (3.0 L). The mixture was stirred at 10 °C for 1 hour and filtered. The filter cake was washed with water (700 mL) and dissolved in DCM (1.00 L). The organic phase was dried over sodium sulfate , filtered and concentrated under reduced pressure to give Compound 5 (107 g, 68%) as a green solid. 1 H NMR (400 MHz DMSO-d 6 ) δ ppm 7.52 (s, 1H), 7.43 (dd, J = 8.4, 7.2 Hz, 1H), 7.13 (s, 1H), 6.95-6.99 (m, 2H), 6.42 (s, 2H), 5.75 (s, 1H), 4.47-4.51 (m, 1H), 2.32-2.33 (m, 1H), 2.14-2.20 (m, 3H), 1.32 (s, 9H); HPLC purity, 100.0%; SFC purity, 100.0% ee.

[00245] Synthesis of 2-fluoro-4-(hydroxymethyl)benzaldehyde (Compound 8): To a solution of 4-(((tert-butyldimethylsilyl)oxy)methyl)-2-fluorobenzaldehyde (370.0 g, 1.38 mol, 1.00 eq ) in THF (1.85 L) was added a solution of p-toluenesulfonic acid monohydrate (78.7 g, 413.6 mmol, 0.30 eq) in water (1.85 L) drop-wise at 10 °C. The mixture was stirred at 27 °C for 16 hours. TEA (80 mL) was added drop-wise and stirred for 10 minutes. The organic phase was separated and the aqueous phase was extracted with ethyl acetate (600 mL × 4). The combined

organic phase was washed with brine (1.50 L), dried over anhydrous sodium sulfate , filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give Compound 8 (137.5 g, 76%) as a yellow oil. 1 H NMR (400 MHz CDCl 3 ) δ ppm 10.34 (s, 1H), 7.86 (dd, J = 8.0, 7.2 Hz, 1H), 7.25 (s, 1H), 7.22 (d, J = 4.4 Hz, 1H) , 4.79 (d, J = 6.0 Hz, 2H), 1.91 (t, J = 6.0 Hz, 1H).

[00246] Synthesis of tert-Butyl (S)-5-amino-4-(4-((2-fluoro-4-(hydroxymethyl)benzyl)amino)-1,3-dioxoisoindolin-2-yl)-5- oxopentanoate (Compound 2-b): To a solution of Compound 5 (100.0 g, 287.9 mmol, 1.00 eq) and Compound 8 (57.7 g, 374.3 mmol, 1.30 eq) in dry DCM (1.00 L) was added TFA (164.1 g , 1.44 mol, 5.00 eq) at 0 °C. The reaction mixture was stirred at 28 °C for 2 hours. To the solution was added sodium cyanoborohydride (27.1 g, 431.8 mmol, 1.50 eq) at 0 °C. The mixture was stirred at 28 °C for 30 minutes. The reaction mixture was quenched by addition of MeOH (600 mL) and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give Compound 2-b (110.0 g, 74.0%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6) δ ppm 7.56 (s, 1H), 7.50 (dd, J = 8.4, 7.2 Hz, 1H), 7.34 (t, J = 8.0 Hz, 1H), 7.02-7.18 ( m, 4H), 6.94-7.01 (m, 2H), 4.57 (d, J = 6.0 Hz, 2H), 4.47-4.53 (m, 3H), 2.31-2.35 (m, 1H), 2.15-2.22 (m, 3H), 1.31 (s, 9H); HPLC purity, 94.0%; SFC purity, 100.0% ee.

[00247] Synthesis of tert-butyl (S)-5-amino-4-(4-((4-(chloromethyl)-2-fluorobenzyl)amino)-1,3-dioxoisoindolin-2-yl)-5-oxopentanoate (Compound 2-a): To a solution of Compound 2-b (100.0 g, 206.0 mmol, 1.00 eq) in NMP (430.0 mL) was added DIEA (79.9 g, 617.9 mmol, 3.00 eq) and MsCl (47.2 g, 411.9 mmol, 2.00 eq) at 0 °C. The ice bath was removed, and the reaction was stirred at 28°C for 10 hours. The reaction was poured into cooled water (<10°C, 2.0 L) and stirred for 10 minutes. The mixture was extracted with methyl tert-butyl ether (750 mL x 3). The combined organic layer was washed with brine (1.25 L), dried over sodium sulfate , filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give Compound 2-a (86.0 g, 81.2%) as a yellow solid.

1 H NMR (400 MHz DMSO-d 6 ) δ ppm 7.55 (s, 1H), 7.50 (dd, J = 8.4, 7.2 Hz, 1H), 7.38 (t, J = 8.0Hz, 1H), 7.31 (dd, J = 10.8, 1.6 Hz, 1H), 7.23 (dd, J = 8.0, 1.6 Hz, 1H), 7.16 (s, 1H), 7.11 (t, J = 6.4 Hz, 1H), 7.00 (d, J = 7.2 Hz, 1H), 6.95 (d, J = 8.4 Hz, 1H), 4.74 (s, 2H), 4.61 (d, J = 6.4 Hz, 2H), 4.49-4.53 (m, 1H), 2.29-2.38 (m , 1H), 2.16-2.25 (m, 3H), 1.30 (s, 9H); HPLC purity, 98.0%; SFC purity, 100.0% ee.

[00248] Synthesis of tert-butyl (S)-5-amino-4-(4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)-1,3- dioxoisoindolin-2-yl)-5-oxopentanoate (Compound 2): To a solution of 4-(azetidin-3-yl)morpholine hydrochloride (Compound 7 HCl, 30.5 g, 170.7 mmol, 1.00 eq) and DIEA (66.2 g, 512.0 mmol, 3.00 eq) in DMSO (350.0 mL) was added to a solution of Compound 2-a (86 g, 170.65 mmol, 1.00 eq) in DMSO (350.0 mL) drop-wise at 15 °C. The reaction mixture was stirred at 28 °C for 16 hours. The reaction mixture was poured into cold half saturated brine (<10°C, 2.5 L) and extracted with ethyl acetate (1.50 L, 1.00 L, 800.0 mL). The combined organic phase was washed with saturated brine (1.50 L), dried over sodium sulfate , filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give Compound 2 (68.3 g, 65.7%) as a yellow solid. 1 H NMR (400 MHz DMSO-d 6 ) δ ppm 7.55 (s, 1H), 7.50 (dd, J = 8.4, 7.2 Hz, 1H), 7.31 (t, J = 8.0 Hz, 1H), 7.16 (s, 1H), 6.94-7.10 (m, 5H), 4.56 (d, J = 6.4 Hz, 2H), 4.49-4.52 (m, 1H), 3.54-3.55 (m, 6H) 3.31-3.32 (m, 3H), 2.81-2.88 (m, 3H), 2.29-2.38 (m, 1H), 2.15-2.25 (m, 7H), 1.30 (s, 9H); HPLC purity, 100.0%; SFC purity, 100.0% ee.

[00249] Synthesis of (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline -1,3-dione (Compound 1): A solution of Compound 2 (30.0 g, 49.2 mmol, 1.00 eq) and benzenesulfonic acid (31.1 g, 196.8 mmol, 4.00 eq) in acetonitrile (480.0 mL) was stirred at reflux for 3 hours. The reaction was cooled to 20 °C, poured into cold brine:saturated sodium bicarbonate solution (1:1, <10 °C, 2.0 L) and extracted with ethyl acetate (1.0 L). The organic phase was washed with cold brine:saturated sodium bicarbonate solution (1:1, <10°C, 1.00 L) once more. The combined aqueous phase was extracted with ethyl acetate (500.0 mL x 2). The combined organic phase was washed with cold brine (<10°C, 1.0 L), dried over sodium sulfate , filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography to give Compound 1 (17.5 g, 66.0%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 11.10 (s, 1H), 7.54 (t, J = 8.0 Hz, 1H), 7.30 (t, J = 8.0 Hz, 1H), 7.04-7.10 (m , 4H), 7.00 (d, J = 8.4 Hz, 1H), 5.07 (dd, J = 12.8, 5.2 Hz, 1H), 4.58 (d, J = 6.4 Hz, 2H), 3.53-3.55 (m, 6H) , 3.30-3.32 (m, 2H), 2.81-2.89 (m, 4H), 2.54-2.61 (m, 2H), 2.20 (m, 4H) 2.03-2.06 (m, 1H); HPLC purity, 100.0%; SFC purity, 97.2% ee; LCMS (ESI) m/z 536.1 [M+H] + .

Example 2: Synthesis of (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3- morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline -1,3-dione

[00250] Synthesis of tert-butyl (S)-5-amino-4-(4-nitro-1,3-dioxoisoindolin-2-yl)-5-oxopentanoate (Compound 6): Ethyl acetate (245 mL, 5 V ), 3-nitrophthalic anhydride (49.1 g, 0.25 mol, 1 eq), and tert-butyl (S)-4,5-diamino-5-oxopentanoate hydrochloride (59.2 g, 0.25 mol,

1 eq) were charged into a reactor and cooled to 15-20°C. A premade solution of CDI (66.7 g,

0.41 mol, 1.5 eq) in DMF (245 mL, 5 V) was charged and the mixture was stirred at 20-25°C for

1 hour. The reaction was quenched with 15% (wt/wt) aqueous citric acid solution (10 V).

