Home » Uncategorized (Page 2)
Category Archives: Uncategorized
Rogocekib



Rogocekib
CAS 2144751-78-8
MF C19H17FN8O2 MW 408.39
1-({5-[(1R)-1-fluoroethyl]-1,3,4-oxadiazol-2-yl}methyl)-6-(4-methoxypyrrolo[2,1-f][1,2,4]triazin-5-yl)-2-methyl1H-imidazo[4,5-b]pyridine
2-[(1R)-1-fluoroethyl]-5-[[6-(4-methoxypyrrolo[2,1-f][1,2,4]triazin-5-yl)-2-methylimidazo[4,5-b]pyridin-1-yl]methyl]-1,3,4-oxadiazole
dual specificity protein kinase CLK (CDC2-like kinase)inhibitor, antineoplastic, CTX 712, XE88VQP94E
Rogocekib is an orally effective CLK 2 inhibitor, with an IC50 of 1.4 nM, showing anti-tumor activity.
Rogocekib is an orally bioavailable inhibitor of CLK family kinases, with potential antineoplastic activity. Upon oral administration, rogocekib binds to and inhibits the activity of CLK family kinases, thereby inhibiting the phosphorylation of serine/arginine-rich (SR) domain-containing splicing factors (SFs). This modulates RNA splicing, prevents the expression of certain tumor-associated genes, and inhibits tumor cell proliferation. In many cancer cells, core spliceosome proteins, including SF3B1, U2 small nuclear ribonucleoprotein auxiliary factor 1 (U2AF1), serine/arginine-rich splicing factor 2 (SRSF2) and U2 small nuclear ribonucleoprotein auxiliary factor subunit-related protein 2 (ZRSR2), are mutated and aberrantly activated leading to a dysregulation of mRNA splicing. CLK family kinases, an evolutionarily conserved group of kinases, phosphorylates various SR proteins including SR domain-containing SFs.
SYN
https://pubs.acs.org/doi/10.1021/acsmedchemlett.5c00412



(R)-2-fluoropropanoic acid (21)
(R)-Ethyl 2-fluoropropanoate (20) (95 g, 791 mmol) was suspended in 10% sulfuric acid (950 mL), and heated and
refluxed for 3 h. After cooled, sodium chloride was added to saturate the aqueous layer, and the aqueous layer
was extracted with TBME (900 mL x4). The obtained organic layer was dried over MgSO4, and concentrated under
reduced pressure to give the title compound (124 g, 791 mmol calcd as quant., containing TBME).
1H NMR (300 MHz, DMSO-d6) δ 1.35-1.56 (3H, m), 4.91-5.21 (1H, m), 13.19 (1H, brs).
(S)-2-amino-3-phenylpropane-1-ol (R)-2-fluoropropanoate (22)
To a solution of (S)-2-amino-3-phenylpropan-1-ol (119 g, 787 mmol) in EtOH (360 mL) and MeCN (1090 mL) was
added dropwise a solution of 21 (791 mmol, theoretically calcd as quant.) in MeCN (1090 mL) at 65° C to 70° C.
The mixture was stirred at 60° C for 1 h, and further stirred at room temperature for 1 h. Precipitated crystals were
collected by filtration, and washed with MeCN (500 mL) to obtain white crystals (170 g, 699 mmol, 89%).
The obtained crystals(140 g, 575 mmol) were dissolved in EtOH (700 mL) at 60° C, and to the solution was added
MeCN (4200 mL) at 58° C to 65° C. The mixture was stirred at 60° C for 1 h. The mixture was cooled to room
temperature, and then stirred overnight at room temperature. The obtained solid was collected by filtration, and
washed with MeCN to obtain give the title compound (109 g, 448 mmol, 78%) as a white crystal.
(R)-2-((6-bromo-2-methyl-1H-imidazo[4,5-b]pyridin-1-yl)methyl)-5-(1-fluoroethyl)-1,3,4-oxadiazole ((R)-19b)
22 (109 g, 448 mmol) was dissolved in 1M HCl aq. (1500 mL) and brine (1500 mL) and extracted with TBME (1000
mL x4). The organic layer was dried over MgSO4 and concentrated in vacuo to give free salt of 22 (i.e., 21) as a
colorless oil. 50 wt% T3P in EtOAc (419 mL, 704 mmol) was added to a suspension of the above material, 17a (100
g, 351.97 mmol), and DIPEA (246 mL, 1408 mmol) in BuOAc (3000 mL) at room temperature. After being stirred at
50 °C for 30 min, 50 wt% T3P in EtOAc (210 mL, 351.97 mmol) was added to the mixture and then the mixture was
heated and refluxed for 3 h. After cooling, to the mixture was added sat NaHCO3 aq. (3000 mL), then the insoluble
material was removed by filtration. The filtrate was extracted with EtOAc (1500 mL x2). The organic layer was
separated, washed with water and brine, then passed through NH silica gel eluted with EtOAc. The residue was
concentrated in vacuo and the resulting precipitate was washed with IPE (3000 mL) to give the title compound
(57.8 g, 170 mmol, 48.3%) as an off-white solid.
1H NMR (300 MHz, DMSO-d6) δ 1.62-1.79 (3H, m), 2.62 (3H, s), 5.83-6.14 (3H, m), 8.38 (1H, d, J = 1.9 Hz), 8.45 (1H,
d, J = 1.9 Hz). MS m/z 340.0, 341.9 [M+H]+
.
1-((5-((1R)-1-fluoroethyl)-1,3,4-oxadiazol-2-yl)methyl)-6-(4-methoxypyrrolo[2,1-f][1,2,4]triazin-5-yl)-2-methyl1H-imidazo[4,5-b]pyridine ((R)-19, CTX-712)
A mixture of (4-methoxypyrrolo[2,1-f][1,2,4]triazin-5-yl)boronic acid (79 g, 409.39 mmol), (R)-19b (100 g, 294
mmol), Pd(Amphos)Cl2 (2.00 g, 2.97 mmol), 2 M Cs2CO3 aq. (295 mL, 590 mmol) and DME (2000 mL) was stirred at
80 °C for 1 h. After cooled to 50 °C, the mixture was diluted with THF (1000 mL). The mixture was poured into
NaHCO3 aq. (1600 mL) and extracted with EtOAc (1000 mL x3). The organic layer was separated, washed with 5%
ammonia aq. (1600 mLx2) and brine (1600 mL), dried over MgSO4 and concentrated in vacuo to give a yellow solid.
To the solution of obtained solid in THF (8000 mL) and water (200 mL) was added NH silica gel (2400 g) and stirred
for 3.5 h at room temperature. The insoluble material was removed by filtration and washed with THF (15 L). The
filtrate was concentrated in vacuo to give a yellow solid. The solid was washed with TBME to give the title
compound (98 g, 240 mmol, 82 %) as a pale yellow solid. A mixture of the above material (115 g, 270 mmol) and
activated carbon (Ecosorb, 33 g) in EtOH/water = 9/1 (2200 mL) and water (1100 mL) was stirred at 55 °C for 1 h.
The insoluble material was removed by filtration, and washed EtOH (550 mL). The resultant solution was diluted
with water (1600 mL) at 55 °C and stirred at room temperature overnight. After cooled to 5 °C, the mixture was
stirred for 3 h. The solid was collected by filtration and washed with EtOH/water = 1/1 (1000 mL) to give a
colorless crystal (88 g, 207 mmol, 77% as a water adduct).
1H NMR (300 MHz, DMSO-d6) δ 1.58-1.82 (3H, m), 2.67 (3H, s), 3.96 (3H, s), 5.83-6.18 (3H, m), 7.06 (1H, d, J = 2.7
Hz), 8.06 (1H, d, J = 2.7 Hz), 8.23 (2H, t, J = 1.0 Hz), 8.59 (1H, d, J = 2.0 Hz). MS m/z 409.1 [M+H]+
.

PAT
Patent document 1:
WO 2010/016526
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2010016526&_cid=P10-MIIA44-38372-1
WO 2011/096535
SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2023190967&_cid=P10-MII9ZT-35263-1
SYN
SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=JP275206879&_cid=P10-MII9SJ-29591-1
PAT
- Condensed Heterocyclic CompoundsPublication Number: KR-102431405-B1Priority Date: 2016-04-28Grant Date: 2022-08-10
- COMPOUND, MEDICINE, AND, USE OF THE COMPOUND OR SALT THEREOFPublication Number: BR-112018072039-B1Priority Date: 2016-04-28
- Fused Heterocyclic CompoundsPublication Number: CN-109415384-BPriority Date: 2016-04-28Grant Date: 2022-01-11
- Condensed heterocyclic compoundPublication Number: EP-3450436-B1Priority Date: 2016-04-28Grant Date: 2022-07-27
- Fused heterocyclic compoundPublication Number: US-11390634-B2Priority Date: 2016-04-28Grant Date: 2022-07-19
- condensed heterocyclic compoundPublication Number: ES-2927529-T3Priority Date: 2016-04-28Grant Date: 2022-11-08
- CONDENSED HETEROCYCLIC COMPOUNDPublication Number: HR-P20221277-T1Priority Date: 2016-04-28
- Fused heterocyclic compoundPublication Number: US-10577382-B2Priority Date: 2016-04-28Grant Date: 2020-03-03
- Fused heterocyclic compoundPublication Number: US-2019106437-A1Priority Date: 2016-04-28
- Fused heterocyclic compoundPublication Number: US-2020140462-A1Priority Date: 2016-04-28
- Fused heterocyclic compoundPublication Number: US-10981934-B2Priority Date: 2016-04-28Grant Date: 2021-04-20
- Fused heterocyclic compoundPublication Number: US-2021115067-A1Priority Date: 2016-04-28
- Medicament for treatment and/or prevention of cancerPublication Number: WO-2024048541-A1Priority Date: 2022-08-30
- Biomarker for treatment of solid cancer by imidazo[4,5-b]pyridine derivativePublication Number: WO-2023190967-A1Priority Date: 2022-03-31
- Fused heterocyclic compoundPublication Number: CA-3021185-A1Priority Date: 2016-04-28
- Condensed heterocyclic compoundPublication Number: EP-3450436-A1Priority Date: 2016-04-28
- Fused heterocyclic compoundsPublication Number: JP-WO2017188374-A1Priority Date: 2016-04-28



AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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]. Akinori Yoda, et al. CTX-712, a Novel Clk Inhibitor Targeting Myeloid Neoplasms with SRSF2 Mutation. Blood. (2021) 205–206[2]. Zhen Qin, et al. Development of Cdc2-like Kinase 2 Inhibitors: Achievements and Future Directions. J Med Chem. 2021 Sep 23;64(18):13191-13211. [Content Brief]
///////rogocekib, CTX 712, XE88VQP94E
Riselcaftor





Riselcaftor
CAS 2799652-36-9
MF C29H28N2O5S MW 516.61
(2R,4R)-2-(2-methoxy-5-methylphenyl)-N-(2-methylquinoline-5-sulfonyl)-4-phenyloxolane-2-
carboxamide
(2R,4R)-2-(2-methoxy-5-methylphenyl)-N-(2-methylquinolin-5-yl)sulfonyl-4-phenyloxolane-2-carboxamide
cystic fibrosis transmembrane regulator (CFTR)protein modulator, 726GWJ6KQQ
Riselcaftor (Example 33) is a CFTR modulator, with an EC50 of 20.1 nM in human bronchial epithelial cells. Riselcaftor can be used for research of cystic fibrosis.
SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=US367940046&_cid=P11-MIH63N-23616-1
Example 33
(2R,4R)-2-(2-methoxy-5-methylphenyl)-N-(2-methylquinoline-5-sulfonyl)-4-phenyloxolane-2-carboxamide
PAT
- Modulators of the Cystic Fibrosis Transmembrane Conductance Regulator Protein and Methods of UsePublication Number: US-2022211692-A1Priority Date: 2021-01-06
- Modulators of the cystic fibrosis transmembrane conductance regulator protein and methods of usePublication Number: WO-2022150174-A1Priority Date: 2021-01-06



AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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

