Home » PHASE 3
Category Archives: PHASE 3
Atumelnant




Atumelnant
CAS 2392970-97-5
MF C33H42F3N5O3 MW 613.7 g/mol
CRN04894, NR57FH6U1N
CRINETICS PHARMA, Orphan Drug Status, Congenital adrenal hyperplasia
N-[(3S)-1-azabicyclo[2.2.2]octan-3-yl]-6-(2-ethoxyphenyl)-3-[(2R)-2-ethyl-4-[1-(trifluoromethyl)cyclobutanecarbonyl]piperazin-1-yl]pyridine-2-carboxamide
N-[(3S)-1-azabicyclo[2.2.2]octan-3-yl]-6-(2-ethoxyphenyl)-3-{(2R)-2-ethyl-4-[1-(trifluoromethyl) cyclobutane-1-carbonyl]piperazin-1-yl}pyridine-2-carboxamide
Adrenocorticotropic hormone receptor antagonist
- OriginatorCrinetics Pharmaceuticals
- ClassAmides; Antineoplastics; Antisecretories; Benzene derivatives; Cyclobutanes; Ethers; Fluorocarbons; Ketones; Piperazines; Pyridines; Quinuclidines; Small molecules
- Mechanism of ActionMelanocortin type 2 receptor antagonists
- Orphan Drug StatusYes – Congenital adrenal hyperplasia
- Phase IICongenital adrenal hyperplasia; Cushing syndrome
- No development reportedEctopic ACTH syndrome
- 21 Aug 2025Atumelnant receives Orphan Drug status for Congenital adrenal hyperplasia in the US
- 07 Aug 2025Crinetics pharmaceuticals plans phase II/III clinical trial for Cushing’s disease in 1H 2026
- 08 May 2025Crinetics Pharmaceuticals plans the phase III CALM-CAH trial for Congenital adrenal hyperplasia (In adults) (PO), in the second half of 2025
Atumelnant (INNTooltip International Nonproprietary Name; developmental code name CRN04894) is an investigational new drug developed by Crinetics Pharmaceuticals for the treatment of adrenocorticotropic hormone (ACTH)-dependent endocrine disorders.[1] It is a selective antagonist of the melanocortin type 2 receptor (MC2R), also known as the ACTH receptor, which is primarily expressed in the adrenal glands.[1][2] The drug is orally active.[1] Atumelnant is being evaluated to treat conditions such as congenital adrenal hyperplasia (CAH) and ACTH-dependent Cushing’s syndrome caused for example by pituitary adenomas.[3]
Atumelnant is an orally bioavailable nonpeptide antagonist of the adrenocorticotropic hormone (ACTH) receptor (ACTHR; melanocortin receptor 2; MC2R), with potential steroid hormone production inhibitory activity. Upon oral administration, atumelnant competes with ACTH for receptor binding to MC2R in the adrenal cortex and inhibits ACTH signaling. This may inhibit the synthesis and secretion of steroid hormones. MC2R, a member of the melanocortin receptor subfamily of type 1 G protein-coupled receptors, plays a key role in adrenal steroidogenesis.
PAPER
Discovery of CRN04894: A Novel Potent Selective MC2R Antagonist
Publication Name: ACS Medicinal Chemistry Letters
Publication Date: 2024-03-19, PMCID: PMC11017392, PMID: 38628803
DOI: 10.1021/acsmedchemlett.3c00514
PATENTS
- Melanocortin subtype-2 receptor antagonists and uses thereofPublication Number: IL-279152-B2Priority Date: 2018-06-05
- Melanocortin subtype-2 receptor (mc2r) antagonists and uses thereofPublication Number: US-2024300920-A1Priority Date: 2018-06-05
- Melanocortin subtype-2 receptor antagonists and uses thereofPublication Number: IL-279152-B1Priority Date: 2018-06-05
- Melanocortin subtype-2 receptor (mc2r) antagonists and uses thereofPublication Number: JP-2024009837-APriority Date: 2018-06-05
- Melanocortin subtype-2 receptor (MC2R) antagonists and uses thereofPublication Number: KR-102695210-B1Priority Date: 2018-06-05Grant Date: 2024-08-13
- Melanocortin subtype-2 receptor (mc2r) antagonists and uses thereofPublication Number: US-2024109866-A1Priority Date: 2018-06-05
- Melanocortin subtype-2 receptor (MC2R) antagonists and uses thereofPublication Number: CN-112533904-BPriority Date: 2018-06-05Grant Date: 2024-10-29