EtOAc (5 V) was added, the mixture was agitated and the phases split and separated. Tea

aqueous layer was extracted with EtOAc (5 V) and the combined organic layers were washed

twice with a 5% (wt/wt) aqueous citric acid solution (5 V each wash). The organic layer was

distilled at reduced pressure to 5 V and further continuously distilled at reduced pressure with the addition of iPrOH (10 V), maintaining a constant volume at 5 V. The final distillate was diluted to 13 V with iPrOH and used in the next step without further handling. 91% solution yield. [00251] Synthesis of tert-butyl (S)-5-amino-4-(4-amino-1,3-dioxoisoindolin-2-yl)-5-oxopentanoate (Compound 5): The solution of Compound 6 in iPrOH was charged to a hydrogenation reactor. 10% palladium on carbon (50% wet, 4.65g 5 wt%) was charged. The reaction mixture was stirred under 50-60 psi H2 at 40-50 o C for 16 hrs. The reaction mixture was filtered and the filter cake was washed three times with iPrOH (1 V each wash). The solution was distilled at reduced pressure to 5 V, cooled to ambient temperature and seeded (1 wt%). Water (20 V) was charged at 20-25 o C. The resulting slurry was cooled to 3-8 o C for 4-8 hrs. The solids were collected by filtration and washed three times with cold water (1.5 V each wash). The solids were dried at 35-45 o C under reduced pressure to give Compound 5 in 87% yield. 1 H NMR (500 MHz DMSO-d 6 ) δ (ppm): 7.52 (s, 1H), 7.43 (dd, J = 8.4, 7.0 Hz, 1H), 7.13 (s, 1H), 6.97 (ddd, J = 10.9, 7.7, 0.61 Hz, 2H), 6.43 (s, 2H), 4.49 (m, 1H), 2.33 (m, 1H), 2.17 (m, 3H), 1.32 (s, 9H); HPLC purity, 99.2%; Chiral purity, 99.9% ee; LCMS (ESI) m/z 348.2, [M+H] + , 292.2 [Mt-Bu+H] + . Residual IPA: 0.7 mol% by 1 H NMR.

[00252] Synthesis of 4-(1-(4-bromo-3-fluorobenzyl)azetidin-3-yl)morpholine (Compound 4): A mixture of 4-bromo-3-flurobenzaldehyde (Compound 14, 82 g, 396 mmol ) and 4-(azetidin-3-yl)-morpholine hydrochloride (Compound 7 HCl, 72 g, 396 mmol) in acetonitrile (820 ml) was agitated at 25±5°C for at least 3 hours. The mixture was cooled to 10±5°C and sodium triacetoxyborohydride (130 g, 594 mmol) was added in four portions while maintaining the temperature of the mixture below 30°C. The temperature of the mixture was adjusted to 25±5°C and stirred for at least 30 min until reaction completion. The mixture was transferred to a precooled (10-15°C) solution of aqueous citric acid (152 g in 400 ml water, 792 mmol) while maintaining the temperature below 30°C. Once the quenching process was complete, the mixture was concentrated to ~ 560 ml (7 volumes) while keeping the temperature at or below 45°C. The mixture was then washed with toluene (320 ml). To the aqueous phase was added THF and the pH was adjusted to above 12 with aqueous NaOH solution (320 ml, 10 N). The phases were separated, and the aqueous phase was removed. The organic phase was washed with brine and subsequently concentrated with addition of THF (~ 3L) until KF ≤ 0.10%. The mixture was filtered to remove any inorganics and the product Compound 4 was isolated as a solution in THF with 95% yield.

[00253] Synthesis of sodium (2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)phenyl) (hydroxy)methanesulfonate (Compound 13): A solution of Compound 4 (520 g, 1.58 mol) in

THF (380 ml) was cooled to −15 ± 5 °C. A solution of iPrMgCl LiCl (1.3 M, 1823 ml, 2.37 mol) in THF was added over the course of at least 1 hour while maintaining the temperature below −10 °C. After addition was complete, the temperature of the reaction mixture was adjusted to 0 ± 5 °C and stirred for at least 1 hour. Once magnesiation was complete, the mixture was cooled to −15 ± 5 °C (target −15 °C to −20 °C) and a solution of DMF (245 ml g, 3.16 mol) in THF (260 ml) was added slowly over the course of at least 1 hour while maintaining the temperature below −10 °C. The temperature of the mixture was then adjusted to −15 ± 5 °C and agitated for at least 4 hours.

[00254] Upon reaction completion, the reaction mixture was charged into an aqueous 3 N HCl solution (2600 ml) over the course of at least 1 hour while maintaining the temperature below −5 °C. The temperature of the mixture was then adjusted to 5 ± 5 °C and agitation was stopped, letting the mixture settle for at least 15 minutes. The layers were separated. The lower aqueous layer containing the product was washed with 2-MeTHF (2600 ml). The aqueous layer was then charged with 2-MeTHF (2600 ml) and the temperature of the batch was adjusted to −10 ± 5 °C. To the cooled mixture, an aqueous 5 N NaOH (728 ml, 3.64 mol) solution was added while maintaining the temperature below −5 °C until the pH of the mixture was between 10 and 11. The temperature of the mixture was adjusted to 5 ± 5 °C and agitated for at least 15 minutes. The agitation of the mixture was stopped and the mixture allowed to settle for at least 15 minutes. The layers were separated, and the lower aqueous layer was back extracted two times with 2-MeTHF (2600 ml). The combined organic layer was washed with water (1040 mL) and the organic solution was evaporated to dryness, affording 372 g of crude Compound 3 freebase as an oil (yield 85%). 1 H NMR (DMSO-d 6 ) δ (ppm): 10.18 (s, 1H), 7.78 (t, J =7.7 Hz, 1H), 7.23-7.35 (m, 2H), 3.66 (s, 2H), 3.51 -3.60 (m, 4H), 3.26-3.47 (m, 2H), 2.72-2.97 (m, 3H), 2.12-2.32 (m, 4H).

[00255] The crude Compound 3 freebase (4.3 kg) was adsorbed onto silica gel (8.6 kg) with 100% DCM, loaded onto a 60 L column containing 12.9 kg silica gel (packed with 100% DCM), and eluted with DCM ( 86 L), followed successively by 1% MeOH/DCM (40 L), 3% MeOH/DCM (80 L) and 10% MeOH/DCM (40 L). The fractions were collected and concentrated at or below 38˚C to give Compound 3 as a purified oil (3.345 kg, yield 66%).

[00256] A portion of Compound 3 (1.0 kg, 3.59 mol) was dissolved in ethanol (16.0 L, 16

vol) at 20±5 °C and the mixture heated to 40 °C. A solution of Na2S2O5 (622.0 g, 3.27 mol; 0.91 eq) in water (2 L, 2 vol) was prepared at 20±5 °C and added to the freebase solution at 40 °C to obtain an off-white suspension. The batch was agitated and maintained at 40 °C for 2 hrs, then cooled to 20±5 °C and agitated for 1 to 2 hrs. The batch was filtered and washed with ethanol (2×2.0 L, 2×2 vol) to obtain an off-white solid. The wet cake was dried under vacuum at 40 °C for 18 hrs to afford about 1.88 kg of Compound 13.

[00257] Synthesis of 2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzaldehyde (Compound 3): Compound 13 (1.88 kg) was dissolved in ethyl acetate (15.0 L) at 20±5 °C . A 2 M Na 2 CO 3 solution (total 15.0 L used) was added to adjust the pH to 10.0. The batch was agitated for 1 to 1.5 hrs at 20±5 °C. After the reaction was complete, the phases were separated and the organic layer was washed with brine (2.0 L). The organic layer was concentrated to dryness at 35-38 °C to afford 852.0 g of Compound 3 as a colorless oil (yield 81%).

[00258] Synthesis of 2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzaldehyde bis-oxalic acid salt (Compound 3 bis-oxalic acid salt): A portion of the Compound 3 oil (187 g, 0.67 mol) was dissolved in isopropanol (1125 ml) and water (375 ml). A first portion (~30%) of this freebase mixture (480 ml) was slowly added over the course of at least 30 minutes to a solution of oxalic acid (125 g, 1.38 mol) in IPA (1125 ml)/water (375 ml) at 60 ± 5 °C. A second portion (~20%) of the freebase mixture (320 ml) was slowly added over the course of at least 30 minutes to the reaction mixture at 60 ± 5 °C. The reaction mixture was agitated at 60 ± 5 °C for at least 90 minutes. A third portion (~25%) of the freebase mixture (~ 400 ml) was slowly added over the course of at least 30 minutes to the reaction mixture at 60 ± 5 °C and the reaction mixture was agitated at 60 ± 5 °C for at least 90 minutes. The remaining freebase solution (400 ml) was slowly added over the course of at least 30 minutes to the reaction mixture at 60 ± 5 °C and the reaction mixture was agitated at 60 ± 5 °C for at least 90 minutes. The temperature of the mixture was adjusted to 20 ± 5 °C (target 20 °C) over the course of at least 1 hour and the mixture was agitated for at least 16 hours at 20 ± 5 °C and then filtered. The cake was washed three times with IPA (2 x 375 ml) and dried in the drying oven at ≤ 40 °C with a slow bleed of nitrogen to afford 261 g of Compound 3 bis-oxalic acid salt (yield 85%). 1 H NMR (DMSO-d 6 ) δ (ppm): 10.21 (s, 1H), 7.87 (t, J = 7.6 Hz, 1H), 7.42-7.56 (m, 2H), 4.31 (s, 2H), 3.89 -4.03 (m, 2H), 3.75-3.89 (m, 2H), 3.60 (br t, J = 4.3 Hz, 4H), 3.26 (br t, J = 6.9 Hz, 1H), 2.37 (br s, 4H) .