……
//////Riselcaftor, 726GWJ6KQQ
Pudafensine


Pudafensine
CAS 1320346-14-2
MFC17H19NO4 MW 301.34 g/mol
7-{[(1R,3s,5S)-8-azabicyclo[3.2.1]octan-3-yl]oxy}-3-methoxy2H-1-benzopyran-2-one
monoamine reuptake inhibitor, erectile dysfunction, neuropathic pain, NS18313, NS 18313, L9NG7US8GE, IP2015, IP 2015
Pudafensine is a monoamine reuptake inhibitor being developed as a potential treatment for erectile dysfunction (ED) and neuropathic pain. As a drug candidate, it works by preferentially inhibiting the reuptake of dopamine and serotonin. It is designed to be a first-line treatment for patients with organic ED who are not adequately served by existing therapies like PDE5 inhibitors.
How it works
- Pudafensine is a monoamine reuptake inhibitor that increases the levels of dopamine and serotonin in the brain by preventing their reabsorption into neurons.
- It has been shown in animal models and human trials to improve erectile function and reduce pain, including neuropathic pain.
Potential uses
- Erectile Dysfunction (ED): Pudafensine is being investigated for its potential to help men with organic ED who do not respond well to or cannot tolerate current treatments. Phase IIb clinical trial results are expected in late 2023.
- Neuropathic Pain: A clinical trial on pain involving pudafensine indicated it reduced allodynia and was well-tolerated with a favorable safety profile compared to pregabalin.
Development status
- Initiator Pharma is developing pudafensine as an oral tablet.
- Phase IIb studies for erectile dysfunction and Phase II studies for neuropathic pain have been completed, with positive results.
- The company is exploring its use in treating patients who are inadequately treated with existing medications.
Erectile dysfunction (ED)
Pudafensine, Initiator’s most advanced drug program has successfully demonstrated efficacy in a Clinicial Phase 2a Proof-of-Concept study and in a Phase 2b study to treat patients who suffer from organic erectile dysfunction (ED) that do not respond or cannot tolerate the currently marketed drugs in the PDE5i class (e.g. Viagra®, Cialis®, Levitra®).
Pudafensine strengthens the natural erection response by having a dual-action, both a central effect initiating erection and a peripheral effect potentiating erection through smooth muscle relaxation. Pudafensine is aimed for treatment of organic erectile dysfunction in patients who have erectile dysfunction (ED) due to abnormalities of the penile arteries and/or veins. Most common risk factors for organic ED are diabetes, overweight, lack of exercise, high cholesterol, high blood pressure, and cigarette smoking. Since Initiator Pharma was founded and pudafensine acquired, all preclinical development of the drug candidate to enable an application for clinical trials (CTA) has been carried out by the company’s auspices. Pudafensine is developed as a tablet that is taken orally on-demand. It is the company’s goal to be able to create a new “First-Line” treatment (recommended treatment) for the large group of men who have organic erectile dysfunction, who are sub-optimally treated with PDE5i products or for whom PDE5i treatment is contraindicated.
In Q4 2023 positive results from the Phase IIb clinical trial with pudafensine (IP2015) was announced. The Phase 2b trial is a randomized, double-blind, placebo-controlled, parallel-dosing group trial studying the efficacy and safety of high and low doses of pudafensine (IP2015) and placebo in otherwise healthy patients suffering from moderate to severe ED. The study comprises 130 patients divided into 3 parallel arms receiving a higher and a lower dose of pudafensine and placebo, respectively, with treatment duration of 4 weeks with frequent assessments of erectile dysfunction, safety and pharmacokinetics. The study has been conducted at the MAC clinical sites in the UK.
The study demonstrated statistically significant efficacy on the primary endpoint (related to improvements in intercourse settings) compared to placebo [p=0.034] and baseline [p=0.046]. Furthermore, the results were consistent throughout the study. Several other clinical endpoints related to improved intercourse activities (obtained from the International Index of Erectile Function Questionnaire, IIEF-15) demonstrated significant effects compared to the baseline. The frequency and type of adverse effects were mild to moderate and comparable to those observed in the placebo group. There was no reporting of critical safety observations.
Neuropathic pain
Pudafensine have shown effects in a human model of pain ie. in a clinical Phase I study in healthy subjects dosed with the drug pudafensine and challenged with a pain-inducing ingredient (capsaicin).
The Phase I study was a randomized, double blind, placebo controlled study in 24 healthy male subjects, investigating the effects on pain measures (hyperalgesia, allodynia, and subjects pain rating) of single doses of pudafensine, pregabalin as an active control, and placebo. The pain was induced by intradermal capsaicin. Pudafensine demonstrated a statistically significant effect on allodynia (p=0.049) and showed a dose-dependent effect on the measured pain parameters. Pregabalin (p=0.083) and IP2015 (p=0.051) tended to reduce hyperalgesia, although the effects on hyperalgesia were not statistically significant compared to placebo-treated subjects.
Syn
US20130040985
https://patentscope.wipo.int/search/en/detail.jsf?docId=US76705962&_cid=P22-MIFE0H-55553-1

endo-Benzoic acid 8-methyl-8-aza-bicyclo[3.2.1]oct-3-yl ester

Benzoylchloride (84.3 g, 600 mmol) was added during 30 min at <30° C. to a mixture of tropine (70.6 g, 500 mmol), potassium tert-butoxide (67.3 g, 600 mmol) and THF (500 ml). The mixture was stirred at room temperature for 2 h. Water (1 L) was added followed by extraction with diethylether (2×500 ml). The organic phase was washed twice with water (2×200 ml) followed by a solution of saturated aqueous sodium chloride (200 ml). The ether phase was dried and hydrochloric acid in ethanol (170 ml, 3 M) was added. The precipitated hydrochloride was filtered and washed with diethylether. The free base was obtained by adding an excess of aqueous ammonia followed by extraction with a mixture of ethylacetate and diethylether. Yield 66.8 g (54%).
endo-Benzoic acid 8-aza-bicyclo[3.2.1]oct-3-yl ester

2,2,2-Trichloroethylchloroformate (75.0 ml, 544 mmol) was added dropwise to a mixture of endo-benzoic acid 8-methyl-8-aza-bicyclo[3.2.1]oct-3-yl ester (66.8 g, 272 mmol) and dry toluene (500 ml). The mixture was allowed to stir for 1 h at room temperature, followed by 15 h at 100° C. Water (250 ml) was added followed by stirring 1 h. The phases were separated and the organic phase was washed twice with water (2×200 ml). The mixture of the intermediate 3-benzoyloxy-8-aza-bicyclo[3.2.1]octane-8-carboxylic acid trichloromethyl ester, was dried and evaporated. Acetic acid (350 ml) was added followed by addition of zinc (53.4 g, 817 mmol) over 3 h time period. Water (100 ml) was added, cooled by adding ice and made alkaline by adding concentrated aqueous ammonia (ca: 400 ml) and the mixture was extracted with dichloromethane (2×300 ml). Yield 44.5 g (61%).
endo-3-Benzoyloxy-8-aza-bicyclo[3.2.1]octane-8-carboxylic acid tert-butyl ester

Di-tert-butyl-dicarbonate (39.9 g, 183 mmol) solved in THF (100 ml) was added to a stirred mixture of endo-benzoic acid 8-aza-bicyclo[3.2.1]oct-3-yl ester (44.5 g, 166.4 mmol), triethylamine (67.4 g, 666 mmol) and THF (250 ml) during 0.5 h at room temperature, followed by stirring for 1 h. Water (1 L) was added and the mixture was extracted with diethylether (2×300 ml). The collected ether phase was washed twice with water (2×200 ml), dried and evaporated. Yield 60.1 g (100%).
endo-3-Hydroxy-8-aza-bicyclo[3.2.1]octane-8-carboxylic acid tert-butyl ester

A mixture of endo-3-benzoyloxy-8-aza-bicyclo[3.2.1]octane-8-carboxylic acid tert-butyl ester (55.0 g, 166 mmol), potassium hydroxide (11.2 g 199 mmol) and ethanol (99%, 400 ml) was stirred for 3 days at room temperature. Potassium benzoate was separated by filtration and the filtrate was evaporated. Diethylether (200 ml) was added and remaining potassium benzoate was separated by filtration and the filtrate was evaporated. The product was triturated with petroleum. Yield 30.0 g (80%). Mp 139.5-140.8° C.
xample 1
Exo-tert-butyl-3-(3-methoxy-2-oxo-chromen-7-yl)oxy-8-azabicyclo[3.2.1]octane-8-carboxylate (Intermediate)

Triphenylphosphine (1.15 g, 4.37 mmol) was solved in toluene (20 ml) and cooled to <20° C. Diethylazodicarboxylate (40% in toluene) (2.0 ml, 4.37 mmol) was added to the mixture below 20° C., followed by stirring for 10 minutes. endo-3-Hydroxy-8-aza-bicyclo[3.2.1]octane-8-carboxylic acid tert-butyl ester (0.828 g, 3.64 mmol) was added and after 10 minutes 7-hydroxy-3-methoxy-chromen-2-one (0.70 g, 3.64 mmol) (prepared according to J. Med. Chem. 1999, 42, p2662-2672) was added to the mixture. The temperature raised to 25° C. due to an exothermic reaction. The mixture precipitates. The mixture was allowed to stir for 15 h at room temperature. Water (20 ml) and sodium hydroxide (0.5 ml, 4 M) was added followed by stirring. The mixture was cooled on an ice-bath, filtered and washed with water and diethylether. Yield 0.92 g (63%).
Exo-7-[(-8-azabicyclo[3.2.1]octan-3-yl)oxy]-3-methoxy-chromen-2-one hydrochloride (Compound 1.1)

Exo-tert-butyl-3-(3-methoxy-2-oxo-chromen-7-yl)oxy-8-azabicyclo[3.2.1]octane-8-carboxylate (0.92 g, 2.29 mmol) and hydrogen chloride (15 ml, 1 M) in acetic acid was mixed as a solution and stirred at room-temperature and precipitated after a few minutes. The product was filtered and washed with diethylether. Yield 0.48 g (62%). LC-ESI-HRMS of [M+H]+ shows 302.13856 Da. Calc. 302.138689 Da, dev. −0.4 ppm.
Syn
WO2011092061
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2011092061&_cid=P22-MIFE80-61015-1






SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2024008808&_cid=P22-MIFDSB-50229-1

SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2024089247&_cid=P22-MIFDSB-50229-1
SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2024146892&_cid=P22-MIFDSB-50229-1
PAT
- Compound for treatment of erectile dysfunctionPublication Number: WO-2024146892-A1Priority Date: 2023-01-03
- Compound for treatment of painPublication Number: WO-2024089247-A1Priority Date: 2022-10-28
- Compound for treatment of female sexual dysfunctionPublication Number: WO-2024008808-A1Priority Date: 2022-07-08



AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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

……
//////////Pudafensine, monoamine reuptake inhibitor, erectile dysfunction, neuropathic pain, NS18313, NS 18313, L9NG7US8GE, IP2015, IP 2015
Privosegtor



Privosegtor
CAS 1361200-34-1
MF C25H38FN5O4, MW 491.6 g/mol
GLYCINAMIDE, N-(2-(2-FLUOROPHENYL)ETHYL)GLYCYL-N-(2-METHYLPROPYL)GLYCYL-N2-(3-(2-OXO-1-PYRROLIDINYL)PROPYL)-
N-(2-(2-FLUOROPHENYL)ETHYL)GLYCYL-N-(2-METHYLPROPYL)GLYCYL-N2-(3-(2-OXO-1-PYRROLIDINYL)PROPYL)GLYCINAMIDE
N-(2-(2-FLUOROPHENYL)ETHYL)GLYCYL-N-(2-METHYLPROPYL)GLYCYL-N2-(3-(2-OXOPYRROLIDIN-1-YL)PROPYL)GLYCINAMIDE
N-[2-(2-fluorophenyl)ethyl]glycyl-N-(2-methylpropyl)glycyl-N2[3-(2-oxopyrrolidin-1-yl)propyl]glycinamide
serum/ glucocorticoid-regulated kinase 2 (Sgk2) activator, Phase 2, Optic neuritis, orphan drug, BN-201, BN 201, G-79, G 79, KCN37L7EIH
- OriginatorBionure
- DeveloperBionure; Oculis Pharma
- ClassAnti-inflammatories; Antiglaucomas; Eye disorder therapies; Neuroprotectants; Peptides; Small molecules
- Mechanism of ActionBrain derived neurotrophic factor agonists; Insulin-like growth factor I stimulants; Neuron modulators; Serum-glucocorticoid regulated kinase stimulants
- Orphan Drug StatusYes – Optic neuritis
- Phase IIOptic neuritis
- PreclinicalMultiple sclerosis; Neurotrophic keratopathy
- No development reportedGlaucoma; Neuromyelitis optica
- 06 Oct 2025Oculis Holding plans the PIONEER-2 trial in Optic neuritis in first half of 2026
- 06 Oct 2025Oculis Holding plans the PIONEER-3 trial in Optic nerve disorders in mid-2026
- 06 Oct 2025Oculis Holding completes End-of-phase II meeting with US FDA and receives positive feedback for registrational PIONEER program in Optic neuritis and Optic nerve disorders
OCS-05 in Patients With Optic Neuritis
CTID: NCT04762017
Phase: Phase 2
Status: Completed
Date: 2025-09-22
N-[2-[(2-amino-2-oxoethyl)-[3-(2-oxopyrrolidin-1-yl)propyl]amino]-2-oxoethyl]-2-[2-(2-fluorophenyl)ethylamino]-N-(2-methylpropyl)acetamide (BN201) is a small peptide molecule, a first-in-class neuroprotective compound. BN201 promotes the survival of cultured neural cells when subjected to oxidative stress or when deprived of trophic factors. BN201 promotes neuronal differentiation, the differentiation of precursor cells to mature oligodendrocytes in vitro, and the myelination of new axons. BN201 modulates several kinases participating in the insulin growth factor 1 pathway including serum-glucocorticoid kinase and midkine, inducing the phosphorylation of NDRG1 and the translocation of the transcription factor Foxo3 to the cytoplasm. In vivo, BN201 prevents axonal and neuronal loss, and it promotes remyelination in models of multiple sclerosis, chemically induced demyelination, and glaucoma. Bionure, a spin-off from Hospital Clínic de Barcelona that is based in California, is developing BN201 for multiple sclerosis, acute optic neuritis (AON) and glaucoma. BN201 was granted with orphan designation status for optic neuritis by the FDA. Optic neuritis is often an early sign of multiple sclerosis. The efficacy, safety, and capacity of the drug to cross the blood-brain barrier have been demonstrated in animal models, but the drug has not yet entered clinical testing.
PAT
Agonists of neurotrophin receptors and their use as medicaments
Publication Number: WO-2012028959-A1
Priority Date: 2010-08-31
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2012028959&_cid=P10-MIDYQ0-58943-1
PAT
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021084013&_cid=P10-MIDYSN-60542-1
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below throughout all the description, the compound of formula (I) is selected from the group consisting of G79 ([N-(2-(2′-fluorophenyl)ethyl)- glycyl]-[N-(2-methylpropyl)-glycyl]-N-[3-(2′-oxopyrrolidinyl)-propyl]glycinamide, BN201 , Chemical Formula: C25H38FN5O4; MW 491.5987), G-80 ([N-(2-(2′-fluorophenyl)ethyl)- glycyl]-[N-(2-methyl-propyl)glycyl]-N-[2-(4′-sulfamoyl-phenyl)ethyl]glycinamide, BN 119, Chemical Formula: C26H36FN5O5S; MW 549.658) and G81 ([N-(2-(1 -pyrrolidinyl)ethyl)- glycyl]-[N-(2-methyl-propyl)glycyl]-N-[2-(4′-sulfamoyl-phenyl)ethyl]glycinamide, BN 120, Chemical Formula: C24H4oN6OS; MW 524.6766):