- Melanocortin subtype-2 receptor (MC2R) antagonists and uses thereofPublication Number: US-10981894-B2Priority Date: 2018-06-05Grant Date: 2021-04-20
- Melanocortin subtype-2 receptor (mc2r) antagonists and uses thereofPublication Number: US-2021002254-A1Priority Date: 2018-06-05
- Melanocortin subtype-2 receptor (mc2r) antagonists and uses thereofPublication Number: US-2021238164-A1Priority Date: 2018-06-05
- Melanocortin subtype-2 receptor (MC2R) antagonists and uses thereofPublication Number: US-11566015-B2Priority Date: 2018-06-05Grant Date: 2023-01-31
- Melanocortin subtype-2 receptor (MC2R) antagonists and their usesPublication Number: JP-7359783-B2Priority Date: 2018-06-05Grant Date: 2023-10-11
- Melanocortin subtype-2 receptor (mc2r) antagonists and uses thereofPublication Number: US-2020216415-A1Priority Date: 2018-06-05
- Melanocortin subtype-2 receptor (MC2R) antagonists and uses thereofPublication Number: US-10766877-B2Priority Date: 2018-06-05Grant Date: 2020-09-08
- Melanocortin subtype-2 receptor (MC2R) antagonists and uses thereofPublication Number: CN-112533904-APriority Date: 2018-06-05
- Melanocortin subtype-2 receptor (mc2r) antagonists and uses thereofPublication Number: EP-3802500-A1Priority Date: 2018-06-05
- Melanocortin subtype-2 receptor (MC2R) antagonists and uses thereofPublication Number: KR-20210005995-APriority Date: 2018-06-05
- Melanocortin subtype-2 receptor (MC2R) antagonists for the treatment of diseasePublication Number: CN-117043146-APriority Date: 2021-03-19
- Melanocortin subtype-2 receptor (MC2R) antagonists and uses thereofPublication Number: US-10562884-B2Priority Date: 2018-06-05Grant Date: 2020-02-18
- Melanocortin subtype-2 receptor (MC2R) antagonists and uses thereofPublication Number: US-10604507-B2Priority Date: 2018-06-05Grant Date: 2020-03-31
- Melanocortin subtype-2 receptor (mc2r) antagonists and uses thereofPublication Number: US-2019367481-A1Priority Date: 2018-06-05
- Melanocortin subtype-2 receptor (mc2r) antagonists and uses thereofPublication Number: US-2020010452-A1Priority Date: 2018-06-05
Melanocortin subtype-2 receptor (mc2r) antagonist for the treatment of diseasePublication Number: US-2022313691-A1Priority Date: 2021-03-19 - Melanocortin subtype-2 receptor (mc2r) antagonist for the treatment of diseasePublication Number: WO-2022197798-A1Priority Date: 2021-03-19
- Melanocortin subtype-2 receptor (mc2r) antagonist for the treatment of diseasePublication Number: TW-202302108-APriority Date: 2021-03-19
- Melanocortin subtype-2 receptor (mc2r) antagonist for the treatment of diseasePublication Number: AU-2022240609-A1Priority Date: 2021-03-19
- Melanocortin subtype-2 receptor (mc2r) antagonist for the treatment of diseasePublication Number: EP-4308553-A1Priority Date: 2021-03-19
- Melanocortin subtype-2 receptor (mc2r) antagonist for the treatment of acth-dependent cushing’s syndromePublication Number: WO-2024211343-A1Priority Date: 2023-04-05
- Crystalline melanocortin subtype-2 receptor (mc2r) antagonistPublication Number: TW-202430167-APriority Date: 2022-12-16
- Crystalline melanocortin subtype-2 receptor (mc2r) antagonistPublication Number: US-2024208963-A1Priority Date: 2022-12-16
- Crystalline melanocortin subtype-2 receptor (mc2r) antagonistPublication Number: WO-2024130091-A1Priority Date: 2022-12-16
- Treatment of congenital adrenal hyperplasia and polycystic ovary syndromePublication Number: WO-2023163945-A1Priority Date: 2022-02-2
SYN
PATENT
https://patentscope.wipo.int/search/en/detail.jsf?docId=US278278493&_cid=P22-MFXDN2-76849-1