[00259] Synthesis of tert-butyl (S)-5-amino-4-(4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)-1,3- dioxoisoindolin-2-yl)-5-oxopentanoate (Compound 2):

Acetonitrile (6.8 L, 8.0 X Vol) was added to a 30 L jacketed cylindrical reactor. Compound 5 (0.845 kg, 1.00 X Wt) and Compound 3 bis-oxalic acid salt (1.35 kg, 1.60 The contents of the reactor were equilibrated with agitation at 20 ± 5 °C. Trifluoroacetic acid (0.19 L, 0.22 X Vol) was added dropwise, maintaining the batch temperature at 20 ± 5 °C. The reaction mixture was stirred at 20 ± 5 °C for no less than 5 minutes and then sodium triacetoxyborohydride (0.13 kg, 015 X Wt) was added as a solid, maintaining the batch temperature at 20 ± 5 °C. The process of adding trifluoroacetic acid and then sodium triacetoxyborohydride was repeated an additional 5 times. After the last addition, the reaction mixture was sampled to determine the reaction progress. The reaction was held at 20 ± 5 °C overnight. The reaction mixture was then quenched with water (3.4 L, 4.0 X Vol), maintaining the batch temperature at 20 ± 5 °C. The mixture was then stirred at 20 ± 5 °C for no less than 30 minutes and the resulting slurry filtered through a 3 L sintered glass filter, directing the filtrates to clean containers. The reactor was rinsed with acetonitrile (0.4 L, 0.5 X Vol) and the rinse passed through the contents of the 3 L sintered glass filter, directing the filtrate to the containers containing the main batch. The contents of the containers were concentrated to ~5 X Vol under reduced pressure at a bath temperature of no more than 30 °C. The residue was transferred to a clean reactor, was rinsing with 2-MeTHF (2.5 L, 3.0 X Vol) to complete the transfer. Additional 2-MeTHF (10.1 L, 12.0 X Vol) was added to the reactor, followed by water (3.4 L, 4.0 X Vol). The mixture was agitated for no less than 15 minutes at 20 ± 5 °C, then allowed to settle for no less than 10 minutes at 20 ± 5 °C before transferring the bottom aqueous layer to new containers. An aqueous sodium bicarbonate solution (5.3 L, 6.3 X Vol, 9% wt/wt) was added to the reactor with stirring over 30 minutes, maintaining batch temperature no more than 25 °C. The mixture was agitated for no more than 15 minutes at 20 ± 5 °C, then allowed to settle for no less than 10 minutes at 20 ± 5 °C before the bottom aqueous layer was transferred to new containers. The aqueous sodium bicarbonate wash was repeated an additional 2 times to reach a pH of about 6.6 for the spent aqueous layer. A saturated aqueous solution of NaCl(0.85 L, 1.0 X Vol) was then added to reactor with agitation. The mixture was agitated for no less than 15 minutes at 20 ± 5 °C, then allowed to settle for no less than 10 minutes before the bottom aqueous layer was transferred to new containers. The remaining organics were concentrated under reduced pressure to a batch volume of ~5 X Vol at a bath temperature of about 40 °C. Acetonitrile (5.1 L, 6.0 X Vol) was added to the residual volume and the resulting solution concentrated to a batch volume of ~ 5 X Vol under reduced pressure at bath temperature of about 40 °C. The process of adding acetonitrile and concentrating under vacuum was repeated two more times to reach the distillation endpoint with a water content of about 1%. The acetonitrile solution was transferred to a clean container along with two 1.7 L (2.0 X Vol) rinses and held at 5 °C overnight. The acetonitrile solution was then filtered through a 3 L sintered glass filter, followed by a 1.7 L (2.0 X Vol) acetonitrile rinse, directing the filtrates to a clean container. The filtrate was transferred to a clean reactor and the container rinsed twice with 1.7 L (2.0 X Vol) of acetonitrile to complete the transfer. Enough acetonitrile (roughly 0.6 L) was added to adjust the total volume in the reactor to about 14 L. A solution assay of the contents of the reactor was obtained to calculate the amount of Compound 2 present for use in the next step (result = 1.3 kg = 1.00 X Wt for remainder of process).

[00260] Synthesis of (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline -1,3-dione bis-besylate salt (Compound 1 bis-besylate salt): The solution of Compound 2 in acetonitrile from the previous step was diluted with acetonitrile (roughly 2 L) such that the total volume in the reactor was about 16 L. The solution was cooled with stirring to 10 ± 5 °C and held within that range for 96 hours. Benzenesulfonic acid (1.86 kg, 1.43 X Wt) was added while sparging the reaction mixture with nitrogen gas and maintaining the batch temperature at 10 ± 10 °C. The temperature of the reactor was then adjusted to 20 ± 5 °C and the mixture stirred at that temperature for 60 minutes. The total volume of reaction mixture was adjusted back to 16 L to account for solvent lost during sparging by the addition of acetonitrile (roughly 0.4 L). The reaction mixture was then heated to 55 ± 5 °C over the course of about 30 minutes and held in that range for 15 to 16 hours for reaction completion. The mixture was then cooled to 50 ± 5 °C and MTBE (3.9 L, 3.0 X Vol) was added, maintaining the batch temperature at 50 ± 5 °C. The mixture was allowed to stir at 50 ± 5 °C for about 1.5 hours to establish a self-seeded slurry. Additional MTBE (3.9 L, 3.0 X Vol) was added to the reactor over the course of about 1.75 hours at 50 ± 5 °C. The slurry was cooled to 20 ± 5 °C over the course of about 1.75 hours and held in that temperature range overnight. The slurry was filtered using a Buchner funnel. The reactor was rinsed twice with

MTBE (3.9 L each, 3.0 X Vol) and the rinse was used to wash the solids in the Buchner funnel. The solids were dried on drying trays for about 23 hours at 40 °C under reduced pressure (15-150 mbar), yielding 1.62 kg (77.9%) of Compound 1 bis-besylate salt.

[00261] Synthesis of (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline -1,3-dione hydrochloride salt (Compound 1 HCl): A suspension of Compound 1 bis-besylate salt (120 g, 1 equiv.) in 2-MeTHF (25 L/kg) was added to a reactor and agitated at 10 °C. A solution of KHCO 3 (32.5 g, 2.4 equiv) in water (1.8 L, 6 L/kg) was added to the slurry over the course of 40 minutes. The mixture was stirred for an additional 30 minutes. The batch was then allowed to settle, at which point the aqueous (bottom) layer was separated and discarded. An aqueous solution of NaCl (5%, 5 L/kg, 575 ml) was added to the organic layer and the mixture was agitated for 10 minutes, after which point the temperature was raised to 20 °C. The batch was allowed to settle, at which point the aqueous (bottom) layer was discarded. The brine was repeated a second time.

Additional 2-MeTHF (500 ml) was added to dilute the organic layer, resulting in a concentration of about 20 mg product per ml. A solution of HCl (total 0.98 eq.) in 2-MeTHF was prepared and a portion (20% of total, corresponding to ~0.2 eq.) then added to the reaction mixture over the course of about 10 min. Seeds of Compound 1 hydrochloride (~5% wt) were added, but did not dissolve. The batch was held under vigorous agitation for one hour. To the slurry, the remaining portion of the HCl solution (~0.78 eq.) was added over the course of 3 hours at a constant rate. Vigorous agitation was maintained. After addition was complete, the batch was held for one hour, after which the batch was filtered, washed three times with 3 L/kg of 2-MeTHF . The filter cake was placed in a vacuum oven at 22 °C for 12 hours, at which point the temperature was raised to 40 °C. Dry cake of Compound 1 hydrochloride (58g, 75% yield) was obtained and packaged. Achiral HPLC purity: 98.91%; chiral HPLC purity: 99.68%.

Example 4: Synthesis of (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3- morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline -1,3-dione

[00264] Synthesis of tert-butyl (S)-5-amino-4-(4-nitro-1,3-dioxoisoindolin-2-yl)-5-oxopentanoate (Compound 6): To a solution of 3-nitrophthalic anhydride (Compound 12,

35.15 g, 176.6 mmol, 1.00 eq) in ethyl acetate (350 mL) was added tert-butyl (4S)-4,5-diamino-5-oxo-pentanoate hydrochloride (Compound 9 HCl, 43.22 g, 181.1 mmol, 1.025 eq) ), DMF (70

mL) and 2-MeTHF (110 mL) at 25°C. 2,6-Lutidine (23.4 mL, 201 mmol, 1.14 eq) was added

slowly to maintain the temperature at or below 25°C. The mixture was aged at 25°C for 1 hour before being cooled to 5°C. CDI (4.17 g, 25.7 mmol, 0.146 eq) was added and stirred until the temperature returned to 5°C. Another portion of CDI (4.62 g, 28.5 mmol, 0.161 eq) was added and stirred until temperature returned to 5°C. CDI (8.87 g, 54.7 mmol, 0.310 eq) was added and stirred until the temperature returned to 5°C. CDI (8.91 g, 54.9 mmol, 0.311 eq) was added and stirred until the temperature returned to 5°C. The mixture was warmed to 20°C and CDI (16.4 g, 101.1 mmol, 0.573 eq) was added, and the mixture was aged at 20°C for 16 hours. The mixture was cooled to 5°C and a solution of 30 wt% citric acid and 5 wt% NaCl (350 mL) was added slowly while maintaining the temperature. The mixture was warmed to 20°C and aged for 30 minutes. The phases were split and separated. The organic phase was diluted with EtOAc (175 mL) and washed with a solution of 5 wt% citric acid (175 mL), and concentrated by distillation (75 torr, 50°C) to a volume of 175 mL EtOAc. The solvent was changed to iPrOH by constant volume distillation (75 torr, 50°C) with 350 mL iPrOH to a final volume of 175 mL. The distillate was diluted with 200 mL iPrOH to afford Compound 6 as a solution for use in the next step. 1 H NMR (500 MHz, CDCl 3 ) δ (ppm): 8.18 – 8.13 (m, 2H), 7.96 (t, J = 7.8 Hz, 1H), 6.34 (s, 1H), 5.59 (s, 1H), 4.90 (dd, J = 10.1, 4.6 Hz, 1H), 2.61 (ddt, J = 14.6, 10.1, 6.1 Hz, 1H), 2.49 (ddt, J = 14.2, 8.7, 5.2 Hz, 1H), 2.44 – 2.29 ( m, 2H), 1.44 (s, 9H).