G79 (BN201) G80 (BN119) G81 (BN120)
Compounds of formula (I) can be prepared as disclosed in WO2012028959.
PAT
- Agonists of Neurotrophin Receptors and Their Use as MedicamentsPublication Number: US-2012052094-A1Priority Date: 2010-08-31
- Agonists of Neurotrophin Receptors and Their Use as MedicamentsPublication Number: US-2015005239-A1Priority Date: 2010-08-31
- Agonists of neurotrophin receptors and their use as medicamentsPublication Number: US-2017121367-A1Priority Date: 2010-08-31
- Agonists of neurotrophin receptors and their use as medicamentsPublication Number: US-8791076-B2Priority Date: 2010-08-31Grant Date: 2014-07-29
- Agonists of neurotrophin receptors and their use as medicamentsPublication Number: US-9453047-B2Priority Date: 2010-08-31Grant Date: 2016-09-27
- Combination Therapy Methods, Compositions and KitsPublication Number: KR-20220109378-APriority Date: 2019-07-03
- Combination therapy methods, compositions and kitsPublication Number: US-2022378866-A1Priority Date: 2019-07-03
- Agonists of neurotrophin receptors and their use as medicamentsPublication Number: EP-2611775-A1Priority Date: 2010-08-31
- Agonists of neurotrophin receptors and their use as medicamentsPublication Number: EP-2611775-B1Priority Date: 2010-08-31Grant Date: 2016-03-16
- Agonists of neurotrophin receptors and their use as medicamentsPublication Number: US-10106577-B2Priority Date: 2010-08-31Grant Date: 2018-10-23
- Combination therapy methods, compositions and kitsPublication Number: WO-2021001464-A1Priority Date: 2019-07-03
- Combination therapy methods, compositions and kitsPublication Number: AU-2020298782-A1Priority Date: 2019-07-03
- Combination therapy methods, compositions and kitsPublication Number: CN-114206329-APriority Date: 2019-07-03
- Combination therapy methods, compositions and kitsPublication Number: EP-3993784-A1Priority Date: 2019-07-03
- Combination therapy methods, compositions and kitsPublication Number: JP-2022539999-APriority Date: 2019-07-03
- Boron-nitrogen compound, organic electroluminescence composition, and organic electroluminescence device containing samePublication Number: WO-2022121951-A1Priority Date: 2020-12-10
- New treatment regimen for the treatment of neurological diseases or conditionsPublication Number: WO-2021084013-A1Priority Date: 2019-10-30
- Novel Therapeutic Approaches for the Treatment of Neurological Diseases or ConditionsPublication Number: CN-115052595-APriority Date: 2019-10-30
- New treatment regimen for the treatment of neurological diseases or conditionsPublication Number: EP-4051263-A1Priority Date: 2019-10-30
- New treatment regiment for the treatment of neurological diseases or conditionsPublication Number: US-2022387385-A1Priority Date: 2019-10-30
- A plant zinc-increasing compound inoculant and its preparation method and applicationPublication Number: CN-117286034-APriority Date: 2023-09-11
- A plant zinc-enhancing composite bacterial agent and its preparation method and applicationPublication Number: CN-117286034-BPriority Date: 2023-09-11Grant Date: 2024-11-15
- Compound, pharmaceutical composition comprising the same, and process for synthesizing the samePublication Number: TW-202432095-APriority Date: 2022-12-22
- Synthesis of small molecule agonists of neuroptrophinPublication Number: WO-2024133860-A1Priority Date: 2022-12-22
- Boron-nitrogen compound, organic electroluminescent composition and organic electroluminescent device containing samePublication Number: WO-2022121920-A1Priority Date: 2020-12-10




AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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

……
- Development and validation of PAMPA-BBB QSAR model to predict brain penetration potential of novel drug candidatesPublication Name: Frontiers in PharmacologyPublication Date: 2023-12-01PMCID: PMC10722238PMID: 38108064DOI: 10.3389/fphar.2023.1291246
- A Phase 1 randomized study on the safety and pharmacokinetics of OCS-05, a neuroprotective disease modifying treatment for Acute Optic Neuritis and Multiple SclerosisPublication Name: Scientific ReportsPublication Date: 2023-03-29PMCID: PMC10060579PMID: 36991169DOI: 10.1038/s41598-023-32278-0
- Retrospective assessment of rat liver microsomal stability at NCATS: data and QSAR modelsPublication Name: Scientific ReportsPublication Date: 2020-11-26PMCID: PMC7693334PMID: 33244000DOI: 10.1038/s41598-020-77327-0
- A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoproteinPublication Name: Molecular PharmacologyPublication Date: 2019-11PMCID: PMC6790066PMID: 31515284DOI: 10.1124/mol.119.115964
- Predictive models of aqueous solubility of organic compounds built on A large dataset of high integrityPublication Name: Bioorganic & Medicinal ChemistryPublication Date: 2019-07-15PMCID: PMC8274818PMID: 31176566DOI: 10.1016/j.bmc.2019.05.037
/////////Privosegtor, Phase 2, Optic neuritis, orphan drug, BN-201, BN 201, G-79, G 79, KCN37L7EIH
Pregabalin naproxencarbil



Pregabalin naproxencarbil
CAS 1221072-91-8
MF C25H33NO7 MW459.5 g/mol
(3S)-3-[({[(1R)-1-{[(2S)-2-(6-methoxynaphthalen-2-yl)propanoyl]oxy}ethoxy]carbonyl}amino)methyl]-5-
methylhexanoic acid
(3S)-3-[[[(1R)-1-[(2S)-2-(6-methoxynaphthalen-2-yl)propanoyl]oxyethoxy]carbonylamino]methyl]-5-methylhexanoic acid
gabamimetic, analgesic, ZVG8DDT3FJ
- OriginatorXgene Pharmaceutical
- ClassAminobutyric acids; Analgesics; Antiepileptic drugs; Antipyretics; Antirheumatics; Anxiolytics; Drug conjugates; Gabapentinoids; Naphthaleneacetic acids; Neuroprotectants; Nonsteroidal anti-inflammatories; Small molecules
- Mechanism of ActionCACNA2D1 protein modulators; Cyclooxygenase inhibitors
- Phase II/IIIPostoperative pain
- Phase IIAcute pain; Cancer pain; Pain
- Phase I/IIBack pain; Neuropathic pain
- No development reportedDiabetic neuropathies
- 15 Jul 2025XG005 licensed to NeuroGen in China, Hong Kong, and Macau
- 31 Dec 2024Efficacy and adverse events data from phase-II/III trial in Postoperative pain released by Xgene Pharmaceutical
- 25 Oct 2024Xgene Pharmaceutical completes the phase II/III trial in Postoperative pain in USA (PO)
PAT
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019219000&_cid=P22-MID0L3-49648-1

Example 1: Exemplary synthesis of the compound of Formula I and crystallization thereof

Step A: Synthesis of 1-chloroethyl 2-fluorophenyl carbonate (3)
[0306]
A suitable reaction vessel was charged with water and sodium bicarbonate followed by the starting material 2-fluoro-phenol (1) . The mixture was cooled to a temperature of about 0~5℃ and 1-chloroethyl chloroformate (2) was added slowly while maintaining the temperature at 0~5℃. The temperature was raised to about 15 ± 5℃. When the reaction was judged complete by the disappearance of 2-fluoro-phenol (criteria: ≤ 2.0%, by HPLC) the reaction was worked up. n-Heptane was added and the organic phase was separated, washed with water and brine. The solution was concentrated, then toluene was added and the solution was concentrated again. The toluene addition and the concentration cycle were repeated once more.
[0307]
Step B: Synthesis of (S) – ( (R, S) -1- ( (2-fluorophenoxy) carbonyloxy) ethyl 2- (6-methoxynaphthalen-2-yl) propanoate (4)
[0308]
To a solution of naproxen in toluene in a suitable reaction vessel, 1-chloroethyl 2-fluorophenyl carbonate (3) and cuprous oxide was added. The temperature of the mixture was raised to about 115 ±5℃. When the reaction was judged complete by the disappearance of 1-chloroethyl 2-fluorophenyl carbonate (criteria: ≤ 2.5%, by HPLC) the reaction was worked up. Methyl tert-butyl ether was added at about 50 ± 5℃. The resulting mixture was filtered and the filtrate was collected at about 25 ± 5℃. Purified water was added into the filtrate and then the mixture was cooled to about 0 ± 5℃. The mixture was alkalified with ammonium hydroxide to a pH of about 9~11 and the organic phase was separated and washed with ammonium hydroxide and brine. The solution was concentrated, then acetonitrile was added and the solution was concentrated again. The acetonitrile addition and the concentration cycle were repeated some more times until the residual toluene was not more than 10% (by GC method) .
[0309]
Step C: Synthesis of (S) -3- ( ( ( (R) -1- ( (S) -2- (6-methoxynaphthalen-2-yl) propanoyloxy) ethoxy) carbonyl-amino) methyl) -5-methylhexanoic acid (the compound of Formula I)
[0310]
To a solution of the mixture of (S) – ( (R, S) -1- ( (2-fluorophenoxy) carbonyloxy) ethyl 2- (6-methoxynaphthalen-2-yl) propanoate (4) in acetonitrile and methyl tert-butyl ether in a suitable reaction vessel, purified water and pregabalin was charged. Triethylamine was added slowly while maintaining the temperature at about 15 ± 5℃. The temperature was raised to about 25 ± 3℃ for reaction. When the reaction was judged complete by the disappearance of (S) – ( (R, S) -1- ( (2-fluorophenoxy) carbonyloxy) ethyl 2- (6-methoxynaphthalen-2-yl) propanoate (criteria: ≤ 0.5%, by HPLC) the reaction was worked up. The resulting mixture was acidified with KHSO 4to pH 3~5 and extracted with methyl tert-butyl ether. The combined organic layers were washed with purified water and brine. The organic phase was then concentrated. Isopropanol was added and the solution was concentrated again. The isopropanol addition and the concentration cycle was repeated once or more times until the total residual acetonitrile and methyl tert-butyl ether was not more than 5% (by GC method) . n-Heptane was added into the mixture at about 40~45 ℃ and stirred and then the temperature was gradually lowered at set appropriate intervals to crystallize (S) -3- ( ( ( (R) -1- ( (S) -2- (6-methoxynaphthalen-2-yl) propanoyloxy) ethoxy) carbonyl-amino) methyl) -5-methylhexanoic acid (5) from the system. When the precipitation was complete, the heterogeneous mixture was centrifuged and the solid was collected.
[0311]
The crude product of (S) -3- ( ( ( (R) -1- ( (S) -2- (6-methoxynaphthalen-2-yl) propanoyloxy) ethoxy) carbonyl-amino) methyl) -5-methylhexanoic acid (5) was added to a solution of isopropanol and water in a suitable vessel. The mixture was stirred while raising the temperature to 45 ± 3 ℃ until all the solid was dissolved, then the temperature was lowered gradually at set appropriate intervals to recrystallize (S) -3- ( ( ( (R) -1- ( (S) -2- (6-methoxynaphthalen-2-yl) propanoyloxy) ethoxy) carbonyl-amino) methyl) -5-methylhexanoic acid from the system. When the precipitation had stopped, the heterogeneous mixture was centrifuged and the expected pure (S) -3- ( ( ( (R) -1- ( (S) -2- (6-methoxynaphthalen-2-yl) propanoyloxy) ethoxy) carbonyl-amino) methyl) -5-methylhexanoic acid (Formula I) was collected.
Example 2: Alternative synthetic route of (S) -3- ( ( ( (R) -1- ( (S) -2- (6-methoxynaphthalen-2-yl) propanoyloxy) ethoxy) carbonyl-amino) methyl) -5-methylhexanoic acid

PAT
- Crystalline form of 1-(acyloxy)-alkyl carbamate drug complex of naproxen and pregabalinPublication Number: JP-7441181-B2Priority Date: 2018-05-14Grant Date: 2024-02-29
- Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: TW-I837128-BPriority Date: 2018-05-14Grant Date: 2024-04-01
- Method for preparing 1-(acyloxy)-alkyl carbamate drug complex of naproxen and pregabalin and intermediate thereofPublication Number: KR-102750784-B1Priority Date: 2018-05-14Grant Date: 2025-01-06
- GABA conjugates and methods of use thereofPublication Number: US-9186341-B2Priority Date: 2008-10-08Grant Date: 2015-11-17
- Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: US-2021221768-A1Priority Date: 2018-05-14
- Crystalline morphology of the 1- (acyloxy) -alkylcarbamate drug complex of naproxen and pregabalinPublication Number: JP-2021530434-APriority Date: 2018-05-14
- Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: AU-2019271799-B2Priority Date: 2018-05-14Grant Date: 2023-10-12
- Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: EP-4227293-A2Priority Date: 2018-05-14
- Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: EP-4227293-A3Priority Date: 2018-05-14
- Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: AU-2019271799-A1Priority Date: 2018-05-14
- crystalline forms of 1- (acyloxy) -alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: BR-112020022885-A2Priority Date: 2018-05-14
- Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: CN-112424158-APriority Date: 2018-05-14
- Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: EP-3793974-A1Priority Date: 2018-05-14
- Crystalline form of 1-(acyloxy)-alkyl carbamate drug complex of naproxen and pregabalinPublication Number: KR-20210013081-APriority Date: 2018-05-14
- Naproxen and pregabalin 1-(acyloxy)-alkyl carbamate drug conjugate purification processPublication Number: TW-202436285-APriority Date: 2022-11-24
- Naproxen and pregabalin 1- (acyloxy) -alkyl carbamate drug conjugate purification processPublication Number: WO-2024109817-A1Priority Date: 2022-11-24
- Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalinPublication Number: WO-2019219000-A1Priority Date: 2018-05-14
- Process for making 1-(acyloxy)-alkyl-carabmate drug conjugates of naproxen and pregabalinPublication Number: CA-3099775-A1Priority Date: 2018-05-14
- Naproxen (NAPROXEN) and pregabalin (PREGABALIN) 1-(acetyl)-alkylcarbamate drug conjugate crystalline formPublication Number: TW-202016065-APriority Date: 2018-05-14



AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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

……
.//////////Pregabalin naproxencarbil, gabamimetic, analgesic, ZVG8DDT3FJ
Plosaracetam



Plosaracetam
CAS 1651179-19-9
MF C13H10ClF3N4O MW330.69 g/mol
(4R)-1-[(5-chloro-1H-1,2,4-triazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one
(4R)-1-[(5-chloro-1,2,4-triazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one
(4R)-1-[(5-Chloro-1H-1,2,4-triazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)-2-pyrrolidinone
(4R)-1-[(5-chloro-1H-1,2,4-triazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one
2-Pyrrolidinone, 1-[(5-chloro-1H-1,2,4-triazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)-, (4R)-
synaptic vesicle glycoprotein 2A (SV2A) positive modulator, ABBV-552, ABBV552, SDI-118, SDI118, ABBV 552, ABBV552, SDI 118, SDI118, W3LYF2KQ6F
Plosaracetam (INNTooltip International Nonproprietary Name; developmental code names ABBV-552, SDI-118) is a synaptic vesicle glycoprotein 2A (SV2A) ligand which is under development for the treatment of Alzheimer’s disease and other cognition disorders.[1][3][4][2] In contrast to earlier SV2A ligands like levetiracetam and brivaracetam, polsaracetam does not have anticonvulsant activity and instead shows pro-cognitive effects.[2] The drug is being developed by UCB Biopharma and AbbVie.[1][3] As of October 2024, it is in phase 2 clinical trials for Alzheimer’s disease and phase 1 trials for cognition disorders.[1][3]
Plosaracetam is a small molecule drug. The usage of the INN stem ‘-racetam’ in the name indicates that Plosaracetam is a piracetam type amide type nootrope agent. Plosaracetam is under investigation in clinical trial NCT05199142 (A Study to Evaluate the Safety, Tolerability, and Pharmacodynamics of SDI-118 in Elderly Male and Female Study Participants With Cognitive Decline). Plosaracetam has a monoisotopic molecular weight of 330.05 Da.
PAT
- Compounds for Enhancing the Cognitive FunctionPublication Number: US-2016185761-A1Priority Date: 2013-08-02
- Compounds for enhancing the cognitive functionPublication Number: US-9630948-B2Priority Date: 2013-08-02Grant Date: 2017-04-25
- Compounds for enhancing the cognitive functionPublication Number: WO-2015014785-A1Priority Date: 2013-08-02
- Connections to improve cognitive functionPublication Number: DK-3027606-T3Priority Date: 2013-08-02Grant Date: 2018-04-30
- Compounds for enhancing the cognitive functionPublication Number: EP-3027606-A1Priority Date: 2013-08-02
- Compounds for enhancing the cognitive functionPublication Number: EP-3027606-B1Priority Date: 2013-08-02Grant Date: 2018-02-28
- Compounds to enhance cognitive functionPublication Number: ES-2666134-T3Priority Date: 2013-08-02Grant Date: 2018-05-03
- Compounds for enhancing the cognitive functionPublication Number: SI-3027606-T1Priority Date: 2013-08-02
SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2015014785&_cid=P11-MI5R7J-79014-1
Example 1 : Synthesis of (4R)-1 -[(5-chloro-1H-1,2,4-triazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 7.

1.1 Synthesis of tert-butyl 2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidine-1 -carboxylate 3 and enantiomers.
To a solution of tert-butyl 2-oxo-2,5-dihydro-1 H-pyrrole-1-carboxylate 1 (10 g, 1 eq., 54.6 mmol) in dioxane/water (100 ml/30 ml) are added at room temperature (3,4,5-trifluorophenyl)boronic acid 2 (19.2 g, 2 eq., 109.2 mmol), cesium fluoride (24.9 g, 3 eq., 163.8 mmol), (±)-2,2′-bis(diphenyl-phosphino)-1 , 1′-binaphthyl (1.5 g, 4.5%, 2.5 mmol), potassium carbonate (22.6 g, 3 eq., 163.8 mmol) and chloro(1 ,5-cyclooctadiene)rhodium(l)dimer (0.82 g, 1.5%, 8.2 mmol). The mixture is heated at 1 10°C for 2 h. Solvent are removed under reduced pressure and the residue is purified by chromatography over silicagel (eluent: CI-^C^/MeOH/NI-^OH 96/3.5/0.5 v/v/v) to afford tert-butyl 2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidine-1-carboxylate 3. The enantiomers are
resolved by chiral chromatography (chiralpak IC, 150*4.6 mm, eluent: heptane/AcOEt/diethylamine 80/20/0.1 v/v/v) to afford tert-butyl (4R)-2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidine-1-carboxylate 3A (second eluted, 5.1 g), and its enantiomer tert-butyl (4S)-2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidine-1-carboxylate 3B (first eluted, 5.2 g) as white solids.
Compound 3A:
Yield: 30%.
LC-MS (MH+): 316.
alphaD (MeOH, 25°C): -19.9.
1.2 Synthesis of (4R)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 4.
At 0°C, TFA (20 ml, 261 mmol) is added to a solution of tert-butyl (4R)-2-oxo-4-(3,4,5-trifluorophenyl)pyrrolidine-1-carboxylate 3A (8 g, 1 eq., 25.4 mmol) in dichloromethane (100 ml). The mixture is stirred at room temperature for 2 h. Then, TFA and solvent are removed under reduced pressure. The crude mixture is poured in an aqueous saturated solution of NaHCC>3 (100 ml) and extracted with AcOEt (3*200 ml). The combined organic extracts are dried over MgS04 and concentrated under reduced pressure. The conversion is total and the evaporation affords 5.5 g of (4R)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 4, which is used in the next step without any further purification.
LC-MS (MH+): 216; LC-MS (MKT): 214.
alphaD (MeOH, 22°C): -20.1.
1.3 Synthesis of (4R)-1 -(hydroxymethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 5.
To a solution of (4R)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 4 (5.5 g, 1 eq., 25.6 mmol) in THF (20 ml) are added potassium tert-butoxide (0.049 g, 0.02 eq., 0.44 mmol) and paraformaldehyde (0.95 g, 1.2 eq., 31.1 mmol) at room temperature. After overnight stirring at 60°C, the mixture is quenched with brine (100 ml) and the aqueous phase is extracted with AcOEt (2*100 ml). The combined organic extracts are dried over MgS04 and concentrated under reduced pressure yielding 4.7 g of (4R)-1-(hydroxymethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 5, which is used in the next step without any further purification.
LC-MS (MH+): 246.
H NMR (DMSO) δ 7.34 (dd, J-| =9.2 Hz, J2=6.8 Hz, 2 H), 5.87 (t, J=6.8 Hz, 1 H), 4.70 (m, 2 H), 3.78 (m, 1 H), 3.62 (m, 1 H), 3.40 (m, 1 H), 2.68 (m, 1 H), 2.43 (dd, J<l =16.6 Hz, J2=8.6 Hz, 1 H).
1.4 Synthesis of (4R)-1 -[(5-chloro-1 H-1 ,2,4-triazol-1-yl)methyl]-4-(3,4,5-trifluoro- phenyl)pyrrolidin-2-one 7.
1 ) To a cold solution (0°C) of (4R)-1-(hydroxymethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 5 (4.7 g, 1 eq., 19.4 mmol) in CH2CI2 (200 mL) is added oxalyl chloride (3.7 ml, 2 eq., 38 mmol). After stirring for 30 minutes at 0°C, the reaction mixture is evaporated in vacuum yielding (4R)-1-(chloromethyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 6 which is dissolved in THF (100 ml) to afford Solution A.
2) To a cold solution (0°C) of 5-chloro-1 H-1 ,2,4-triazole (3.0 g, 1.5 eq., 29.1 mmol) in THF (100 ml) is added NaH 95% in mineral oil (0.9 g, 2 eq., 38.7 mmol). The reaction mixture is stirred during 30 minutes at 0°C to afford Solution B.
3) Solution A is added to solution B at 0°C and the reaction mixture is maintained under stirring overnight at room temperature. The mixture is quenched with water (100 ml) and extracted with AcOEt (2*100 mL). The combined organic extracts are washed with brine (100 ml), dried over MgS04 then concentrated under reduced pressure yielding 7 g of compound 7 as crude material. The crude residue is purified by chromatography on silicagel (eluent: CH2Cl2/MeOH/NH4OH 95/5/0.5 v/v/v) and recrystallized from iPr20/EtOH affording 1.6 g of (4R)-1-[(5-chloro-1 H-1 ,2,4-triazol-1-yl)methyl]-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one 7 as a white solid.
Yield: 25%.
LC-MS (MH+): 331/333.
H NMR (DMSO) δ 8.12 (s, 1 H), 7.32 (dd, J-| =9.2 Hz, J2=6.9 Hz, 2 H), 5.63 (d, J=1.5 Hz, 2 H), 3.81 (t, J=8.6 Hz, 1 H), 3.62 (t, J=8.4 Hz, 1 H), 3.39 (m, 1 H), 2.71 (dd, J<l =16.7 Hz, J2=8.8 Hz, 1 H), 2.54 (d, J=9.1 Hz, 1 H).
alphaD (MeOH, 25°C): + 9.2.



AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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 names | ABBV-552; ABBV552; SDI-118; SDI118 |
| Routes of administration | Oral[1] |
| Drug class | Synaptic vesicle glycoprotein 2A (SV2A) ligand[2] |
| Identifiers | |
| IUPAC name | |
| CAS Number | 1651179-19-9 |
| PubChem CID | 90467376 |
| ChemSpider | 129532952 |
| UNII | W3LYF2KQ6F |
| KEGG | D13077 |
| ChEMBL | ChEMBL5314929 |
| Chemical and physical data | |
| Formula | C13H10ClF3N4O |
| Molar mass | 330.70 g·mol−1 |
| 3D model (JSmol) | Interactive image |
| SMILES | |
| InChI | |
References
- “ABBV 552”. AdisInsight. 28 October 2024. Retrieved 26 February 2025.
- Botermans W, Koole M, Van Laere K, Savidge JR, Kemp JA, Sunaert S, et al. (2022). “SDI-118, a novel procognitive SV2A modulator: First-in-human randomized controlled trial including PET/fMRI assessment of target engagement”. Frontiers in Pharmacology. 13 1066447. doi:10.3389/fphar.2022.1066447. PMC 9887116. PMID 36733374.
- “Delving into the Latest Updates on Plosaracetam with Synapse”. Synapse. 22 February 2025. Retrieved 26 February 2025.
- “ABBV-552”. ALZFORUM. 28 February 2023. Retrieved 26 February 2025.
/////////Plosaracetam, ABBV-552, ABBV552, SDI-118, SDI118, ABBV 552, ABBV552, SDI 118, SDI118, W3LYF2KQ6F
Padoprazan