Example 31: N-[(3S)-1-azabicyclo[2.2.2]octan-3-yl]-6-(2-ethoxyphenyl)-3-[(2R)-2-ethyl-4-[1-(trifluoromethyl)cyclobutanecarbonyl]piperazin-1-yl]pyridine-2-carboxamide (Compound 1-410)

Step 31-1, Preparation of 6-(2-ethoxyphenyl)-3-[(2R)-2-ethyl-4-[1-(trifluoromethyl)cyclobutanecarbonyl]piperazin-1-yl]pyridine-2-carboxylic acid
Step 31-2, Preparation of N-[(3S)-1-azabicyclo[2.2.2]octan-3-yl]-6-(2-ethoxyphenyl)-3-[(2R)-2-ethyl-4-[1-(trifluoromethyl)cyclobutanecarbonyl]piperazin-1-yl]pyridine-2-carboxamide



AS ON JUNE2025 4.45 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

……
References
- “Crinetics Pharmaceuticals”. AdisInsight. 21 January 2025. Retrieved 25 February 2025.
- “Atumelnant (CRN04894)”. crinetics.com. 14 August 2020.
- Varlamov EV, Gheorghiu ML, Fleseriu M (December 2024). “Pharmacological management of pituitary adenomas – what is new on the horizon?”. Expert Opinion on Pharmacotherapy. 26 (2): 119–125. doi:10.1080/14656566.2024.2446625. PMID 39718553.
| Clinical data | |
|---|---|
| Other names | CRN04894 |
| Routes of administration | Oral[1] |
| Drug class | Melanocortin MC2 receptor antagonist[1] |
| Identifiers | |
| IUPAC name | |
| CAS Number | 2392970-97-5 |
| PubChem CID | 146361282 |
| IUPHAR/BPS | 13339 |
| ChemSpider | 129750231 |
| UNII | NR57FH6U1N |
| KEGG | D13102 |
| Chemical and physical data | |
| Formula | C33H42F3N5O3 |
| Molar mass | 613.726 g·mol−1 |
| 3D model (JSmol) | Interactive image |
| SMILES | |
| InChI | |
////////Atumelnant, CRN04894, CRN 04894, NR57FH6U1N, CRINETICS PHARMA, Orphan Drug Status, Congenital adrenal hyperplasia, PHASE 3
Admilparant



Admilparant, (BMS-986278)
CAS 2170126-74-4
MF C22H31N5O5 MW 445.5 g/mol
(1S,3S)-3-({2-methyl-6-[1-methyl-5-({[methyl(propyl)carbamoyl]oxy}methyl)-1H-1,2,3-triazol-4-l]pyridin-3-yl}oxy)cyclohexane-1-carboxylic acid
lysophosphatidic acid receptor 1 (LPA1) antagonist
- 4UN9AOU6G8
- BMS986278
- (1S,3S)-3-((2-Methyl-6-(1-methyl-5-(((methyl(propyl)carbamoyl)oxy)methyl)-1H-1,2,3-triazol-4-yl)pyridin-3-yl)oxy)cyclohexane-1-carboxylic acid

Admilparant is an investigational new drug being developed by Bristol-Myers Squibb for the treatment of idiopathic pulmonary fibrosis (IPF) and progressive pulmonary fibrosis (PPF). It is a first-in-class lysophosphatidic acid receptor 1 (LPA1) antagonist.[1][2]
As of 2024, admilparant is in Phase III clinical trials for both IPF and PPF.[2][3]
SYN
Publication Name: Journal of Medicinal Chemistry, Publication Date: 2021-10-28, PMID: 34709814
DOI: 10.1021/acs.jmedchem.1c01256