[00265] Synthesis of tert-butyl (S)-5-amino-4-(4-amino-1,3-dioxoisoindolin-2-yl)-5-oxopentanoate (Compound 5): To a solution of Compound 6 in iPrOH (375 mL) was added 5% palladium on carbon (1.23 g, 3.5 wt%, wet). The mixture was purged with nitrogen five times and with hydrogen three times. The mixture was pressurized with hydrogen (50 psi) and aged at 50°C for 16 hours. The mixture was cooled to room temperature and purged with nitrogen three times, filtered to remove catalyst, and the filter cake was washed with iPrOH (20mL) three times. The filtrate was concentrated to 200 mL, seeded (0.454 g, 1.3 wt%) at 22 °C, and aged for 45 minutes. Water (1325 mL) was added over 3 hours at 22°C. After the addition of water, the mixture was cooled to 8°C over 2 hours and aged for 1 hour at 8°C. The slurry was filtered, and the cake was rinsed with cold water (200 mL) three times and dried under vacuum at 50°C to yield Compound 5 as a yellow solid (47.97 g, 80.6% yield, 99.62% LC purity, 103% 1 H NMR potency). 1 H NMR (500 MHz, CDCl 3 ) δ (ppm): 7.46 (dd, J = 8.3, 7.0 Hz, 1H), 7.19 (d, J = 7.2 Hz, 1H), 6.89 (d, J = 8.3 Hz, 1H), 6.28 (s, 1H), 5.41 (s, 1H), 5.28 (s, 2H), 4.83 (dd, J = 9.3, 6.0 Hz, 1H), 2.52 (p, J = 7.0 Hz, 2H), 2.36 – 2.29 (m, 2H), 1.44 (s, 9H). 13 C NMR (126 MHz, CDCl 3 ) δ (ppm): 171.80, 171.12, 169.64, 168.27, 145.70, 135.50, 132.20, 121.43, 112.98, 80.99, 53.04, 32.23, 28.02 , 24.36. LCMS (ESI): m/z 291.9 [M+H – tBu]

[00266] Synthesis of 4-(1-(4-bromo-3-fluorobenzyl)azetidin-3-yl)morpholine bis-methanesulfonic acid salt (Compound 4 bis-methanesulfonic acid salt): A mixture of 4-bromo-3- fluorobenzaldehyde (Compound 14, 102 g, 493 mmol) and 4-(azetidin-3-yl)morpholine hydrochloride (Compound 7 HCl, 90 g, 493 mmol) in acetonitrile (1000 ml) was agitated at a temperature of about 20 to 25 °C for 2 to 3 hours. The slurry was cooled to temperature of about 10 to 15 °C and sodium triacetoxyborohydride (STAB, 162 g, 739 mmol) was added in 4 portions over the course of about 45 minutes while maintaining the batch temperature at no more than 30 °C. The slurry was stirred at a temperature of about 20 to 25 °C for at least 30 minutes and then quenched by an aqueous citric acid solution (191 g, 986 mmol in 500 ml of water) at a temperature of about 40 to 45 °C over the course of 2 hours. Upon completion of the quenching process, the batch volume was reduced by vacuum distillation to about 700 ml at a temperature of no more than 45 °C. Cyclopentylmethylether (CPME, 400 ml) was added to the aqueous solution to afford a final volume of about 1100 ml. The pH was adjusted to about 8 to 9 by addition of an aqueous solution of 10 N NaOH (added volume about 430 ml). The phases were separated, and the aqueous phase discarded. The organic phase was washed with brine (100 ml) twice such that the pH was no more than 8 and the volume was adjusted to about 1000 ml with addition of extra CPME. The batch was distilled at constant volume under reduced pressure with addition of CPME until KF was no more than 0.15%. CPME was added (if needed) to adjust the batch to a volume of 1000 ml at the end of distillation. The dry CPME solution was seeded (500 to 750 mg) at ambient temperature. The seeded, dry CPME slurry was heated to a temperature of 50 to 60 °C and then charged with methanesulfonic acid in 200 ml of CPME over the course of 4 to 5 hours. The slurry was then cooled to 20 °C over the course of 4 to 5 hours and kept at 20 °C for 3 to 4 hours, filtered, rinsed with CPME and dried in a vacuum oven at 35 to 40 °C over 16 hours to give Compound 4 bis-methanesulfonic acid salt as a white solid. 1 H NMR (500 MHz DMSO-d 6 ) δ (ppm): 10.62 (br s, 1-2H), 7.85 (t, J = 7.8 Hz, 1H), 7.58 (dd, J = 9.5 Hz, 1.9 Hz, 1H), 7.34 (dd, J = 8.2 Hz, 1.8 Hz, 1H), 4.55-4.24 (m, 7H), 3.84 (br s, 4H), 3.14 (m, 4H); HPLC purity, 99.8%, LCMS (ESI) m/z 329.1 /331.1[M/M+2] + . XRPD pattern of the product is shown in FIG.10. DSC thermogram of the product is shown in FIG.11.

[00267] Preparation of 4-(1-(4-bromo-3-fluorobenzyl)azetidin-3-yl)morpholine (Compound 4): A slurry of Compound 4 bis-methanesulfonic acid salt (70 g, 134 mmol) in t -butyl methyl ether was cooled to 10±5°C. An aqueous solution of NaOH (2 N, 201 ml, 403 mmol) was added over the course of at least 30 minutes while maintaining the batch temperature at about 15 °C. After the addition of NaOH , the batch temperature was raised to 20±5°C and agitated over the course of about 20 minutes. The organic layer was separated and washed with water (210 ml) three times. The organic layer was subsequently concentrated with addition of THF (~ 1.05L) until KF ≤ 0.10%. The product Compound 4 was isolated as a solution in THF

with 95% solution yield.

[00268] Preparation of 2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzaldehyde dihydrochloride (Compound 3 di-HCl): A solution of Compound 4 (44 g, 134 mmol) in THF (total volume ~ 350 ml) was then cooled to −20 ± 5 °C. A solution of iPrMgCl LiCl (1.3 M, 176 ml, 228 mmol) in THF was added over the course of half an hour while maintaining the temperature below −10 °C. After the addition was complete, the batch was stirred at −20 ± 5 °C for 16 to 22 hours. DMF (21 ml, 268 mmol) was then added slowly over the course of 30 minutes while maintaining the batch temperature no more than -15 °C. The batch was stirred at −20 ± 5 °C for 6 to 24 hours. 2-MeTHF (350 ml) was then added to the batch over the course of 30 minutes, followed by slow addition of 3 N HCl (235 ml, 704 mmol) while keeping the batch temperature no more than -10 °C. After the addition of aqueous HCl, the batch was warmed to 0 ± 5 °C and 2 N aqueous NaOH (154 ml, 309 mmol) was added slowly to adjust the solution pH to about 8 to 9. The batch was stirred for about 30 minutes and then warmed to 20 ± 5 °C. The organic layer was separated and washed with 15% aqueous NaCl (3 x 140 ml). The organic layer was subsequently concentrated with addition of 2-MeTHF until KF ≤ 0.10%.

[00269] A portion of the free base of 2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzaldehyde (37.4 g, 134 mmol) so obtained was dissolved in 2-MeTHF (total ~ 420 ml) , to which isopropanol (420 ml) and water (21 ml) were added at 20 ± 5 °C. The batch was then heated to 50 ± 5 °C and a solution of HCl in IPA (5 to 6 N, 28 ml, half of total HCl volume) was added over the course of 1 hour. The batch was seeded with 2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzaldehyde dihydrochloride (700 mg) and aged for 1 hour. The remaining HCl (28 ml) was then added over the course of 1 hour. The batch was agitated at 50 ± 5 °C for 4 hours and then cooled to 20 ± 5 °C for 8 hours. The slurry was filtered, washed with IPA (210 ml), and the filter cake dried under vacuum at 50 ± 5 °C to afford Compound 3 dihydrochloride salt (36 g, yield 75%). 1 H NMR (DMSO-d 6 ) δ (ppm): 12.32-12.55 (m, 1H), 10.23 (s, 1H), 7.93 (t, J =7.6 Hz, 1H), 7.66 (d, J = 10.5 Hz , 1H), 7.58 (d, J = 7.9 Hz, 1H), 4.80 (br s, 2H), 4.48-4.70 (m, 2H), 4.30 (br s, 4H), 3.78-4.00 (m, 5H), 2.93-3.15 (m, 2H). Two polymorphic forms were obtained. XRPD pattern and DSC thermogram of Form A (anhydrous) are shown in FIG.5 and FIG.6, respectively. XRPD pattern, TGA thermogram and DSC thermogram of Form B (hydrate) are shown in FIG.7, FIG.8, and FIG.9, respectively.

[00270] Synthesis of tert-butyl (S)-5-amino-4-(4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)-1,3- dioxoisoindolin-2-yl)-5-oxopentanoate (Compound 2): A mixture of Compound 5 (12 g, 34.5 mmol, 1.0 eq) and Compound 3 dihydrochloride (14.56 g, 41.5 mmol, 1.2 eq) in MeCN (96 ml) was cooled to 0-5 °C. Trifluoroacetic acid (TFA, 2.0 ml, 26 mmol, 0.75 eq) was added, followed by sodium triacetoxyborohydride (STAB, 2.75 g, 12.95 mmol, 0.375 eq) while maintaining the internal temperature below 10 °C. The addition of TFA and STAB was repeated three additional times. After a total of four additions of TFA and STAB, the reaction was aged at 0-5°C for 1 hour. A 10% brine solution (108 ml) was then added to the reaction mixture over the course of 1 hour and partitioned with IPAc (96 ml). The mixture was warmed to 20-25 °C and aged for 30 minutes. The layers were then separated and the organic layer was washed with 2.0 M K3PO4 (114 ml). The pH of the spent aqueous layer should have a pH of about 8.5 – 9.0. The layers were separated again and the organic phase was washed with 8.5% NaHCO 3 (2 x 60 ml), with 30 minutes between each wash, followed by 24% brine (60 ml). The organic fraction was distilled to 72 ml at an internal temperature near 50°C. Toluene (72 ml) was added to bring the volume to 144 ml and distillation continued at constant volume at 50°C with feed and bleed until water content < 0.1. The mixture was heated to 50°C and acetonitrile (48 ml) was added, followed by slow addition of heptane (144 ml) while maintaining the internal temperature above 45 °C. The reaction was held at 50 °C for 2 hours. Once complete, the reaction was slowly cooled to 20-25°C over the course of 4 hours and held at 20-25°C overnight (16 hour). The yellow slurry was then filtered and the yellow cake displacement washed with 1:3:3 mixture of acetonitrile/heptane/toluene (3 x 48 ml). The final cake was then dried under reduced pressure at 50 °C under nitrogen to provide Compound 2 (87.7% isolated molar yield) with >99.0% LCAP. HPLC purity, 99.85%; Chiral purity, >99.9% ee. 1 H NMR (DMSO-d6, 500 MHz) δ (ppm) 7.55 (s, 1H), 7.51 (dd, J = 7.2, 8.4 Hz, 1H), 7.32 (t, J = 7.9 Hz, 1H), 7.16 ( s, 1H), 7.0-7.1 (m, 5H), 4.57 (d, J = 6.3 Hz, 2H), 4.5-4.5 (m, 1H), 3.5-3.6 (m, 6H), 3.3-3.4 (m, 3H), 2.8-2.9 (m, 3H), 2.3-2.4 (m, 1H), 2.1-2.3 (m, 7H), 1.31 (s, 9H); LCMS m/z 610.3 [M+H] + .