Padoprazan
CAS 2756367-23-2
MF C19H20FN3O4S MW 405.4 g/mol
1-[5-(2-fluorophenyl)-4-methoxy-1-(6-methoxypyridine-3-sulfonyl)-1Hpyrrol-3-yl]-N-methyl methanamine
1-[5-(2-fluorophenyl)-4-methoxy-1-[(6-methoxy-3-pyridinyl)sulfonyl]pyrrol-3-yl]-N-methylmethanamine
1-(5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrol-3-yl)- N -methylmethanamine
proton pump inhibitor, 95BJ28E2RP, ID-120040002, ID 120040002
Padoprazan is a new-generation potassium-competitive acid blocker (P-CAB) used to treat acid-related disorders like gastroesophageal reflux, according to MedchemExpress.com and Patsnap Synapse. It works by inhibiting the proton pump in the stomach and is different from traditional proton pump inhibitors (PPIs) because it is not dependent on acid activation. Padoprazan is currently undergoing Phase 3 clinical trials in Korea, notes THE BIO (더바이오).
Key facts about padoprazan
- Drug class: Potassium-competitive acid blocker (P-CAB), a type of proton pump inhibitor, according to DrugBank and GlpBio.
- Mechanism: It inhibits the proton pump in the stomach to reduce acid production and is not acid-activated like older PPIs, per DrugBank.
- Indications: Used for acid-related conditions like gastroesophageal reflux, reports Patsnap Synapse.
- Status: Currently undergoing Phase 3 clinical trials in Korea, says THE BIO (더바이오).
- Development: It is a new-generation drug being developed by companies like Daewon Pharmaceutical.
Padoprazan is a small molecule drug. The usage of the INN stem ‘-prazan’ in the name indicates that Padoprazan is a proton pump inhibitor, not dependent on acid activation. Padoprazan has a monoisotopic molecular weight of 405.12 Da.
PAT
- NEW INHIBITOR OF ACID SECRETION AND USE OF THE SAMEPublication Number: PE-20231652-A1Priority Date: 2020-06-17
- Novel acid secretion inhibitors and use thereofPublication Number: TW-I839161-BPriority Date: 2020-06-17Grant Date: 2024-04-11
- Novel acid secretion inhibitor and use thereofPublication Number: US-2023373954-A1Priority Date: 2020-06-17
- Novel acid secretion inhibitor and use thereofPublication Number: EP-4148050-B1Priority Date: 2020-06-17Grant Date: 2024-12-18
- Novel acid secretion inhibitors and use thereofPublication Number: TW-I797645-BPriority Date: 2020-06-17Grant Date: 2023-04-01
- Acid secretion inhibitor and use thereofPublication Number: US-11767311-B2Priority Date: 2020-06-17Grant Date: 2023-09-26
- Novel Acid Secretion Inhibitor and use thereofPublication Number: AU-2021293694-B2Priority Date: 2020-06-17Grant Date: 2023-12-21
- Novel acid secretion inhibitor and use thereofPublication Number: CN-115884968-BPriority Date: 2020-06-17Grant Date: 2024-06-21
- Novel acid secretion inhibitors and their usesPublication Number: JP-7404561-B2Priority Date: 2020-06-17Grant Date: 2023-12-25
- Novel acid secretion inhibitors and use thereofPublication Number: KR-102432523-B1Priority Date: 2020-06-17Grant Date: 2022-08-16
- Novel acid secretion inhibitor and use thereofPublication Number: CN-115884968-APriority Date: 2020-06-17
- Novel acid secretion inhibitor and use thereofPublication Number: JP-2023524172-APriority Date: 2020-06-17
- Novel acid secretion inhibitor and use thereofPublication Number: US-2023192650-A1Priority Date: 2020-06-17
- Novel acid secretion inhibitors and use thereofPublication Number: TW-202325702-APriority Date: 2020-06-17
- Novel acid secretion inhibitor and use thereofPublication Number: WO-2021256861-A1Priority Date: 2020-06-17
- Novel acid secretion inhibitors and use thereofPublication Number: TW-202214588-APriority Date: 2020-06-17
- Novel Acid Secretion Inhibitor and use thereofPublication Number: AU-2021293694-A1Priority Date: 2020-06-17
- Novel acid secretion inhibitor and use thereofPublication Number: CA-3182882-A1Priority Date: 2020-06-17
- Novel acid secretion inhibitor and use thereofPublication Number: EP-4148050-A1Priority Date: 2020-06-17
- Novel salt of 1-sulfonyl pyrrole derivative, preparation method thereof and pharmaceutical composition comprising thereofPublication Number: TW-I828476-BPriority Date: 2021-12-15Grant Date: 2024-01-01
- Novel salt of 1-sulfonyl pyrrole derivative, preparation method thereof and pharmaceutical composition comprising thereofPublication Number: WO-2023113458-A1Priority Date: 2021-12-15
- Novel salt of 1-sulfonyl pyrrole derivative, method for preparing same, and pharmaceutical composition including samePublication Number: WO-2023113474-A1Priority Date: 2021-12-15
- Novel salt of 1-sulfonyl pyrrole derivative, method for preparing same, and pharmaceutical composition including samePublication Number: US-2025042872-A1Priority Date: 2021-12-15
- Novel acid secretion inhibitors and use thereofPublication Number: KR-20210156234-APriority Date: 2020-06-17
- Novel salt of 1-sulfonylpyrrole derivative, preparation method thereof and pharmaceutical composition comprising the samePublication Number: CN-118541361-APriority Date: 2021-12-15
- Novel salt of 1-sulfonyl pyrrole derivative, preparation method thereof and pharmaceutical composition comprising thereofPublication Number: KR-20230091056-APriority Date: 2021-12-15
- Novel salts of 1-sulfonyl pyrrole derivatives, methods for producing the same, and pharmaceutical compositions containing the samePublication Number: KR-20240119083-APriority Date: 2021-12-15
- Novel salt of 1-sulfonyl pyrrole derivative, preparation method thereof and pharmaceutical composition comprising thereofPublication Number: TW-202334114-APriority Date: 2021-12-15
- Novel formulation comprising acid secretion inhibitorsPublication Number: KR-20240161598-APriority Date: 2023-05-04
- Method for preparation of 6-methoxypyridine-3-yl derivativesPublication Number: TW-202411216-APriority Date: 2022-05-23
- Method for preparing 6-methoxypyridin-3-yl derivativesPublication Number: WO-2023229322-A1Priority Date: 2022-05-23
- Method for preparation of 6-methoxypyridine-3-yl derivativesPublication Number: KR-20230163283-APriority Date: 2022-05-23
- NOVEL SALT OF A DERIVATIVE OF 1-SULFONYLPYRROL, METHOD OF PREPARATION THEREOF AND PHARMACEUTICAL COMPOSITION THAT INCLUDES THE SAMEPublication Number: AR-127964-A1Priority Date: 2021-12-15
SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021256861&_cid=P22-MI13VU-05837-1



Synthesis Example 1. Synthesis of Example 1
[267]
[Example 1] 1-(5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrol-3-yl)- N -methylmethanamine
[268]
(1) Synthesis of step methyl 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrole-3-carboxylate
[269]Methyl 5-(2-fluorophenyl)-4-methoxy-1
H -pyrrole-3-carboxylate (intermediate 1, 1.0 eq., 1.2 g, 4.8 mmol) was dissolved in THF (20.0 mL), and NaH (2.0 eq., 384.8 mg, 9.6 mmol) was added dropwise at 0 °C and stirred at room temperature for 10 min. 6-Methoxypyridine-3-sulfonyl chloride (1.5 eq., 1.6 g, 7.2 mmol) was added and stirred at room temperature for 1 h. Water was added to the reaction solution, and the mixture was extracted with EA. The organic layer was dried over anhydrous magnesium sulfate, filtered, concentrated, and purified by column chromatography to obtain methyl 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrole-3-carboxylate as a light brown solid. (1.85 g, 91.6%)
[270]
(2) Synthesis of step 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrol-3-yl)methanol
[271]Methyl 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrole-3-carboxylate (1.0 eq., 1.0 g, 2.38 mmol) was dissolved in THF (5.0 mL), and 1.0 M DIBAL in
n -hexane solution (5.0 eq., 11.9 mL, 11.9 mmol) was added dropwise at 0 °C, followed by stirring at room temperature for 1 h. The reaction solution was cooled to 0 °C, quenched with an aqueous Rochelle salt solution, and extracted with EA. The organic layer was dried over anhydrous magnesium sulfate, filtered, concentrated, and purified by column chromatography to obtain 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrol-3-yl)methanol as a yellow oil. (654.8 mg, 70.2%)
[272]
(3) Synthesis of step 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrole-3-carbaldehyde
[273]5-(2-Fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrol-3-yl)methanol (1.0 eq., 500.0 mg, 1.3 mmol) and Dess-Martin periodinane (1.0 eq., 540.4 mg, 1.3 mmol) were dissolved in DCM (10.0 mL) and stirred at room temperature for 1 h. The reaction mixture was concentrated and purified by column chromatography to give 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrole-3-carbaldehyde as a pale brown solid. (388.2 mg, 78.1%)
[274]
(4) Step 1 Synthesis of (5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrol-3-yl)- N -methylmethanamine
[275]5-(2-Fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrole-3-carbaldehyde (1.0 eq., 385.0 mg, 0.99 mmol) was dissolved in THF (5.0 mL), and 2.0 M methylamine in THF (10 eq., 4.9 mL, 9.9 mmol) was added. After stirring at room temperature for 1 h, the reaction mixture was cooled to 0 °C, and NaBH
4 (10 eq., 373.4 mg, 9.9 mmol) was added, followed by stirring at room temperature for 1 h. 6.0
N aqueous hydrogen chloride solution was slowly added dropwise to the reaction solution, and the resulting solid was filtered. The filtered solid was dissolved in water, 1
N aqueous sodium hydroxide solution was added, and extraction was performed with EA. The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated to obtain 1-(5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrol-3-yl)-
N -methylmethanamine as a white solid. (125.8 mg, 28.3%) [M+H] + : 405
SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2023113474&_cid=P22-MI1405-08231-1

7) Preparation of 1-(5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine free base[211]
(1) Step: Synthesis of methyl 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrole-3-carboxylate[212]Methyl 5-(2-fluorophenyl)-4-methoxy-1
H -pyrrole-3-carboxylate (intermediate 1, 1.0 eq., 920 g, 3.69 mol) was dissolved in DMF (9.2 L), and t-BuOK (2.0 eq., 828 g, 7.38 mmol) was added dropwise at 0 °C and stirred for 30 min. 6-Methoxypyridine-3-sulfonyl chloride (1.5 eq., 1.15 kg, 5.54 mol) was added and stirred at 0 °C for 1 h. Water was added to the reaction solution, which was then extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate, filtered, concentrated, and purified by column chromatography to obtain methyl 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrole-3-carboxylate as a white solid. (1.20 kg, 77.4%) [213]
(2) Step: Synthesis of 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrol-3-yl)methanol[214]Methyl 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrole-3-carboxylate (1.0 eq., 1.1 kg, 2.62 mol) was dissolved in THF (11.0 L), and DIBAL 2.0 M in THF solution (3.0 eq., 3.93 L, 7.86 mol) was added dropwise at 0 °C, followed by stirring for 30 min. The reaction solution was quenched with 5% aqueous Rochelle’s salt solution and extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated to obtain 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrol-3-yl)methanol as a light yellow oil. (870 g, 84.8%) [215]
(3) Step: Synthesis of 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrole-3-carbaldehyde[216]5-(2-Fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrol-3-yl)methanol (1.0 eq., 830 g, 2.12 mol) and TEA (4.0 eq., 1.59 kg, 15.7 mol) were dissolved in DMSO (4.15 L), and SO
3 -pyridine (4.0 eq., 1.35 kg, 8.48 mol) was added dropwise, and the mixture was stirred at room temperature for 1.5 h. Water was added to the reaction mixture at 0 °C, and the mixture was extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate, filtered, and concentrated to obtain 5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrole-3-carbaldehyde as a yellow solid. (722 g, 87.6%) [217]
(4) Step: Synthesis of 1-(5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrol-3-yl)- N -methylmethanamine[218]5-(2-Fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1
H -pyrrole-3-carbaldehyde (1.0 eq., 715 g, 1.83 mol) was dissolved in methanol (7.2 L), and methylamine in methanol (5.0 eq., 916 g, 9.16 mol) was added. After stirring at room temperature for 1 h, the reaction mixture was concentrated, dissolved in ethanol (7.2 L), cooled to 0 °C, and NaBH
4 (2.0 eq., 139 g, 3.66 mol) was added, and stirred at 0 °C for 1 h. Water was added to the reaction solution, and extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate, filtered, concentrated, and purified by column chromatography to obtain 1-(5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1 H -pyrrol-3-yl)-
N -methylmethanamine as a brown oil. (347 g, 46.7%)
<Example 1> Preparation of hydrochloric acid salt of 1-(5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine About 500 mg of the free base of 1-(5-(2-fluorophenyl)-4-methoxy-1-((6-methoxypyridin-3-yl)sulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine was weighed and placed in a glass vial, and then dissolved in 2 mL of ethanol while heating at 25°C. Then, 647.44 μL (2 M) hydrochloric acid was added to the vial. The sample was continuously stirred on a magnetic stirrer at room temperature for 24 hours, and after stirring for 24 hours, the solid precipitate was separated by centrifugation. Subsequently, the wet solid was dried at 40°C for 20 hours to obtain a grayish white dried powder.
SYN



AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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

……
/////////Padoprazan, proton pump inhibitor, 95BJ28E2RP, ID-120040002, ID 120040002
Ofirnoflast


Ofirnoflast
CAS 2731294-23-6
MFC23H19F4N7O2 MW501.4 g/mol
N-[4-(4-amino-7-cyclopropyl-7H-pyrrolo[2,3-d]pyrimidin-5-yl)-2-fluorophenyl]-N’-{5-[1-
(trifluoromethyl)cyclopropyl]-1,2-oxazol-3-yl}urea
N-(4-(4-AMINO-7-CYCLOPROPYL-7H-PYRROLO(2,3-D)PYRIMIDIN-5-YL)-2-FLUOROPHENYL)-N’-(5-(1-(TRIFLUOROMETHYL)CYCLOPROPYL)-3-ISOXAZOLYL)UREA
N-(4-(4-AMINO-7-CYCLOPROPYL-7H-PYRROLO(2,3-D)PYRIMIDIN-5-YL)-2-FLUOROPHENYL)-N’-(5-(1-(TRIFLUOROMETHYL)CYCLOPROPYL)-1,2-OXAZOL-3-YL)UREA
OFIRNOLAST [USAN]
OFIRNOFLAST
UREA, N-(4-(4-AMINO-7-CYCLOPROPYL-7H-PYRROLO(2,3-D)PYRIMIDIN-5-YL)-2-FLUOROPHENYL)-N’-(5-(1-(TRIFLUOROMETHYL)CYCLOPROPYL)-3-ISOXAZOLYL)-
OFIRNOFLAST [INN]
serine/ threonine-protein kinase Nek7 inhibitor, antiinflammatory, HT-6184, HT 6184, 54PY2PBN7S
Ofirnoflast is an investigational drug, a NEK7 inhibitor, that targets and disrupts the formation of the NLRP3 inflammasome, a key driver of chronic inflammation. Developed by Halia Therapeutics, it is being explored for conditions like myelodysplastic syndromes (MDS), obesity, and Alzheimer’s disease. The drug’s unique mechanism aims to address inflammation at a root cause level, potentially offering a new approach to treating these diseases.
How it works
- Ofirnoflast is a “first-in-class” molecule that selectively inhibits the NEK7 protein.
- NEK7 is essential for the assembly of the NLRP3 inflammasome, a molecular complex that causes chronic inflammation.
- By inhibiting NEK7, ofirnoflast prevents the inflammasome from forming and promotes its disassembly.
- This approach aims to reduce inflammation without causing broad immunosuppression.
Therapeutic applications
- Myelodysplastic Syndromes (MDS): Ofirnoflast has completed a Phase 2 study for this condition and received Orphan Drug Designation from the FDA. It is being investigated for its potential to improve blood cell production by targeting the underlying inflammation.
- Obesity: An ongoing Phase 2 study is exploring ofirnoflast in combination with semaglutide to target inflammation and metabolic issues.
- Alzheimer’s Disease: Ofirnoflast is part of an early-stage program looking into its potential for this disease.
Ofirnoflast is a first-in-class, orally bioavailable NEK7 inhibitor currently undergoing Phase 2 clinical evaluation. It disrupts NLRP3 inflammasome assembly by targeting NEK7’s scaffolding function—blocking complex formation independently of NLRP3 activation status, upstream of caspase activation, pyroptosis, and inflammatory cytokine release. This mechanism offers a novel therapeutic approach for chronic inflammation. Unlike NSAIDs, corticosteroids, cytokine-neutralising biologics, and NLRP3-directed small molecules—which are frequently limited by off-target effects, immunosuppression, or incomplete efficacy—ofirnoflast provides a targeted approach with fewer anticipated liabilities
- A Ph2 Study to Evaluate the Safety, Efficacy and Tolerability of HT-6184 and Semaglutide in Obese Participants With T2DMCTID: NCT07172867Phase: Phase 2Status: Not yet recruitingDate: 2025-09-15
- HT-6184 in Subjects With MDSCTID: NCT07052006Phase: Phase 2Status: Active, not recruitingDate: 2025-07-14
- Evaluating Ability of HT-6184 to Reduce Inflammation and Pain After Third Molar ExtractionCTID: NCT06241742Phase: Phase 2Status: CompletedDate: 2025-03-30
- Study to Evaluate HT-6184 in Healthy SubjectsCTID: NCT05447546Phase: Phase 1Status: CompletedDate: 2023-08-28
SYN
https://www.tandfonline.com/doi/full/10.1080/1061186X.2025.2542856
SYN
COMPD 10
SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021242505&_cid=P11-MHZPDU-32878-1