(1S,3S)-3-((2-Methyl-6-(1-methyl-5-(((methyl(propyl)carbamoyl)-oxy)methyl)-1H-1,2,3-triazol-4-yl)pyridin-3-yl)oxy)cyclohexane-1-carboxylic Acid (33). Compound 33 was prepared using the same
synthetic sequence as 25, except that intermediate 42 was reacted with
N-methylpropan-1-amine instead of 1-cyclobutyl-N-methylmethanamine. 1H NMR (500 MHz, DMSO-d6, 100 °C) δ 11.99−11.46 (m,1H), 7.82 (d, J = 8.3 Hz, 1H), 7.43 (d, J = 8.8 Hz, 1H), 5.65 (s, 2H),
4.89−4.62 (m, 1H), 4.10 (s, 3H), 3.12 (br t, J = 7.2 Hz, 2H), 2.79 (s,3H), 2.69 (tt, J = 9.4, 4.4 Hz, 1H), 2.44 (s, 3H), 2.03 (dt, J = 13.8, 4.5Hz, 1H), 1.92−1.86 (m, 1H), 1.86−1.79 (m, 2H), 1.74−1.68 (m, 1H),
1.68−1.58 (m, 2H), 1.58−1.51 (m, 1H), 1.43 (dq, J = 14.4, 7.1 Hz,2H), 0.76 (br t, J = 7.3 Hz, 3H). 13C NMR (126 MHz, DMSO-d6, 100°C) δ 175.4, 154.7, 150.1, 147.7, 143.9, 141.4, 129.6, 120.0, 118.6, 71.8,
54.5, 49.5, 37.4, 34.4, 33.4, 31.6, 28.7, 27.2, 19.8, 19.4, 18.6, 10.1. m/z446 [M + H]+
. HPLC/UV purity: 99.9% using the following reverse phase chromatographic conditions: Agilent HPLC; Phenomenex Kinetex-C-18; 100 (L) × 4.6 mm2 (i.d.) column; 2.6 μm particle size; wavelength, 220−380 nm; flow rate, 1.0 mL/min; temperature, 35°C; injection volume, 4 μL of 0.25 mg/mL in 1:1 MeCN:H2O; mobilephase A, H2O−0.05% TFA; mobile phase B, MeCN−0.05% TFA; gradient elution, starting at 10−80% B over 10 min and ending at 95% Bafter an additional 4 min; retention time = 8.28 min. Stereoisomeric purity was >99.5% using the following chiral chromatographic conditions: UPC2 Analytical SFC, ChromegaChiral CC4; 250 (L) ×4.6 mm2 (i.d.); 5 μm column; flow rate, 3 mL/min; temperature, 40 °C;injection volume, 10 μL of 0.25 mg/mL in MeCN:MeOH (1:1);mobile phase, 30% MeOH and 70% CO2 at 120 bar retention time =6.05 min. Accurate mass, [M + H]+ at m/z = 446.2398 (−2.03 ppmfrom theoretical for C22H32N5O5). [α]20D = +28.24° (MeOH, c = 0.51).
Elem. Anal. (theoretical): C, 59.31; H, 7.01; N, 15.72. Found: C, 59.35;H, 6.78; N, 15.69. UV (MeOH) at 254 nm (ε = 17,856), 290 nm (ε =7,519), and 296 nm (ε = 8,288). Concentration: adjusted for purity,
0.05154840 g/L or 0.0001157047 mol/L. Melting point = 152−154°C. Accurate mass, [M + H]+ at m/z 466.2398 (−2.03 ppm fromtheoretical for C22H32N5O5).
synthetic sequence as 25, except that intermediate 42 was reacted with N-methylpropan-1-amine instead of 1-cyclobutyl-N-methylmethanamine