[00271] Synthesis of (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline -1,3-dione bis-besylate salt (Compound 1 bis-besylate): To a suspension of Compound 2 (130 g, 1.0 equiv.) in MeCN (1.56 L, 12 L/kg) agitated at 55 °C was added a solution of benzene sulfonic acid (185 g, 5.5

equiv.) in MeCN (0.39 L, 3 L/kg) and water (0.01 L, 2.0 equiv.). The mixture was stirred at 55 °C for 16 hours. After the reaction age, crystalline seeds (1.3 g, 1 wt%) of bis-besylate salt of Compound 1 were charged into the batch, resulting in formation of a yellow slurry. The slurry was then cooled to 20 °C over the course of 90 minutes. 2-MeTHF (1.3 L, 10 L/kg) was added to the batch slowly over 2 hours at 20 °C. The batch was agitated for an additional 4 hours at 20°C. The yellow slurry was then filtered and the yellow cake re-slurried with MeTHF (1.3 L, 10 L/kg) followed by a displacement MeTHF (0.65 L, 5 L/kg) wash. The final cake was then dried under reduced pressure at 50 °C under nitrogen to give Compound 1 bis-besylate salt (160 g, 88.4% yield). HPLC purity: 98.39%; chiral HPLC purity: 100%. XRPD pattern, TGA thermogram and DSC thermogram of the product are shown in FIG.1, FIG.2, and FIG.3, respectively.

[00272] Synthesis of (S)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline -1,3-dione hydrochloride salt (Compound 1 HCl): A suspension of Compound 1 bis-besylate salt (300 g, 1 equiv.) in EtOAc (4.68 L, 15.6 L/kg) and 2-propanol (0.12 L, 0.4 L/kg) was agitated at 15 °C. To the suspension was added a solution of KHCO 3 (82.4 g, 2.5 equiv) in water (1.8 L, 6 L/kg) over 30 minutes. The mixture was heated to 20 °C over 30-60 minutes and then agitated for 30 minutes. The batch was allowed to settle for 30 minutes, at which point the aqueous (bottom) layer was discarded. To the rich organic layer was added water (1.2 L, 4 L/kg) and the reactor contents were agitated for 30 minutes. The batch was allowed to settle for 30 minutes, at which point the aqueous (bottom) layer was discarded. To the rich organic stream was added 2-propanol (2.375 L, 7.9 L/kg) and the stream was then filtered. Water was added to the filtrate to adjust the water content to 8≤KF≤8.2. To the above agitated solution at 20 °C was added 0.2 N HCl (38 mL, 0.025 equiv prepared in EtOAC/IPA 2:1, v/v with 8wt% water) over 10 minutes. To the mixture was added crystalline seeds of Compound 1 hydrochloride salt (1.6 g, 0.5wt%) and the contents of the reactor were agitated at 20 °C for 30 minutes. To the suspension was added 0.2 N HCl (1.44 L, 0.945 equiv. prepared in EtOAC/IPA 2:1, v/v with 8 wt% water) over 4.5 hours. The slurry was agitated for 14 hours, then filtered and washed with EtOAC/IPA (750 mL, 2.5 L/kg, 2:1 v/v with 8 wt% water) followed by IPA (750 mL, 2.5 L/kg). The solids were dried under vacuum at 40 °C to afford Compound 1 hydrochloride salt (170 g, 90% yield). Achiral HPLC purity: 99.91%; chiral HPLC purity: 99.58%. XRPD analysis (FIG.4) confirmed the product (a)

str1
Flag Counter

AS ON DEC2021 3,491,869 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

//////////Golcadomide, CC 99282, CC 1007548, WHO 12305

C1CC(=O)NC(=O)C1N2C(=O)C3=C(C2=O)C(=CC=C3)NCC4=C(C=C(C=C4)CN5CC(C5)N6CCOCC6)F

Igermetostat


structure image
Igermetostat Structure
Igermetostat.png

Igermetostat

C32H46N4O4. 550.73

2409538-60-7

2KES6YSH8D

INN 12655

BENZAMIDE, N-((1,2-DIHYDRO-4,6-DIMETHYL-2-OXO-3-PYRIDINYL)METHYL)-3-(ETHYL(TETRAHYDRO-2H-PYRAN-4-YL)AMINO)-2-METHYL-5-((TRANS-3-(1-PIPERIDINYL)CYCLOBUTYL)OXY)-
IGERMETOSTAT
IGERMETOSTAT [INN]
N-((1,2-DIHYDRO-4,6-DIMETHYL-2-OXO-3-PYRIDINYL)METHYL)-3-(ETHYL(TETRAHYDRO-2H-PYRAN-4-YL)AMINO)-2-METHYL-5-((TRANS-3-(1-PIPERIDINYL)CYCLOBUTYL)OXY)BENZAMIDE
N-((4,6-DIMETHYL-2-OXO-1,2-DIHYDROPYRIDIN-3-YL)METHYL)-3-(ETHYL(OXAN-4-YL)AMINO)-2-METHYL-5-(((1R,3R)-3-(PIPERIDIN-1-YL)CYCLOBUTYL)OXY)BENZAMIDE

PATENT

PATENT

US20230382898

The compound has been disclosed in patent WO2020020374A1. It is an enhancer of Zeste homolog 2 (EZH2) inhibitor, and can be used for preventing or treating EZH2-mediated diseases, including brain cancer, thyroid cancer, cardiac sarcoma, lung cancer, oral cancer, stomach cancer and various other cancers.

PATENT

US20210308141

Example 4: Preparation of the Compound N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-3-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-2-methyl-5-(trans-3-morpholinylcyclobutoxy)benzamide (4)


(MOL)(CDX)
      The title compound was prepared by referring to Example 1 with cyclobutanol replaced by cis-3-morpholinylcyclobutyl-1-ol. The cis-3-morpholinylcyclobutyl-1-ol was prepared as follows:
      3-benzyloxy-cyclobutyl-1-one (3 g, 17 mmol, 1 eq) and morpholine (2.97 g, 34 mmol, 2 eq) were dissolved in DCE (30 mL). After stirring at room temperature for 0.5 h, the solution was added with NaBH(OAc)3 (7.23 g, 34 mmol, 2 eq) and stirred at room temperature for 2 h. The reaction mixture was concentrated under reduced pressure to remove the solvent, adjusted to basicity with aqueous NaHCO3 solution, and extracted with ethyl acetate. The organic phase was washed with brine, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a crude product, which was purified by a phase preparative column to give 4-(3-(benzyloxy)cyclobutyl)morpholine (2.9 g). M/z (ES+), [M+H] +=248.
      4-(3-(benzyloxy)cyclobutyl)morpholine (2.9 g, 117.4 mmol) and methane sulfonic acid (20 mL) was added to dichloromethane (40 mL). The mixture was stirred overnight at room temperature and concentrated under reduced pressure to remove the solvent to give a crude product, which was purified by a preparative column to give cis-3-morpholinylcyclobutyl-1-ol; m/z (ES+), [M+H] +=158. The trans-3-morpholinylcyclobutyl-1-ol can also be separated and obtained by this method.
      m/z (ES +), [M+H] +=553; HPLC t R=1.296 min. 1H NMR (400 MHz, DMSO-d 6) δ 11.53-11.42 (s, 1H), 8.08-8.00 (s, 1H), 6.63-6.58 (d, J=2.4 Hz, 1H), 6.44-6.38 (d, J=2.5 Hz, 1H), 5.89-5.84 (s, 1H), 4.28-4.22 (d, J=4.7 Hz, 2H), 3.63-3.57 (d, J=4.0 Hz, 4H), 3.29-3.17 (t, J=10.5 Hz, OH), 3.02-2.87 (m, OH), 2.39-2.25 (d, J=19.9 Hz, 6H), 2.22-2.17 (s, 3H), 2.15-2.03 (d, J=2.9 Hz, 8H), 1.67-1.57 (d, J=10.7 Hz, 2H), 1.53-1.42 (d, J=8.4 Hz, 2H), 0.82-0.76 (t, J=6.9 Hz, 3H). No isomerization was found in chiral HPLC.

[1]. Peng, et al. Polysubstituted benzene compound and preparation method and use thereof.

World Intellectual Property Organization, WO2020020374 A1. 2020-01-30.