INTERMEDIATE D1
5-(4-AMINO-3-FLUOROPHENYL)-7-CYCLOPROPYL-7H-PYRROLO[2,3-D]PYRIMIDIN-4- AMINE

A mixture of 7-cyclopropyl-5-iodo-7H-pyrrolo[2,3-d]pyrimidin-4-amine (C1, 0.160 g, 0.533 mmol), 2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (0.190 g, 0.800 mmol), and K2CO3 (0.221 g, 1.599 mmol) in 1,4-dioxane (1 mL) and water (0.3 mL) was purged with N2 for 10 min. Pd(PPh3)4 (0.062 g, 0.053 mmol) was then added and the reaction mixture was stirred at 100 °C for 12 h. Following completion of the reaction (as indicated by TLC), the mixture was filtered through a pad celite which was then rinsed with EtOAc (2 x 10 mL). The combined filtrates were concentrated under reduced pressure to yield crude material which was purified by flash chromatography (silica gel 230-400 mesh, eluting with 3% MeOH in DCM), affording
the title compound as a yellow solid (0.110 g, 73% yield).1H NMR (400 MHz, DMSO-d6) δ = 8.14 (s, 1H), 7.13 (s, 1H), 7.05-7.09 (m, 1H), 6.95-6.98 (m, 1H), 6.82-6.86 (m, 1H), 6.10 (bs, 2H), 5.22 (bs, 2H), 3.52-3.58 (m, 1H), 1.00-1.04 (m, 4H). LCMS: 284.1 [M+H].

3-(1-(Trifluoromethyl)cyclopropyl)isoxazol-5-amine (precursor to E6) and 5-(1-(trifluoromethyl)cyclopropyl)isoxazol-3-amine (precursor to E7) were synthesized as reported in Synthesis 2013, 45, 171–173
EXAMPLE 5
1-(4-(4-AMINO-7-CYCLOPROPYL-7H-PYRROLO[2,3-D]PYRIMIDIN-5-YL)-2- FLUOROPHENYL)-3-(3-(1-(TRIFLUOROMETHYL)CYCLOPROPYL)ISOXAZOL-5-YL)UREA

The title compound was prepared following the general procedure for urea formation (Method A), starting from 5-(4-amino-3-fluorophenyl)-7-cyclopropyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine (D1, 0.080 g, 0.282 mmol) and phenyl (3-(1-(trifluoromethyl)cyclopropyl)isoxazol-5-yl)carbamate (E6, 0.088 g, 0.282 mmol), and was obtained as a white solid (0.031 g, 22% yield).1H NMR (400 MHz, DMSO-d6) δ = 10.59 (bs, 1H), 8.84 (bs, 1H), 8.11-8.17 (m, 2H), 7.26-7.37 (m, 3H), 6.20 (s, 1H), 6.16 (bs, 2H), 3.55-3.61 (m, 1H), 1.45-1.49 (m, 2H), 1.38-1.43 (m, 2H), 1.03-1.08 (m, 4H). LCMS: 502.1 [M+H].
PAT
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2024249257&_cid=P11-MHZP9H-30149-1



PAT
- Targeted nek7 inhibition for modulation of the nlrp3 inflammasomePublication Number: US-2023210853-A1Priority Date: 2020-05-08
- Inhibitors of NEK7 kinasePublication Number: US-11713321-B2Priority Date: 2020-05-08Grant Date: 2023-08-01
- Inhibitors of nek7 kinasePublication Number: EP-4146348-B1Priority Date: 2020-05-08Grant Date: 2024-07-03
- Inhibitors of nek7 kinasePublication Number: US-2023416259-A1Priority Date: 2020-05-08
- Inhibitors of NEK7 kinasePublication Number: US-12091413-B2Priority Date: 2020-05-08Grant Date: 2024-09-17
- Inhibitors of nek7 kinasePublication Number: TW-202208356-APriority Date: 2020-05-08
- Inhibitors of NEK7 kinasePublication Number: AU-2021280893-A1Priority Date: 2020-05-08
- Inhibitors of NEK7 kinasePublication Number: CN-115843272-APriority Date: 2020-05-08
- Inhibitors of nek7 kinasePublication Number: EP-4146348-A1Priority Date: 2020-05-08
- Inhibitors of NEK7 kinasePublication Number: KR-20230008763-APriority Date: 2020-05-08
- Polymorphs of nek 7 inhibitorsPublication Number: WO-2024249257-A1Priority Date: 2023-05-26
- Inhibitors of NEK7 kinasePublication Number: US-11161852-B1Priority Date: 2020-05-08Grant Date: 2021-11-02
- Inhibitors of nek7 kinasePublication Number: US-2021355130-A1Priority Date: 2020-05-08
- Inhibitors of nek7 kinasePublication Number: US-2022064173-A1Priority Date: 2020-05-08
- Inhibitors of nek7 kinasePublication Number: WO-2021242505-A1Priority Date: 2020-05-08



AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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

……
///////////ofirnoflast, serine/ threonine-protein kinase Nek7 inhibitor, antiinflammatory, HT-6184, HT 6184, 54PY2PBN7S
Nurandociguat


Nurandociguat
CAS 2781965-75-9
MF C30H36ClF2N5O2 MW 572.1 g/mol
1-[(3R)-1-{4-chloro-4′-[4-(2-methylpropyl)piperazin-1-yl][1,1′-biphenyl]-2-yl} piperidin-3-yl]-5-(difluoromethyl)-1H-pyrazole-4-carboxylic acid
1-[(3R)-1-[5-chloro-2-[4-[4-(2-methylpropyl)piperazin-1-yl]phenyl]phenyl]piperidin-3-yl]-5-(difluoromethyl)pyrazole-4-carboxylic acid
guanylate cyclase activator, BAY 3283142, LPU8429UK5
Nurandociguat is a small molecule drug candidate, previously known as BAY 3283142, that is a guanylate cyclase activator being developed by Bayer for cardiovascular conditions. The “ciguat” stem in its name indicates its function as a guanylate cyclase activator, a mechanism that is also being investigated for related drugs like runcaciguat. It is currently in clinical trials, including a Phase 2 program for chronic kidney disease (CKD).
- Drug class: Guanylate cyclase activator
- Developer: Bayer
- Previous name: BAY 3283142
- Indication: Investigated for cardiovascular conditions
- Current status: In clinical development, including a Phase 2 study for chronic kidney disease (CKD)
- OriginatorBayer
- ClassAntihypertensives; Cardiovascular therapies; Hepatoprotectants; Urologics
- Mechanism of ActionGuanylate cyclase stimulants
- Phase IIRenal failure
- Phase ICardiovascular disorders; Diabetic retinopathy; Hypertension; Liver disorders
- 28 Sep 2025No recent reports of development identified for phase-I development in Renal-failure in Germany (PO, Immediate release)
- 16 Sep 2025(CTIS2024-510856-11-00) (EudraCT2024-510856-11-00): Trial initiation and completion info added; updated DevT; Corrected intro to match DevT as most of the info about indication and countries missing
- 28 May 2025No recent reports of development identified for phase-I development in Renal-failure(In volunteers, In adults) in Japan (PO, Immediate release)
Nurandociguat is a small molecule drug. The usage of the INN stem ‘-ciguat’ in the name indicates that Nurandociguat is a guanylate cyclase activator and stimulator. Nurandociguat has a monoisotopic molecular weight of 571.25 Da.
PAT
- Soluble guanylate cyclase activators for use in the treatment of heart failure with preserved ejection fraction in womenPublication Number: WO-2023237577-A1Priority Date: 2022-06-09
- Substituted pyrazolo piperidine carboxylic acidsPublication Number: WO-2022122910-A1Priority Date: 2020-12-10
- The use of sgc activators for the treatment of ophthalmologic diseasesPublication Number: WO-2022122917-A1Priority Date: 2020-12-10
- Use of sGC activators for the treatment of ophthalmic diseasesPublication Number: CN-115175681-APriority Date: 2020-12-10
- Use of sgc activators for the treatment of ophthalmologic diseasesPublication Number: US-2022241273-A1Priority Date: 2020-12-10
- Use of sGC activators for the treatment of ophthalmic diseasesPublication Number: JP-2023514928-APriority Date: 2020-12-10
- The use of sgc activators for the treatment of ophthalmologic diseasesPublication Number: EP-4259140-A1Priority Date: 2020-12-10
- Use of sGC activators for the treatment of ophthalmic diseasesPublication Number: KR-20230118143-APriority Date: 2020-12-10
- Substituted pyrazolo piperidine carboxylic acidsPublication Number: US-2023265072-A1Priority Date: 2020-12-10
- Use of sGC activators for the treatment of ophthalmic diseasesPublication Number: JP-2024073585-APriority Date: 2020-12-10
- Use of sGC activators for the treatment of ophthalmological diseasesPublication Number: JP-7458683-B2Priority Date: 2020-12-10Grant Date: 2024-04-01
- Use of sgc activators for the treatment of ophthalmologic diseasesPublication Number: US-2023346777-A1Priority Date: 2020-12-10
- Use of sGC activators for treating ophthalmic diseasesPublication Number: CN-115175681-BPriority Date: 2020-12-10Grant Date: 2024-10-25
SYN
SYN
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2022122917&_cid=P20-MHVQYD-96133-1
soluble guanylate cyclase (sGC) activators for use in the treatment and/or prophylaxis of ophthalmologic diseases, including non-proliferative diabetic retinopathy (NPDR), diabetic macular edema (DME), retinal ganglion cell/photoreceptor neurodegeneration and cataract, especially wherein the soluble guanylate cyclase (sGC) activators are compounds selected from the group consisting of





Example 1
1 – [ 1 – { 4-Chloro-4′- [4-(2-methylpropyl)piperazin- 1 -yl] [1,1 ’-biphenyl] -2-yl }piperidin-3-yl] -5- (difluoromethyl)-lH-pyrazole-4-carboxylic acid hydrochloride (Enantiomer 1)

Ethyl 1 – [ 1 – { 5-chloro-2- [(trifluoromethanesulfonyl)oxy]phenyl }piperidin-3-yl] -5-(difluoromethyl)- 1 H-pyrazole-4-carboxylate (prepared in analogy to Example 11A, Enantiomer 1, 80.0 mg, 147 pmol) and l-(2-methylpropyl)-4- [4-(4,4,5 ,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)phenyl]piperazine (Example 18 A 62.8 mg, 97 % purity, 177 pmol) were placed under argon in toluene/ethanol (820/820 pl). 2 M sodium carbonate solution (220 pl, 2.0 M, 440 pmol) and tetrakis(triphenylphosphine)palladium(0) (8.52 mg, 7.37 pmol) were added and the mixture was stirred at 100°C. overnight. The reaction mixture was diluted with ethyl acetate and 1 M hydrochloric acid was added. The aqueous phase was extracted three times with ethyl acetate. The organic phase was dried with sodium sulfate, filtered off and evaporated. The crude mixture was dissolved with THF/ethanol (2.0/0.2 ml), 1 M lithium hydroxide solution (1.5 ml, 1.5 mmol) was added and the mixture was stirred at room temperature overnight. A I M lithium hydroxide solution (740 pl, 740 pmol) was added again. After about 6 h the reaction mixture was evaporated at 50°C. The residue was dissolved in
SUBSTITUTE SHEET (RULE 26)
acetonitrile/water/0.25 ml trifluoroacetic acid and purified by preparative HPLC (RP18 column, acetonitrile/water gradient with the addition of 0.1% trifluoroacetic acid). The crude product was purified by means of thick layer chromatography (dichloromethane/methanol/formic acid: 10/1/0.1). The silica gel mixture was stirred with dichloromethane/1 M hydrochloric acid in dioxane (10/1) in ethanol, filtered off and carefully evaporated at 30°C and lyophilized. 34 mg of the target compound (36% of theory, purity 95%) were obtained.
LC-MS (Method 6): Rt = 1.23 min; MS (ESIpos): m/z = 572 [M-HC1+H]+
‘H-NMR (600 MHz, DMSO-d6) 5 [ppm]: 1.004 (15.87), 1.015 (16.00), 1.500 (0.51), 1.521 (0.57), 1.728 (0.73), 1.750 (0.61), 1.897 (0.57), 1.917 (0.62), 1.975 (0.79), 2.122 (0.42), 2.133 (0.84), 2.144 (1.02), 2.156
(0.79), 2.571 (0.47), 2.587 (0.91), 2.610 (0.52), 3.004 (0.84), 3.022 (2.01), 3.026 (2.20), 3.038 (3.72), 3.048
(2.50), 3.065 (0.75), 3.154 (2.66), 3.161 (2.75), 3.169 (2.36), 3.177 (1.88), 3.224 (0.84), 3.237 (0.70), 3.589
(1.41), 3.602 (1.80), 3.825 (1.02), 3.841 (0.78), 3.866 (1.05), 3.882 (0.75), 4.223 (2.57), 4.445 (0.68), 4.463
(0.97), 4.481 (0.57), 7.045 (0.55), 7.055 (3.63), 7.070 (3.72), 7.084 (2.72), 7.087 (3.09), 7.110 (1.47), 7.113
(1.11), 7.123 (2.19), 7.127 (2.02), 7.163 (3.67), 7.177 (2.19), 7.215 (0.46), 7.428 (0.83), 7.495 (4.24), 7.510
(4.02), 7.515 (2.07), 7.602 (0.82), 7.959 (4.79), 9.484 (0.54).
Example 2
1 – [ 1 – { 4-Chloro-4′- [4-(2-methylpropyl)piperazin- 1 -yl] [1,1 ’-biphenyl] -2-yl }piperidin-3-yl] -5-(difluoromethyl)-lH-pyrazole-4-carboxylic acid (Enantiomer 2)