a
Reagents and conditions: (a) I2 (1.1 equiv)/KI (2.5 equiv)/NaHCO3 (3 equiv)/water (96%); (b) H2 (50 psi)/ Pd/C (cat)/Et3N (2 equiv)/EtOAc (68%); (c) CH3COCl (2.5 equiv)/iPrOH (87−95%); d) (Ph3P)2PdCl2 (5%)/ Et3N/CuI (5%)/RT (75−94%); (e) Ru(II)-(Ph3P)2(Me5Cyp)Cl (5%)/TMSCH2N3/dioxane 50 °C/15 h; (f) Bu4NF/0 °C to RT (51−65% over 2 steps; 3:1 desired:undesired regioisomer); (g) 4-nitrophenyl chloroformate/pyridine/CH2Cl2 (86%); (h) N-cyclobutyl N-methylamine/iPr2NEt/CH2Cl2 (100%); (i) B2(pin)2/KOAc/PdCl2(dppf)/THF/80 °C; (j) NaH2BO4/H2O/RT (76% over 2 steps); (k) 38; 1,1′-(azodicarbonyl)dipiperidine/Bu3P/toluene/50 °C (45%); (l)LiOH/H2O/MeOH (76%).
PAT
https://patentscope.wipo.int/search/en/detail.jsf?docId=US208146892&_cid=P20-MFS2PF-83792-1
PATENT
- Carbamoyloxymethyl triazole cyclohexyl acids as lpa antagonistsPublication Number: US-2022249443-A1Priority Date: 2016-06-21
- Carbamoyloxymethyl triazole cyclohexyl acids as LPA antagonistsPublication Number: US-RE49352-EPriority Date: 2016-06-21Grant Date: 2023-01-03
- Carbamoyloxymethyl triazole cyclohexyl acids as LPA antagonistsPublication Number: AU-2021209334-B2Priority Date: 2016-06-21Grant Date: 2023-06-01
- Carbamoyloxymethyltriazole cyclohexylates as LPA antagonistsPublication Number: JP-7312295-B2Priority Date: 2016-06-21Grant Date: 2023-07-20
- Carbamoyloxymethyl triazole cyclohexyl acids as lpa antagonistsPublication Number: US-2023390249-A1Priority Date: 2016-06-21
- Carbamoyloxymethyltriazolylcyclohexanes as LPA antagonistsPublication Number: CN-109963843-BPriority Date: 2016-06-21Grant Date: 2022-03-11
- Carbamoyloxymethyltriazole cyclohexyl acid as LPA antagonistPublication Number: CN-114601830-APriority Date: 2016-06-21
- Carbamoyloxymethyl triazole cyclohexyl acid as an LPA antagonistPublication Number: KR-102377340-B1Priority Date: 2016-06-21Grant Date: 2022-03-21
- Carbamoyloxymethyl triazole cyclohexyl acids as lpa antagonistsPublication Number: KR-20220038537-APriority Date: 2016-06-21
- Carbamoyloxymethyl triazole cyclohexyl acids as lpa antagonistsPublication Number: KR-102463621-B1Priority Date: 2016-06-21Grant Date: 2022-11-03



AS ON JUNE2025 4.45 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

……
References
- “Admilparant (BMS-986278): Idiopathic Pulmonary Fibrosis Likelihood of Approval”. Pharmaceutical Technology. 25 December 2023. Retrieved 2024-11-23.
- Corte TJ, Behr J, Cottin V, Glassberg MK, Kreuter M, Martinez FJ, et al. (October 2024). “Efficacy and Safety of Admilparant, an LPA1 Antagonist in Pulmonary Fibrosis: A Phase 2 Randomized Clinical Trial”. American Journal of Respiratory and Critical Care Medicine. 211 (2): 230–238. doi:10.1164/rccm.202405-0977OC. PMID 39393084.
- Splete H (16 September 2024). “Admilparant Affects Biomarkers in Pulmonary Fibrosis”. Medscape. Retrieved 2024-11-23.
| Clinical data | |
|---|---|
| Other names | BMS-986278 |
| Identifiers | |
| IUPAC name | |
| CAS Number | 2170126-74-4 |
| PubChem CID | 132232205 |
| DrugBank | DB18011 |
| ChemSpider | 115009679 |
| UNII | 4UN9AOU6G8 |
| KEGG | D12657 |
| ChEMBL | ChEMBL5087506 |
| Chemical and physical data | |
| Formula | C22H31N5O5 |
| Molar mass | 445.520 g·mol−1 |
| 3D model (JSmol) | Interactive image |
| SMILES | |
| InChI | |
References
- Zhou Y, Zhang Y, Zhao D, Yu X, Shen X, Zhou Y, Wang S, Qiu Y, Chen Y, Zhu F: TTD: Therapeutic Target Database describing target druggability information. Nucleic Acids Res. 2024 Jan 5;52(D1):D1465-D1477. doi: 10.1093/nar/gkad751. [Article]
/////////Admilparant, BMS 986278, PHASE 3, Bristol-Myers Squibb, idiopathic pulmonary fibrosis, 4UN9AOU6G8
Baxdrostat