//////////Igermetostat

CCN(C1CCOCC1)c2cc(O[C@H]3C[C@@H](C3)N4CCCCC4)cc(C(=O)NCc5c(C)cc(C)[nH]c5=O)c2C

Oditrasertib,


Obeldesivir.png

Obeldesivir,

2647441-36-7

361.35 g/mol

C16H19N5O5

[(2R,3S,4R,5R)-5-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxyoxolan-2-yl]methyl 2-methylpropanoate

Q55KCM7PXB; ATV006; UNII-Q55KCM7PXB

Obeldesivir (GS-5245ATV006) is an isobutyric ester prodrug of GS-441524 made by Gilead Sciences that is currently in Phase III trials for the outpatient treatment of COVID-19 in high risk patients.[1][2] The purpose of the isobutyric ester modification on obeldesivir is to improve the oral bioavailability of the parent nucleoside, GS-441524. Obeldesivir is hydrolyzed to its parent nucleoside, GS-441524, which is in turn converted to remdesivir-triphosphate (GS-443902) by a nucleoside kinase, adenylate kinase and nucleotide diphosphate kinase.[3][4]

GS-443902 is a bioactive ATP analogue with broad-spectrum antiviral activity [5] and is the same compound formed by remdesivir, though by a different enzymatic pathway. Unlike remdesivir, which is metabolized by enzymes that are highly expressed in the liver, GS-441524 released by obeldesivir is metabolized by enzymes that are evenly expressed throughout the body. Due to their different metabolic pathways, obeldesivir can be administered orally, whereas remdesivir must be administered intravenously for COVID-19 treatment.[citation needed]

The pharmacokinetic properties of obeldesivir and improved was first published by Chinese researchers in May 2022. The Chinese group pursued investigation of obeldesivir independently from Gilead Sciences. Compared to IV administered GS-441524 in rats at 5 mg/kg, orally administered obeldesivir at 25 mg/kg (referred to as “ATV006”) yielded approximately 22% bioavailability.[6] Treatment with obdeldesivir reduced viral load and prevents lung pathology in KI-hACE2 and Ad5-hACE2 mouse models of SARS-CoV-2. A patent filed by Gilead Sciences with a priority date of August 27, 2020,[7] found the bioavailability of GS-441524 after oral administration of obdeldesivir (compound 15) in mice, rats, ferrets, dogs, and cynomolgus macaques to be 41%, 63.9%, 154%, 94%, and 38%, respectively. Across all species evaluated, obeldesivir showed improved oral bioavailability compared to oral administration of the parent nucleoside, GS-441524

PAPER

https://pubs.acs.org/doi/10.1021/acs.jmedchem.3c00750

Abstract

Remdesivir 1 is an phosphoramidate prodrug that releases the monophosphate of nucleoside GS-441524 (2) into lung cells, thereby forming the bioactive triphosphate 2-NTP2-NTP, an analog of ATP, inhibits the SARS-CoV-2 RNA-dependent RNA polymerase replication and transcription of viral RNA. Strong clinical results for 1 have prompted interest in oral approaches to generate 2-NTP. Here, we describe the discovery of a 5′-isobutyryl ester prodrug of 2 (GS-5245, Obeldesivir, 3) that has low cellular cytotoxicity and 3–7-fold improved oral delivery of 2 in monkeys. Prodrug 3 is cleaved presystemically to provide high systemic exposures of 2 that overcome its less efficient metabolism to 2-NTP, leading to strong SARS-CoV-2 antiviral efficacy in an African green monkey infection model. Exposure-based SARS-CoV-2 efficacy relationships resulted in an estimated clinical dose of 350–400 mg twice daily. Importantly, all SARS-CoV-2 variants remain susceptible to 2, which supports development of 3 as a promising COVID-19 treatment.

Synthesis of ((2R,3S,4R,5R)-5-(4-Aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methyl Isobutyrate (3)

To a solution of (3aR,4R,6R,6aR)-4-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-6-(hydroxymethyl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxole-4-carbonitrile 4 (2000 mg, 6.0 mmol) (30) and isobutyric acid (638 mg, 7.2 mmol) in DMF (5 mL), N,N′-diisopropylcarbodiimide (914 mg, 7.2 mmol) was slowly added followed by 4-(dimethylamino)pyridine (737 mg, 6.0 mmol). The reaction mixture was stirred for 4 h and then diluted with ethyl acetate, washed with water and brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was subjected to silica gel chromatography, eluting with 20% MeOH in CH2Cl2 to provide the intermediate ((3aR,4R,6R,6aR)-6-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-6-cyano-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4-yl)methyl isobutyrate. LCMS m/z = 402.2 (M + 1). To a solution of the intermediate (1500 mg) in THF (10 mL), conc. HCl (2 mL) was added, and the mixture was stirred at rt for 4 h. The reaction mixture was diluted with CH2Cl2, washed with water, saturated aqueous bicarbonate, and brine, dried over sodium sulfate, concentrated, and subjected to silica gel chromatography, eluting with 30% MeOH in CH2Cl2 to afford the title compound (660 mg, 50%). 1H NMR (400 MHz, MeOH-d4) δ 7.88 (s, 1H), 6.96–6.85 (m, 2H), 4.50–4.27 (m, 4H), 4.16 (dd, J = 6.2, 5.3 Hz, 1H), 2.56 (p, J = 7.0 Hz, 1H), 1.14 (dd, J = 7.0, 3.8 Hz, 6H). LCMS m/z: 362.1 (M + 1). 13C NMR (400 MHz, CHCl3d3) δ 175.9, 155.6, 147.9, 123.5, 110.2, 100.8, 116.9, 116.6, 81.3, 79.0, 74.0, 70.2, 62.9, 33.2, 18.7, 18.6. HRMS m/z: 362.14615, C16H20N5O5 362.14644.

PATENT

WO-2022047065-A2

PATENT

Example 1. (3aR, 4R, 6R, 6aR)-4-(4-aminopyrrole[2,1-f][1,2,4]triazin-7-yl)-6-(hydroxymethyl- Synthesis of 2,2-dimethyltetrahydrofuran[3,4-d][1,3]dioxolyl-4-carbonitrile (compound 5)
      
        Dissolve 5.62g of compound 69-0 in 30mL of acetone, then add 11.50mL of 2,2-dimethoxypropane and 1.34mL of 98% sulfuric acid, stir at 45°C for half an hour, cool to room temperature, and remove by rotary evaporation Organic solvents. Extract with 100 mL of ethyl acetate and 100 mL of saturated sodium bicarbonate solution. Repeat the extraction three times. Combine the ethyl acetate layers, add anhydrous sodium sulfate to dry, and filter to remove sodium sulfate. The organic solvent was removed by rotary evaporation, and 6.20 g of compound 5 (white solid, yield 97%) was obtained through column chromatography (eluent: petroleum ether/ethyl acetate (v/v) = 1/2). The hydrogen spectrum of the obtained compound 5 was detected, and the results are as follows:
        氢谱:1H NMR(400MHz,Chloroform-d)δ7.95(s,1H),7.11(d,J=4.7Hz,1H),6.69(dd,J=4.8,2.4Hz,1H),5.77(s,2H),5.42(d,J=6.6Hz,1H),5.24(dd,J=6.6,2.4Hz,1H),4.67(q,J=1.9Hz,1H),3.99(dd,J=12.5,1.9Hz,1H),3.84(dd,J=12.5,1.7Hz,1H),1.81(s,3H),1.40(s,3H)。
        Example 2. ((3aR,4R,6R,6aR)-6-(4-aminopyrrole[2,1-f][1,2,4]triazin-7-yl)-6-cyano-2 ,Synthesis of 2-dimethyltetrahydrofuran[3,4-d][1,3]dioxol-4-yl)methylisobutyrate (compound INT-1)
        
        Dissolve 1.50g of compound 5 in 15ml of dichloromethane, then add 0.42mL of isobutyric acid and 55.40mg of 4-dimethylaminopyridine, stir for 10 minutes, add 1.02g of dicyclohexylcarbodiimide, and stir at room temperature. Stir for 24h. After column chromatography separation (eluent: petroleum ether/ethyl acetate (v/v) = 1/1), 1.71 g of compound INT-1 (white solid, yield 94%) was obtained. The hydrogen spectrum and carbon spectrum of the obtained compound INT-1 were detected, and the results are as follows:
        Hydrogen spectrum: 1 H NMR (400MHz, CDCl 3 ,ZQF-RD01-2)δ(ppm):7.99(s,1H),6.99(d,J=4.6Hz,1H),6.62(d,J=4.6Hz,1H),5.72(br,2H),5.49(d,J=6.8Hz,1H),4.93-4.90(dd,J=6.8Hz,4.3Hz,1H),4.61-4.58(q,J=4.4Hz,1H),4.44-4.26(m,2H),2.61-2.50(m,1H),1.77(s,3H),1.42(s,3H),1.17-1.14(q,J=3.8Hz,6H)。
        Carbon spectrum: 13 C NMR (100MHz, CDCl 3 ,ZQF-RD01-2)δ(ppm):176.7,155.2,147.3,123.5,117.2,116.7,115.6,112.6,100.0,83.8,83.0,82.0,81.4,63.1,33.8,26.4,25.6,18.9。

PATENT

WO2023167938

To a solution of (3aR,4R,6R,6aR)-4-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-6-(hydroxymethyl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxole-4-carbonitrile, Intermediate 1 (2000 mg, 6.0 mmol) (Siegel et. al. J. Med. Chem.2017, 60, 1648-1661) in tetrahydrofuran (THF) was added N,N-dimethylaminopyridine (DMAP) (0.03 eq). To the reaction mixture, isobutyric anhydride (1.1 eq) was added slowly. After the completion of the staring material, the reaction mixture was concentrated and purified by flash chromatography using 20% methanol in DCM as an eluant to give ((3aR,4R,6R,6aR)-6-(4- aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-6-cyano-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxol-4- yl)methyl isobutyrate, Intermediate 1a. LCMS: MS m/z: 402.2 (M+1).