Method A
A solution of ethyl l-[l-{4-chloro-4′-[4-(2-methylpropyl)piperazin-l-yl][l,T-biphenyl]-2-yl}piperidin-3-yl]-5-(difluoromethyl)-lH-pyrazole-4-carboxylate (prepared in analogy to Example 17A, Enantiomer 2, 50.8 g, 84.6 mmol) in a THF/methanol mixture 9:1 (1.0 1) was treated with an aqueous solution of lithium hydroxide (850 ml, 1.0 M, 850 mmol) and stirred overnight at room temperature. The reaction mixture was
SUBSTITUTE SHEET (RULE 26)
concentrated, diluted with dichloromethane (1.5 1) and adjusted to pH = 2 with an aqueous solution of hydrogen chloride (2N). The resulting suspension was stirred 45 minutes at room temperature. The solid was filtered, washed with water and dried under vacuum affording 43 g (90 % yield) of the title compound.
LC-MS (Method 7): Rt = 1.27 min; MS (ESIpos): m/z = 572 [M+H]+
‘H-NMR (600 MHz, DMSO-d6) 5 [ppm]: 1.002 (15.68), 1.013 (16.00), 1.080 (0.57), 1.092 (1.18), 1.103 (0.63), 1.498 (0.74), 1.519 (0.83), 1.719 (1.03), 1.741 (0.88), 1.902 (0.78), 1.908 (0.74), 1.922 (0.88), 1.928
(0.83), 1.943 (0.45), 1.978 (1.13), 1.994 (0.74), 2.102 (0.71), 2.112 (0.85), 2.123 (0.70), 2.571 (1.40), 2.591
(0.77), 2.882 (1.10), 3.018 (1.27), 3.035 (3.01), 3.053 (2.14), 3.239 (2.40), 3.254 (2.32), 3.368 (1.13), 3.379
(1.40), 3.391 (1.33), 3.403 (0.92), 3.493 (0.76), 4.463 (0.65), 4.482 (1.12), 4.500 (0.62), 7.033 (4.22), 7.048
(4.45), 7.074 (3.47), 7.077 (4.04), 7.100 (1.85), 7.103 (1.52), 7.113 (2.53), 7.117 (2.34), 7.162 (4.18), 7.175
(2.71), 7.439 (1.03), 7.481 (4.88), 7.495 (4.57), 7.526 (2.04), 7.613 (0.91), 7.952 (5.28).
Method B
1 – { 1 – [4-Chloro-4′-(4-isobutylpiperazin- 1 -yl) [biphenyl] -2-yl]piperidin-3-yl } -5-(difluoromethyl)- 1 H-pyrazole-4-carboxylic acid hydrochloride (prepared in analogy to Example 3, Enantiomer 2, 31.2 mg, 51.3 pmol) were dissolved in 17 ml of dichloromethane and 1 ml of methanol. The solution was shaken once with 1.5 ml of saturated, aqueous sodium bicarbonate solution. The phases were separated. 5 ml of dichloromethane and 3 ml of methanol were added to the organic phase. The organic phase was then dried over sodium sulfate, filtered, evaporated and purified by preparative HPLC (RP18 column, acetonitrile/water gradient, neutral without acid addition). Product fractions were combined and lyophilized. 22 mg of the target compound (74% of theory) were obtained.
LC-MS (Method 3): Rt = 1.73 min; MS (ESIpos): m/z = 572 [M+H]+
‘H-NMR (600 MHz, DMSO-d6) 5 [ppm]: 0.887 (15.60), 0.898 (16.00), 1.493 (0.64), 1.514 (0.70), 1.695 (0.89), 1.718 (0.74), 1.799 (0.48), 1.811 (0.88), 1.822 (1.12), 1.833 (0.92), 1.844 (0.48), 1.890 (0.68), 1.910
(0.74), 1.977 (0.93), 1.995 (0.62), 2.118 (3.91), 2.130 (3.66), 2.516 (5.14), 3.017 (1.09), 3.035 (2.76), 3.053
(1.94), 3.181 (5.03), 3.185 (5.02), 3.267 (1.53), 4.473 (0.55), 4.491 (0.96), 4.509 (0.54), 6.963 (3.96), 6.977
(4.06), 7.048 (3.13), 7.051 (3.31), 7.081 (1.60), 7.084 (1.26), 7.095 (2.21), 7.098 (1.89), 7.152 (3.52), 7.165
(2.42), 7.434 (4.45), 7.448 (4.50), 7.533 (1.51), 7.621 (0.67), 7.930 (4.14).
Example 3
1 – { 1 – [4-Chloro-4′-(4-isobutylpiperazin- 1 -yl) [biphenyl] -2-yl]piperidin-3-yl } -5-(difluoromethyl)- 1 H-pyrazole-4-carboxylic acid hydrochloride (Enantiomer 2)
SUBSTITUTE SHEET (RULE 26)

Method A
A suspension of 1 – [ 1 – { 4-chloro-4′- [4-(2-methylpropyl)piperazin- 1 -yl] [1,1 ’-biphenyl] -2-yl }piperidin-3-yl] -5-(difluoromethyl)-lH-pyrazole-4-carboxylic acid (prepared in analogy to Example 2, Enantiomer 2, 43.5 g, 76.0 mmol) in diethyl ether (870 ml) was treated with a solution of hydrogen chloride in diethyl ether (84 ml, 1.0 M, 84 mmol). The resulting mixture was stirred overnight at room temperature and evaporated affording 46.1 g (quant.) of the title compound.
LC-MS (Method 3): Rt = 1.72 min; MS (ESIpos): m/z = 572 [M+H]+
‘H-NMR (600 MHz, DMSO-d6) 5 [ppm]: 1.026 (15.64), 1.037 (16.00), 1.497 (0.56), 1.519 (0.61), 1.722 (0.78), 1.743 (0.65), 1.903 (0.59), 1.910 (0.53), 1.924 (0.66), 1.930 (0.61), 1.978 (0.82), 1.994 (0.50), 2.142
(0.45), 2.154 (0.91), 2.165 (1.11), 2.176 (0.89), 2.187 (0.45), 2.557 (0.64), 2.577 (1.02), 2.594 (0.55), 2.992
(1.81), 3.002 (2.77), 3.012 (1.87), 3.018 (1.15), 3.036 (2.40), 3.054 (1.60), 3.133 (1.12), 3.148 (1.19), 3.168
(0.53), 3.237 (0.88), 3.250 (0.76), 3.338 (0.81), 3.360 (1.42), 3.379 (0.88), 3.580 (1.61), 3.791 (0.89), 3.819
(1.25), 3.844 (0.81), 4.463 (0.89), 4.474 (0.97), 4.481 (1.26), 4.488 (0.99), 4.499 (0.88), 7.051 (3.56), 7.065
(3.77), 7.077 (2.72), 7.080 (3.14), 7.103 (1.42), 7.106 (1.13), 7.116 (2.00), 7.120 (1.84), 7.165 (3.40), 7.178
(2.22), 7.443 (0.84), 7.489 (4.04), 7.504 (3.79), 7.531 (1.66), 7.618 (0.72), 7.954 (4.33), 10.519 (0.49).
Method B
Ethyl 1 – [ 1 – { 5-chloro-2- [(trifluoromethanesulfonyl)oxy]phenyl }piperidin-3-yl] -5-(difluoromethyl)- 1 H-pyrazole-4-carboxylate (prepared in analogy to Example 14A, Enantiomer 2, 80.0 mg, 150 pmol) and l-(2-methylpropyl)-4- [4-(4,4,5 ,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)phenyl]piperazine (Example 18 A 64.1 mg, 97 % purity, 180 pmol) were dissolved under argon in toluene/ethanol (0.83/0.83 ml). Tetrakis(triphenylphosphine)palladium(0) (8.69 mg, 7.52 pmol) and 2 M sodium carbonate solution (226 pl, 452 pmol) were added and the mixture was stirred at 100°C overnight. The reaction mixture was diluted with ethyl acetate and water. The aqueous phase was acidified with 1 M hydrochloric acid. The phases were
SUBSTITUTE SHEET (RULE 26)
separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic phases were dried over sodium sulfate, filtered and evaporated. The crude product was dissolved in THF/ethanol (3.9/0.39 ml), 1 M aqueous lithium hydroxide solution (1.5 ml, 1.5 mmol) was added and the mixture was stirred overnight at room temperature. The mixture was evaporated, the residue was dissolved in acetonitrile/TFA/water and purified using preparative HPLC (RP18 column, acetonitrile/water gradient with the addition of 0.1% TFA). The product fractions were combined and evaporated. The residue was mixed with 0.1 M hydrochloric acid in dioxane, carefully evaporated at 30°C (twice) and then lyophilized. 53 mg of the target compound (55% of theory, purity 95%) were obtained.
LC-MS (Method 4): Rt = 0.91 min; MS (ESIpos): m/z = 572 [M-HC1+H]+
‘H-NMR (400 MHz, DMSO-d6) 5 [ppm]: 1.004 (15.46), 1.020 (16.00), 1.491 (0.44), 1.522 (0.50), 1.722 (0.68), 1.753 (0.55), 1.890 (0.47), 1.920 (0.55), 1.967 (0.84), 2.129 (0.76), 2.146 (0.96), 2.163 (0.76), 2.582
(0.91), 2.613 (0.48), 2.999 (0.86), 3.010 (1.71), 3.025 (3.88), 3.041 (2.30), 3.131 (0.88), 3.161 (1.25), 3.177
(2.08), 3.213 (1.75), 3.242 (1.16), 3.467 (1.06), 3.496 (0.84), 3.503 (0.60), 3.519 (0.54), 3.525 (0.50), 3.549
(0.75), 3.555 (0.84), 3.572 (1.57), 3.582 (1.48), 3.589 (1.38), 3.601 (2.78), 3.608 (1.89), 3.633 (0.44), 3.640
(0.41), 3.811 (0.94), 3.847 (1.32), 3.878 (0.71), 4.329 (0.49), 4.439 (0.46), 4.466 (0.73), 4.477 (0.52), 4.839
(0.49), 7.047 (3.30), 7.070 (3.64), 7.082 (2.61), 7.087 (3.29), 7.104 (1.46), 7.109 (0.86), 7.124 (2.34), 7.129
(2.03), 7.160 (3.99), 7.181 (1.96), 7.388 (0.88), 7.490 (4.02), 7.512 (3.81), 7.519 (2.20), 7.650 (0.72), 7.959
(3.78), 9.708 (0.41).
[OC]D20 = -73.05°, c = 0.465g/100cm3, trichloromethane.
Enantiomer 2 has an absolute configuration of R as shown in example 3 A below.
1 – { 3(2?)- 1 – [4-Chloro-4′-(4-isobutylpiperazin- 1 -yl) [biphenyl] -2-yl]piperidin-3-yl } -5-(difluoromethyl)- 1H-pyrazole-4-carboxylic acid hydrochloride
Example 3A
1 – { 3(7?)- 1 – [4-Chloro-4′-(4-isobutylpiperazin- 1 -yl) [biphenyl] -2-yl]piperidin-3-yl } -5-(difluoromethyl)- 1H-pyrazole-4-carboxylic acid hydrochloride hemihydrate
SUBSTITUTE SHEET (RULE 26)

100 mg 1 – { 1 – [4-Chloro-4′-(4-isobutylpiperazin- 1 -yl) [biphenyl] -2-yl]piperidin-3-yl } -5-(difluoromethyl)-lH-pyrazole-4-carboxylic acid hydrochloride (Enantiomer 2) (example 3) were solved at 60°C in 3,5 ml 2 -propanol, wherein the 2-propanol was dosed portion wise in lOOpl -portions at 60°C until a clear solution was obtained. Afterwards the vessel was closed with a septum and placed into a slowly cooling sand bath from 60°C to roomtemperature over the weekend -> small amounts of solids were detected. Thereafter the septum was provided with a canula, in order to slowly let the solvent evaporate. After 4 weeks crystals were collected and inspected under a microscope.



AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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

……
/////////Nurandociguat, guanylate cyclase activator, BAY 3283142, LPU8429UK5
Muvadenant


Muvadenant
CAS 2459881-03-7
MF C21H26N4O4S , 430.5 g/mol
(5S)-N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy[1,3]thiazolo[4,5-c]pyridin-2-yl]-7-oxa-2-azaspiro[4.5] decane-2-carboxamide
(5S)-N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1,3]thiazolo[4,5-c]pyridin-2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide
adenosine receptor antagonist, antineoplastic, 6LSF69F6A8, M1069 , M 1069
Muvadenant is a small molecule drug. The usage of the INN stem ‘-adenant’ in the name indicates that Muvadenant is a adenosin receptor antagonist. Muvadenant has a monoisotopic molecular weight of 430.17 Da.
Adenosine is an ubiguitous modulator of numerous physiological activities, particularly within the cardiovascular, nervous and immune systems. Adenosine is related both structurally and metabolically to the bioactive nucleotides adenosine triphosphate (ATP), adenosine diphosphate (ADP), adenosine monophosphate (AMP) and cyclic adenosine monophosphate (cAMP), to the biochemical methylating agent S-adenosyl-L-methione (SAM) and structurally to the coenzymes NAD, FAD and coenzym A and to RNA.
Via cell surface receptors, adenosine modulates diverse physiological functions including induction of sedation, vasodilatation, suppression of cardiac rate and contractility, inhibition of platelet aggregability, stimulation of gluconeogenesis and inhibition of lipolysis. Studies show that adenosine is able to activate adenylate cyclases, open potassium channels, reduce flux through calcium channels, and inhibit or stimulate phosphoinositide turnover through receptor-mediated
mechanisms (Muller C. E. and Stein B., Current Pharmaceutical Design, 2: 501 , 1996; Muller C. E., Exp. Opin. Ther. Patents, 7(5): 419, 1997).
Adenosine receptors belong to the superfamily of G-protein-coupled receptors (GPCRs). Four major subtypes of adenosine receptors have been
pharmacologically, structurally and functionally characterized (Fredholm et al., Pharm. Rev., 46: 143-156, 1994) and referred to as A1, A2A, A2B and A3. Though the same adenosine receptor can couple to different G-proteins, adenosine A1 and A3 receptors usually couple to inhibitory G-proteins referred to as G, and Go which inhibit adenylate cyclase and down-regulate cellular cAMP levels. In contrast, the adenosine A2A and A2B receptors couple to stimulatory G-proteins referred to as Gs that activate adenylate cyclase and increase intracellular levels of cAMP (Linden J., Annu. Rev. Pharmacol. Toxicol., 41 : 775-87 2001).
PAT
- Thiazolopyridine derivatives as adenosine receptor antagonistsPublication Number: US-2022119412-A1Priority Date: 2019-01-22
- Thiazolopyridine derivatives as adenosine receptor antagonistsPublication Number: EP-3914600-B1Priority Date: 2019-01-22Grant Date: 2024-08-07
- Thiazolopyridine derivatives as adenosine receptor antagonistsPublication Number: CN-113939520-BPriority Date: 2019-01-22Grant Date: 2024-11-22
- Thiazolopyridine derivatives as adenosine receptor antagonistsPublication Number: ES-2992081-T3Priority Date: 2019-01-22Grant Date: 2024-12-09
- Thiazolopyridine derivatives as adenosine receptor antagonists.Publication Number: JP-7600119-B2Priority Date: 2019-01-22Grant Date: 2024-12-16
- Thiazolopyridine derivatives as adenosine receptor antagonistsPublication Number: AU-2020211697-A1Priority Date: 2019-01-22
- Thiazolopyridine derivatives as adenosine receptor antagonistsPublication Number: KR-20210116572-APriority Date: 2019-01-22
- Thiazolopyridine derivatives as adenosine receptor antagonistsPublication Number: CN-113939520-APriority Date: 2019-01-22
- Thiazolopyridine derivatives as adenosine receptor antagonistsPublication Number: EP-3914600-A1Priority Date: 2019-01-22
- Thiazolopyridine derivative as an adenosine receptor antagonistPublication Number: JP-2022524914-APriority Date: 2019-01-22
- Combination therapy for cancerPublication Number: CN-117858723-APriority Date: 2021-06-07
- Combination treatment of cancerPublication Number: EP-4351640-A1Priority Date: 2021-06-07
- Combination Treatment of CancerPublication Number: JP-2024520764-APriority Date: 2021-06-07
- Combination Treatment of CancerPublication Number: US-2024279338-A1Priority Date: 2021-06-07
- Thiazolopyridine derivatives as adenosine receptor antagonistsPublication Number: WO-2020152132-A1Priority Date: 2019-01-22
- Novel crystalline forms of (s)-7-oxa-2-aza-spiro[4.5]decane-2-carboxylic acid [7-(3,6-dihydro-2h-pyran-4-yl)-4-methoxy-thiazolo[4,5-c]pyridin-2-yl]-amide and co-crystal forms thereofPublication Number: TW-202415667-APriority Date: 2022-08-02
- Novel crystalline forms of (s)-7-oxa-2-aza-spiro[4.5]decane-2-carboxylic acid [7-(3,6-dihydro-2h-pyran-4-yl)-4-methoxy-thiazolo[4,5-c]pyridin-2-yl]-amide and co-crystal forms thereofPublication Number: WO-2024028273-A1Priority Date: 2022-08-02
- Combination treatment of cancerPublication Number: WO-2022258622-A1Priority Date: 2021-06-07
- Combination treatment of cancerPublication Number: AU-2022288571-A1Priority Date: 2021-06-07
- Combination treatment of cancerPublication Number: CA-3220380-A1Priority Date: 2021-06-07
PAT
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2020152132&_cid=P10-MHPOEV-06540-1



1. (5R)-N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-thiazolo[4,5-c]pyridin- 2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide 24
and (5S)-N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-thiazolo[4,5- c]pyridin-2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide 25

a. 4-chloro-5-iodo-3-nitropyridin-2-ol
Into a 250-mL round-bottom flask was placed 4-chloro-3-nitropyridin-2-ol (10.0 g, 54.4 mmol, 95%), N-lod-succinimid (NIS, 14.2 g, 59.9 mmol, 95%) in acetonitrile (115 mL). The solution was stirred for 1 h overnight at 80°C in an oil bath. The mixture was concentrated and the precipitate formed collected by filtration. The residue was washed with twice with petrol ether (500 mL) dried under vacuum at 60°C overnight. This resulted in 4-chloro-5-iodo-3-nitropyridin-2-ol (16.5 g, 97.9%, 97% purity) as a yellow solid. MS: m/z = 300.9 [M+H]+.
b. 4-chloro-5-iodo-2-methoxy-3-nitropyridine
Into a 500-mL round-bottom flask was placed 4-chloro-5-iodo-3-nitropyridin-2-ol (16.5 g, 53.3 mmol, 97%), Ag2CO3 (15.5 g, 53.3 mmol, 95%) in toluene (310 mL). To this suspension CH3I (15.9 g, 107 mmol, 95%) was added at 50°C and the mixture was stirred at 80°C for 4 h. The precipitate was collected by filtration and discarded. The filtrate was evaporated to dryness under vacuum and the residue purified by silica gel chromatography with ethyl acetate/petroleum ether (15:85).
This resulted in 4-chloro-5-iodo-2-methoxy-3-nitropyridine (9.90 g, 52.6%, 89% purity) as a light yellow solid. MS: m/z = 315.5 [M+H]+.
c. 4-chloro-5-iodo-2-methoxypyridin-3-amine
Into a 250-mL 3-necked round-bottom flask was placed 4-chloro-5-iodo-2-methoxy-3-nitropyridine (9.90 g, 28.0 mmol, 89%), iron (16.5 g, 281 mmol, 95%) and NH 4C (9.40 g, 174 mmol, 99%) in ethanol (152 mL) and water (30 mL). The mixture was stirred for 2 h at 80°C in an oil bath. The reaction mixture was filtered over Celite, washed with ethanol and the mother liquor was concentrated to dryness. The residue was stirred for 30 min. with 100 ml water at 60°dried in vacuo. This resulted in 4-chloro-5-iodo-2-methoxypyridin-3-amine (7.20 g, 75%, 83% purity) as an off-white solid. It was used without further purification in the next step. MS: m/z = 285.9 [M+H]+.
d. N-[7-iodo-4-methoxy-[1,3]thiazolo[4,5-c]pyridin-2-yl]benzamide
Into a 500-mL round-bottom flask was placed 4-chloro-5-iodo-2-methoxypyridin-3-amine (7.20 g, 21.0 mmol, 83%) in acetone (150 mL) and benzoyl isothiocyanate (5.21 g, 31.5 mmol, 99%) was added dropwise at room temperature. The solution was stirred for 1 h at 50 °C in an oil bath. The solids were collected by filtration, washed with acetone and dried in vacuo to give N-[7-iodo-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]benzamide (8.73 g, 91 %, 90% purity) as a white solid. MS: m/z = 412.2 [M+H]+.
e. N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1,3]thiazolo[4,5-c]pyridin- 2-yl]benzamide
To a solution of N-[7-iodo-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]benzamide (6.00 g, 13.1 mmol, 90%) and 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (6.13 g, 27.7 mmol, 95%) in dioxane (200 mL) and water (40.00 mL) were added NaOH (2.90 g, 68.9 mmol, 95%) and Pd(dppf)Cl2* dichloromethane (1.20 g, 1.40 mmol, 95%). After stirring for 1 h at 100°C under a nitrogen atmosphere, the mixture was concentrated to dryness under vacuo. The residue was purified by silica gel chromatography with ethyl acetate/hexane (95:5). This resulted in 3.32 g (62%, 90% purity) of N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]benzamide as colorless solid. MS: m/z = 368.1 [M+H]+.
f. 7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1,3]thiazolo[4,5-c]pyridin-2- amine
To a stirred mixture of N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]benzamide (3.27 g, 8.00 mmol, 90%) in water/methanol (1 :1 , 300 mL) was added NaOH (3.36 g, 80.0 mmol, 95%) at room temperature under nitrogen atmosphere. The mixture was stirred for overnight at 90°C under nitrogen atmosphere and evaporated to dryness. The residue was taken up in water and extracted 3 times with dichloromethane (100 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and evaporated to dryness. The residue was purified by silica gel column chromatography, eluted with petrol ether/ethyl acetate (1 :1) to afford 7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-amine (1.50 g, 68%, 96% purity) as a light brownish solid. MS: m/z = 264.1 [M+H]+.
g. phenyl N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy- [1,3]thiazolo[4,5c]pyridin-2-yl]-N-(phenoxycarbonyl)carbamate
To a stirred solution of 7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-amine (600 mg, 2.19 mmol, 96%) and phenyl chloroformate (1.81 g,
11.0 mmol, 95%) in THF (50 mL) was added K2CO3 (1.59 g, 11.0 mmol, 95%) and pyridine (913 mg, 11.0 mmol, 95%) at room temperature under nitrogen
atmosphere. The mixture was stirred for 6 h at 50° and then after re-cooling to room temperature quenched by the addition of water (300 mL). The mixture was extracted 3 times with dichloromethane (200 mL), the combined organic layers were washed once with brine (200 mL), dried over anhydrous Na2SO4, filtered, and evaporated to dryness under reduced pressure. This resulted in phenyl N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]-N-(phenoxycarbonyl)carbamate (1.00 g, 69%, 76% purity) as a light yellow solid. The crude product was used in the next step directly without further purification. MS: m/z = 504.1 [M+H]+.
h. N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1,3]thiazolo[4,5-c]pyridin- 2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide
To a mixture of phenyl N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]-N-(phenoxycarbonyl)carbamate (1.00 g, 1.52 mmol, 76.) and bis(7-oxa-2-azaspiro[4.5]decane), oxalic acid (1.19 g, 3.03 mmol, 95%) in THF (50 mL) was added diisopropylethyl amine (1.24 g, 9.09 mmol, 95%) at room temperature under nitrogen atmosphere. The mixture was stirred for 1 h at 60°. After re-cooling to room temperature, the mixture was extracted twice with dichloromethane (100 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and evaporated to dryness. The residue was purified by silica gel column chromatography, eluted with petrol ether/ethyl acetate (1 :1) to afford N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide (600 mg, 92%) as a white solid. HPLC: 99.9 % purity, RT = 1.17 min. MS: m/z = 431.1 [M+H]+. 1 H NMR (300 MHz, DMSO-d6) d 1 1.37 (s, 1 H), 7.95 (s, 1 H), 6.25 (s, 1 H), 4.30-4.29 (m, 2H), 3.99 (s, 3H), 3.89 (t, J=5.4Hz, 2H), 3.61-3.29 (m, 8H), 2.55-2.51 (m, 2H), 1.82-1.54 (m, 6H).
i. (5R)-N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-thiazolo[4,5-c]pyridin- 2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide 24
and (5S)-N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-thiazolo[4,5- c]pyridin-2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide 25
N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide (450 mg, 1.044 mmol, 1 equiv, 99.9%) was purified by chiral-preparative HPLC (Preparative HPLC-032, column: ChiralPak IA, 2*25cm, 5 mm; mobile phase, dichloromethane:ethanol (20:80); detector, UV). This resulted in (5R)-N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide (178 mg, 39%) as a white solid. HPLC: 99.7 % purity, RT (chiral) = 3.86 min, 100% ee. MS: m/z = 431.2 [M+H]+. 1 H NMR (400 MHz, DMSO-d6) d 1 1.36 (s, 1 H), 7.94 (s, 1 H), 6.24 (s, 1 H), 4.29-4.27 (m, 2H), 3.97 (s,3H), 3.88 (t, J=5.2 Hz, 2H), 3.51-3.19 (m, 8H), 2.55-2.50 (m, 2H), 1.83-1.53 (m, 6H) and (5S)-N-[7-(3,6-dihydro-2H-pyran-4-yl)-4-methoxy-[1 ,3]thiazolo[4,5-c]pyridin-2-yl]-7-oxa-2-azaspiro[4.5]decane-2-carboxamide (171 mg, 38%) as a white solid. HPLC: 99.8 % purity, RT (chiral) = 5.23 min, 99.9% ee. MS: m/z = 431.2 [M+H]+. 1 H NMR (400 MHz, DMSO-d6) d 1 1.35 (s, 1 H), 7.94 (s, 1 H), 6.24 (s, 1 H), 4.29-4.28 (m, 2H), 3.99 (s, 3H), 3.88-3.85 (m, 2H), 3.61-3.29 (m, 8H), 2.55-2.50 (m,2H), 1.83-1.53 (m,6H).
PAT
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2024028273&_cid=P10-MHPOFP-06905-1



AS ON OCT2025 4.511 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

join me on Linkedin
Anthony Melvin Crasto Ph.D – India | LinkedIn
join me on Researchgate
RESEARCHGATE

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

……
//////////muvadenant, adenosine receptor antagonist, antineoplastic, 6LSF69F6A8, M1069 , M 1069
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO
.....