Baxdrostat
- NF3P9Z8J5Y
- CIN-107
- RO6836191
- 363.5 g/mol
WeightAverage: 363.461
Monoisotopic: 363.194677057
Chemical FormulaC22H25N3O2
N-[(8R)-4-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)-5,6,7,8-tetrahydroisoquinolin-8-yl]propanamide
- (+)-(R)-N-(4-(1-Methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-5,6,7,8-tetrahydroisoquinolin-8-yl)propionamide
- N-((8R)-5,6,7,8-Tetrahydro-4-(1,2,3,4-tetrahydro-1-methyl-2-oxo-6-quinolinyl)-8-isoquinolinyl)propanamide
- N-[(8R)-4-(1-methyl-2-oxo-3,4-dihydroquinolin-6-yl)-5,6,7,8-tetrahydroisoquinolin-8-yl]propanamide
- Propanamide, N-((8R)-5,6,7,8-tetrahydro-4-(1,2,3,4-tetrahydro-1-methyl-2-oxo-6-quinolinyl)-8-isoquinolinyl)-
Baxdrostat is an investigational drug that is being evaluated for the treatment of hypertension.[1] It is an aldosterone synthase inhibitor.[2][3]
Baxdrostat is under investigation in clinical trial NCT06344104 (A Phase III Study to Investigate the Efficacy and Safety of Baxdrostat in Asian Participants With Uncontrolled Hypertension on Two or More Medications Including Participants With Resistant Hypertension).
LIT
https://patentscope.wipo.int/search/en/detail.jsf?docId=US76841362&_cid=P21-MEZ3MG-55484-1
Example 3-1
(+)-(R)—N-(4-(1-Methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-5,6,7,8-tetrahydroisoquinolin-8-yl)propionamide

In analogy to the procedures described for the preparation of intermediate A-2 [E] and for the preparation of intermediate B-1, Suzuki reaction of (+)-(R)-4-bromo-5,6,7,8-tetrahydroisoquinolin-8-amine (intermediate B-3b) with 1-methyl-6-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-3,4-dihydro-1H-quinolin-2-one (intermediate A-1) gave (R)-6-(8-amino-5,6,7,8-tetrahydroisoquinolin-4-yl)-1-methyl-3,4-dihydroquinolin-2(1H)-one and after subsequent reaction with propionyl chloride the title compound as colorless solid. MS: 364.2 (M+H +).
Pat
CN 117247371
https://patentscope.wipo.int/search/en/detail.jsf?docId=CN418385740&_cid=P12-MEZHY3-66430-1


| Example 1 |

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

| Step A |
| Compound (S)-N-(4-bromo-6,7-dihydroisoquinolin-8(5H))-tert-butylsulfonylimide (1.65 g, 5 mmol) was dissolved in 20 mL of dichloromethane, and 20 mL of trifluoroacetic acid was added. The mixture was stirred and reacted for 1 hour. The reaction solution was concentrated under reduced pressure. The residue was purified by reverse-phase preparative column chromatography to obtain compound (R)-4-bromo-5,6,7,8-tetrahydroisoquinolin-8-amine (1.07 g, 94% yield). LC/MS (ESI): m/z = 226.0 [M+H] + . |
| Step B |
| To a mixture of (R)-4-bromo-5,6,7,8-tetrahydroisoquinolin-8-amine (0.86 g, 3.8 mmol), pinacol diboron (2 g, 4 mmol), AcOK (1.10 g, 11.4 mmol) in toluene (10 mL) was added Pd(dppf)Cl 2 (0.27 g, 0.38 mmol). The mixture was degassed and stirred at 130 ° C for 3 hours. The reaction mixture was filtered and concentrated to give a residue. EtOAc (10 mL) and water (10 mL) were added to the residue. The organic phase was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 ) and eluted with 30-40% ethyl acetate in petroleum ether to afford (R)-8-amino-5,6,7,8-tetrahydroisoquinoline-4-boronic acid pinacol ester (0.68 g, 65% yield). LC/MS (ESI): m/z = 274.1 [M+H] + . |
| Step C |
| To a reaction flask, add 6-bromo-1-methyl-3,4-dihydroquinolin-2(1H)-one (0.72 g, 3.0 mmol), (R)-8-amino-5,6,7,8-tetrahydroisoquinolin-4-boronic acid pinacol ester (0.99 g, 3.6 mmol), bistriphenylphosphine palladium dichloride (210 mg, 0.3 mmol), and potassium phosphate monohydrate (204 mg, 0.9 mmol). Dissolve the mixture in dioxane and water (9:1, 30 mL). Replace the atmosphere with nitrogen three times and allow the mixture to react overnight at 90°C with stirring. Cool to room temperature, dilute the reaction solution with ethyl acetate and water, and extract with ethyl acetate. The resulting organic phase is then washed with water and saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to obtain (R)-6-(8-amino-5,6,7,8-tetrahydroisoquinolin-4-yl)-1-methyl-3,4-dihydroquinolin-2(1H)-one (0.81 g, 88% yield). LC/MS (ESI): m/z = 307.1 [M+H] + . The target compound, Baxdrostat, was then prepared using a method similar to the last step in Example 1. |
| Example 3 |