Concentrated HCl (5 eq, 1mL) was added to a solution of Intermediate 1a (1000 mg) in acetonitrile (10 mL), which was then stirred at room temperature for 2 hours. LCMS showed the product formation. The reaction was stopped after 4 hours. The reaction mixture was diluted with ethyl acetate and quenched with saturated bicarbonate. The organic layer was separated, washed with brine, dried over sodium sulphate, and concentrated. The residue was purified by flash chromatography using 30% methanol DCM as an eluant. The collected fractions were concentrated to give Intermediate 2.1H NMR (400 MHz, Methanol-d4) δ 7.88 (s, 1H), 6.96 – 6.85 (m, 2H), 4.50 – 4.27 (m, 4H), 4.16 (dd, J = 6.2, 5.3 Hz, 1H), 2.56 (p, J = 7.0 Hz, 1H), 1.14 (dd, J = 7.0, 3.8 Hz, 6H); LCMS: MS m/z: 362.1 (M+1).

str1
Flag Counter

AS ON DEC2021 3,491,869 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

//////////

Names
IUPAC name[(2R,3S,4R,5R)-5-(4-Aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxyoxolan-2-yl]methyl 2-methylpropanoate
Identifiers
CAS Number2647441-36-7 
3D model (JSmol)Interactive image
ChEMBLChEMBL4863829
ChemSpider115037299
PubChemCID162513664
UNIIQ55KCM7PXB
showInChI
showSMILES
Properties
Chemical formulaC16H19N5O5
Molar mass361.358 g·mol−1
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

References

  1. ^ “Clinical Trials Register”.
  2. ^ “Gilead touts Veklury resilience against mutated coronavirus, plots phase 3 for new COVID oral antiviral”. 19 October 2022.
  3. ^ Cho, A.; Saunders, O. L.; Butler, T.; Zhang, L.; Xu, J.; Vela, J. E.; Feng, J. Y.; Ray, A. S.; Kim, C. U. (2012). “Synthesis and antiviral activity of a series of 1′-substituted 4-aza-7,9-dideazaadenosine C-nucleosides”Bioorganic & Medicinal Chemistry Letters22 (8): 2705–2707. doi:10.1016/j.bmcl.2012.02.105PMC 7126871PMID 22446091.
  4. ^ Yan, Victoria C.; Muller, Florian L. (2020). “Advantages of the Parent Nucleoside GS-441524 over Remdesivir for Covid-19 Treatment”ACS Medicinal Chemistry Letters11 (7): 1361–1366. doi:10.1021/acsmedchemlett.0c00316PMC 7315846PMID 32665809.
  5. ^ Gordon, C. J.; Tchesnokov, E. P.; Woolner, E.; Perry, J. K.; Feng, J. Y.; Porter, D. P.; Götte, M. (2020). “Remdesivir is a direct-acting antiviral that inhibits RNA-dependent RNA polymerase from severe acute respiratory syndrome coronavirus 2 with high potency”The Journal of Biological Chemistry295 (20): 6785–6797. doi:10.1074/jbc.RA120.013679PMC 7242698PMID 32284326.
  6. ^ Cao, Liu; et al. (2022). “The adenosine analog prodrug ATV006 is orally bioavailable and has preclinical efficacy against parental SARS-CoV-2 and variants”Science Translational Medicine14 (661): eabm7621. doi:10.1126/scitranslmed.abm7621PMC 9161374PMID 35579533.
  7. ^ “Espacenet – search results”.

//////////Obeldesivir, GS-5245ATV006

 CC(C)C(OC[C@H]1O[C@@](C#N)(C2=CC=C3C(N)=NC=NN32)[C@H](O)[C@@H]1O)=O

Oditrasertib


Oditrasertib.png
Structure of Oditrasertib

Oditrasertib

Molecular FormulaC14H15F2N3O2
Molecular Weight295.2846
  • UNII-I1YQT8HC89
  • I1YQT8HC89
  • cas 2252271-93-3
  • 4-(3,3-Difluoro-2,2-dimethylpropanoyl)-3,5-dihydro-2H-pyrido(3,4-F)(1,4)oxazepine-9-carbonitrile

PYRIDO(3,4-F)-1,4-OXAZEPINE-9-CARBONITRILE, 4-(3,3-DIFLUORO-2,2-DIMETHYL-1-OXOPROPYL)-2,3,4,5-TETRAHYDRO-

Oditrasertib is a serine/threonine kinase (STK) inhibitor.

PATENT

[WO2018213632A1]

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018213632

str1
Flag Counter

AS ON DEC2021 3,491,869 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

//////////

///////////Oditrasertib,

CC(C)(C(F)F)C(=O)N1CCOC2=C(C=NC=C2C1)C#N

ORZILOBEN


Orziloben Chemical Structure

ORZILOBEN,

CAS 1555822-28-0

 2-methyl-3-(pentyloxy)benzoic acid

Molecular Weight222.28
FormulaC13H18O3

Orziloben is a medium chain fatty acid (MCFA) analogue[1].

Patent

WO2020074964

The rising global epidemic of obesity and its comorbidities, e.g., type 2 diabetes mellitus and hyperlipidemia, is placing an enormous burden both on public health (mortality and morbidity) and on the available public health resources required to treat these conditions.

Current drugs that treat hyperlipidemia (e.g., statins, omega-3 fatty acids, fibrates) have mostly neutral effects on glycemic control, whilst drugs targeting glycemic control e.g., insulin, thiazolidinediones (TZDs), have adverse effects upon bodyweight and (for TZDs) other unwanted side-effects restricting their use.

In addition to hyperlipidemia and type 2 diabetes, a marked increase in the prevalence of non-alcoholic fatty liver disease (NAFLD) has occurred. NAFLD has become the most common chronic liver condition in Western populations in relation to the obesity and type 2 diabetes epidemics. The prevalence of non-alcoholic steatohepatitis (NASH), a form of NAFLD that is associated with hepatic inflammation and ballooning of hepatocytes, is expected to increase by 63% between 2015 and 2030 in the United States (Estes, Hepatology, 2018; 67(1): 123-133), where NASH is expected to become the leading cause of liver transplantation by 2020. As liver fibrosis, but not inflammation, is associated with mortality and morbidity in NASH patients, drugs which prevent progression/induce regression of fibrosis are also a focus of biomedical research.

The development of novel compounds that simultaneously target both hyperlipidemia and glycemic control, without the adverse side-effects (e.g., weight gain) typically associated with insulin sensitising drugs is thus a desirable goal. Such compounds would be even more attractive if they could additionally prevent the progression/reverse hepatic fibrosis and reduce hepatic steatosis. The present invention addresses these needs for new treatment methods, compounds, and pharmaceutical compositions.

str1
Flag Counter

AS ON DEC2021 3,491,869 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]. Skjaeret, et al. Preparation of benzoic acid derivatives for treatment of metabolic and liver diseases. World Intellectual Property Organization, WO2020074964 A1 2020-04-16.

/////////ORZILOBEN, 1555822-28-0, OBESITY

O=C(C1=C(C)C(OCCCCC)=CC=C1)O

Opevesostat


Opevesostat.png

img

structure image

Opevesostat

  • ODM208

2231294-96-3
Chemical Formula: C21H26N2O5S

Molecular Weight: 418.508

2-[(1,3-dihydro-2H-isoindol-2-yl)methyl]-5-{[1-(methanesulfonyl)piperidin-4-yl]methoxy}-4H-pyran-4-one

structure image

Opevesostat tosylate

2832831-08-8

Molecular Formula
C21H26N2O5S.C7H8O3S
Molecular Weight
590.71

4H-Pyran-4-one, 2-[(1,3-dihydro-2H-isoindol-2-yl)methyl]-5-[[1-(methylsulfonyl)-4-piperidinyl]methoxy]-, 4-methylbenzenesulfonate (1:1)

2-((1,3-DIHYDRO-2H-ISOINDOL-2-YL)METHYL)-5-((1-(METHYLSULFONYL)-4-PIPERIDINYL)METHOXY)-4H-PYRAN-4-ONE, TOSYLATE

useful in the treatment of a steroid receptor, in particular androgen receptor (AR), dependent conditions and diseases, and to pharmaceutical compositions containing such compounds.

Prostate cancer is worldwide the most common cancer in men. Even though the 5-year survival rate of patients with localized prostate cancer is high, the prognosis for those patients, who develop castration-resistant prostate cancer (CRPC) within that 5-year follow-up period, is poor.

The androgen receptor (AR) signalling axis is critical in all stages of prostate cancer. In the CPRC stage, disease is characterized by high AR expression, AR amplification and persistent activation of the AR signalling axis by residual tissue/tumor androgens and by other steroid hormones and intermediates of steroid biosynthesis. Thus, treatment of advanced prostate cancer involves androgen deprivation therapy (ADT) such as hormonal manipulation using gonadotropin-releasing hormone (GnRH) agonists/antagonists or surgical castration, AR antagonists or CYP17A1 inhibitors (such as abiraterone acetate in combination with prednisone).

Although therapies can initially lead to disease regression, eventually majority of the patients develop a disease that is refractory to currently available therapies. Increased progesterone levels in patients treated with abiraterone acetate has been hypothesized to be one of the resistance mechanisms. Several nonclinical and clinical studies have indicated upregulation of enzymes that catalyse steroid biosynthesis at the late stage of CRPC. Very recently it has been published that 11β-OH androstenedione can be

metabolized into 11-ketotestosterone (11-K-T) and 11-ketodehydrotestosterone (11-K-DHT) which can bind and activate AR as efficiently as testosterone and dihydrotestosterone. It has been shown that these steroids are found in high levels in plasma and tissue in prostate cancer patients, suggesting their role as AR agonists in CRPC. Furthermore, it has been addressed that prostate cancer resistance to CYP17A1 inhibition may still remain steroid dependent and responsive to therapies that can further suppress de novo intratumoral steroid synthesis upstream of CYP17A1, such as by CYP11A1 inhibition therapy (Cai, C. et al, Cancer Res., 71(20), 6503-6513, 2011).

Cytochrome P450 monooxygenase 11A1 (CYP11A1), also called cholesterol side chain cleavage enzyme, is a mitochondrial monooxygenase which catalyses the conversion of cholesterol to pregnenolone, the precursor of all steroid hormones. By inhibiting CYP11A1, the key enzyme of steroid biosynthesis upstream of CYP17A1, the total block of the whole steroid biosynthesis can be achieved. CYP11A1 inhibitors may therefore have a great potential for treating steroid hormone dependent cancers, such as prostate cancer, even in advanced stages of the disease, and especially in those patients who appear to be hormone refractory. It has been recently shown that a compound having CYP11A1 inhibitory effect significantly inhibited tumor growth in vivo in a murine CRPC xenograft model (Oksala, R. et al, Annals of Oncology, (2017) 28 (suppl.

PATENT

WO2018115591

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018115591&_cid=P20-LQXJT7-60871-1

STR1

Example 4. SIMILAR

N-((4-(((6-(Isoindolin-2-ylmethyl)-4-oxo-4H-pyran-3-yl)oxy)methyl)cyclohexyl)- methyl)methanesulfonamide (Compound 173)

To a solution of 5-hydroxy-2-(isoindolin-2-ylmethyl)-4H-pyran-4-one (0.10 g, 0.41 mmol) in DMF (2 ml) were added (4-(methylsulfonamidomethyl)cyclohexyl)methyl methanesulfonate (0.14 g, 0.45 mmol) and K2CO3 (0.12 g, 0.8 mmol). The reaction mixture was heated at 80 °C for 2 h. The mixture was cooled to RT, water (10 ml) was added and the product was extracted with EtOAc. The combined extracts were washed with water, dried with Na2SO4, filtered and evaporated. The crude product was purified by column chromatography to afford the title compound (0.06 g). 1H NMR (400 MHz, Chloroform-d) δ ppm 0.92 – 1.11 (m, 4 H) 1.40 – 1.63 (m, 2 H) 1.78 – 2.00 (m, 4 H) 2.91 – 2.99 (m, 5 H) 3.65 (d, J=6.46 Hz, 2 H) 3.77 (s, 2 H) 4.03 (s, 4 H) 5.04 (br t, J=6.31 Hz, 1 H) 6.49 (s, 1 H) 7.20 (s, 4 H) 7.59 (s, 1 H).

ntermediate 58: 5-Hydroxy-2-(isoindolin-2-ylmethyl)-4H-pyran-4-one

STR2

To a stirred solution of 2-(chloromethyl)-5-hydroxy-4H-pyran-4-one (2.0 g, 12.5 mmol) in acetonitrile (50 mL) were added DIPEA (3.22 mL, 25.0 mmol) and isoindoline (1.78 g, 25.0 mmol) at RT. When the reaction was complete, the precipitated solid was filtered and washed with EtOAc. The title compound was collected as pale brown solid (1.1 g). LC-MS: m/z 244.1 (M+H)+.

///////////////////

str1
Flag Counter

AS ON DEC2021 3,491,869 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@amcrasto

//////////

/////////Opevesostat, ODM 208

 O=C1C=C(CN2CC3=C(C=CC=C3)C2)OC=C1OCC4CCN(S(=O)(C)=O)CC4

Sotuletinib HCl


img
Sotuletinib (hydrochloride).png
Structure of SOTULETINIB

Sotuletinib HCl

CAS: 2222138-31-8 (HCl)
Chemical Formula: C20H23ClN4O3S

Molecular Weight: 434.939

7W3V82OQ0P

Synonym: Sotuletinib HCl; Sotuletinib hydrochloride, Sotuletinib monohydrochloride, BLZ945; BLZ 945; BLZ-945;

IUPAC/Chemical Name: 4-((2-(((1R,2R)-2-hydroxycyclohexyl)amino)benzo[d]thiazol-6-yl)oxy)-N-methylpicolinamide hydrochloride

2-PYRIDINECARBOXAMIDE, 4-((2-(((1R,2R)-2-HYDROXYCYCLOHEXYL)AMINO)-6-BENZOTHIAZOLYL)OXY)-N-METHYL-

Sotuletinib, also known as BLZ945, is a potent and selective CSF-1R kinase inhibitor. BLZ945 showed effects of CSF1R inhibition on other tumor-infiltrating immune cells. BLZ945 attenuates the turnover rate of TAMs while increasing the number of CD8+ T cells that infiltrate cervical and breast carcinomas. BLZ945 decreases the growth of malignant cells in the mouse mammary tumor virus-driven polyomavirus middle T antigen (MMTV-PyMT) model of mammary carcinogenesis. BLZ945 prevents tumor progression in the keratin 14-expressing human papillomavirus type 16 (K14-HPV-16) transgenic model of cervical carcinogenesis.

Sotuletinib (BLZ945) is an experimental drug in development for the treatment of amyotrophic lateral sclerosis (ALS). It works as a colony-stimulating factor 1 (CSF1) receptor inhibitor.[1][2][3]

  • OriginatorCelgene Corporation; Novartis
  • ClassAmides; Amines; Antineoplastics; Benzothiazoles; Cyclohexanols; Ethers; Pyridines; Small molecules
  • Mechanism of ActionMacrophage colony stimulating factor receptor antagonists
  • Phase IIAmyotrophic lateral sclerosis
  • Phase I/IISolid tumours
  • 05 Dec 2022Novartis Pharmaceuticals terminates a phase I/II trials in Solid tumours (Combination therapy, Late-stage disease, Metastatic disease) in Taiwan, Japan, Israel (PO) in US, Israel, Italy, Japan, Singapore, Spain, Taiwan and Switzerland (EudraCT2015-005806-12) (NCT02829723)
  • 14 Feb 2022Adverse events and pharmacodynamics data from preclinical macaque model study in brain disorders presented at the 29th Conference on Retroviruses and Opportunistic Infections
  • 03 Dec 2020Chemical structure information added

An orally bioavailable inhibitor of colony stimulating factor 1 receptor (CSF-1R; CSF1R), with potential antineoplastic activity. CSF1R inhibitor BLZ945 selectively binds to CSF1R expressed on tumor-associated macrophages (TAMs), blocks the activity of CSF1R, and inhibits CSF1R-mediated signal transduction pathways. This inhibits the activity and proliferation of TAMs, and reprograms the immunosuppressive nature of existing TAMs. Altogether, this reduces TAM-mediated immune suppression in the tumor microenvironment, re-activates the immune system, and improves anti-tumor cell responses mediated by T-cells. CSF1R, also known as macrophage colony-stimulating factor receptor (M-CSFR) and CD115 (cluster of differentiation 115), is a cell-surface receptor for its ligand, colony stimulating factor 1 (CSF1); this receptor is overexpressed by TAMs in the tumor microenvironment, and plays a major role in both immune suppression and the induction of tumor cell proliferation.

PATENT

The free base and salts of the compound of formula (I) may be prepared for example, according to the procedures given in International Patent Application No. PCT/US2007/066898 filed on Apr. 18, 2007 and published as WO2007/121484 on Oct. 25, 2007. The compound of formula (I) has the chemical name: 4-(2-((1R,2R)-2-hydroxycyclohexylamino)benzothiazol-6-yloxy)-N-methylpicolinamide and is also known as BLZ945.

STR1
STR2
STR3
str4

Step 1. Preparation of 4-(2-((lR,2R)-2-aminocyclohexylamino)benzo[d]thiazol-6-yloxy)-N-methylpicolinamide
To the solution of N-methyl-4-(2-(methylsulfinyl)benzo[d]thiazol-6-yloxy)picolinamide (15 mg, 43 μmole) in 400 μL of NMP was added (lR,2R)-cyclohexane-1,2-diamine (17 mg, 150 μmole). The reaction solution was stirred at 105°c for 24 hours. The crude reaction solution was purified on prep HPLC and evaporated in vaccuo to give 4-(2-((lR,2R)-2-aminocyclohexylamino)benzo[d]thiazol-6-yloxy)-N-methylpicolinamide (12 mg, 30 μmole) as white powder. ES/MS m/z 398.1(MH+).

PATENT

 US-2020093801

https://patents.google.com/patent/US20200093801A1

PATENT

CN116139135

PATENT

US20200190057

PATENT

CN110475555

str1
Flag Counter

AS ON DEC2021 3,491,869 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@amcrasto

//////////

///////////

References

  1. ^ Pognan, François; Buono, Chiara; Couttet, Philippe; Galarneau, Jean-René; Timsit, Yoav; Wolf, Armin (29 October 2022). “Liver enzyme delayed clearance in rat treated by CSF1 receptor specific antagonist Sotuletinib”Current Research in Toxicology3: 100091. doi:10.1016/j.crtox.2022.100091ISSN 2666-027X.
  2. ^ Thongchot, Suyanee; Duangkaew, Supani; Yotchai, Wasan; Maungsomboon, Sorranart; Phimolsarnti, Rapin; Asavamongkolkul, Apichat; Thuwajit, Peti; Thuwajit, Chanitra; Chandhanayingyong, Chandhanarat (2 December 2022). “Novel CSF1R-positive tenosynovial giant cell tumor cell lines and their pexidartinib (PLX3397) and sotuletinib (BLZ945)-induced apoptosis”Human Cell36 (1): 456–467. doi:10.1007/s13577-022-00823-0.
  3. ^ Martinez-Gonzalez, Loreto; Martinez, Ana (1 February 2023). “Emerging clinical investigational drugs for the treatment of amyotrophic lateral sclerosis”. Expert Opinion on Investigational Drugs32 (2): 141–160. doi:10.1080/13543784.2023.2178416.

////////Sotuletinib HCl, BLZ945,  BLZ 945, BLZ-945,

O=C(NC)C1=NC=CC(OC2=CC=C3N=C(N[C@H]4[C@H](O)CCCC4)SC3=C2)=C1.[H]Cl

Follow New Drug Approvals on WordPress.com

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

Join 2,841 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