| Step A |
| 4-Bromo-6,7-dihydroisoquinolin-8(5H)-one (1.88 g, 6.9 mmol) and (S)-tert-butylsulfenamide (2.51 g, 20.7 mmol) were dissolved in 20 mL of tetrahydrofuran. Ethyl titanate (10.08 mL, 48.28 mmol) was added and the mixture was heated to 65°C with stirring for 48 hours. After cooling to room temperature, ethyl acetate and water were added and stirred for 15 minutes. The resulting solid was removed by filtration. The organic phase was separated and dried over anhydrous sodium sulfate, filtered, and evaporated to dryness under reduced pressure to obtain the crude product (S,Z)-N-(4-bromo-6,7-dihydroisoquinolin-8(5H)-tert-butylsulfenimide), which was used directly in the next step. LC/MS (ESI): m/z = 376.2 [M+H] + . |
| Step B |
| Compound (S,Z)-N-(4-iodo-6,7-dihydroisoquinoline-8(5H)-tert-butylsulfonyl imide) (2.26 g, 6 mmol) was dissolved in 15 mL of tetrahydrofuran and cooled to -45°C. Sodium borohydride (0.36 g, 9.0 mmol) was added, and the mixture was allowed to return to room temperature and stirred for 18 hours. The mixture was quenched with ice water and extracted with dichloromethane. The resulting organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to obtain compound (S)-N-(4-iodo-6,7-dihydroisoquinoline-8(5H))-tert-butylsulfonyl imide (1.04 g, 46% yield). LC/MS (ESI): m/z = 378.0 [M+H] + . |
| Step C |
| To a mixture of (S)-N-(4-iodo-6,7-dihydroisoquinoline-8(5H))-tert-butylsulfonimide (0.76 g, 2 mmol), pinacol diboronate (1.05 g, 2.1 mmol), and AcOK (0.578 g, 6 mmol) in toluene (10 mL) was added Pd(dppf)Cl 2 (0.144 g, 0.2 mmol). The mixture was degassed and stirred at 130 ° C for 3 hours. The reaction mixture was filtered and concentrated to give a residue. EtOAc (15 mL) and water (10 mL) were added to the residue. The organic phase was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 ) and eluted with 30-40% ethyl acetate in petroleum ether to afford (S)-N-tert-butylsulfonamido-6,7-dihydroisoquinolin-8(5H)-4-boronic acid pinacol ester (0.51 g, 68% yield). LC/MS (ESI): m/z = 378.2 [M+H] + . |
| The next three steps were carried out in the same manner as in Example 1 to prepare the target compound Baxdrostat. |
LIT
https://medicalxpress.com/news/2025-08-stubborn-high-blood-pressure-experimental.html
A new treatment has been shown to significantly lower blood pressure in people whose levels stay dangerously high, despite taking several existing medicines, according to the results of a Phase III clinical trial led by a UCL Professor. Globally, around 1.3 billion people have high blood pressure (hypertension), and in around half of cases the condition is uncontrolled or treatment resistant. These individuals face a much greater risk of heart attack, stroke, kidney disease, and early death. In the UK the number of people with hypertension is around 14 million.
The international BaxHTN trial, led by Professor Bryan Williams (UCL Institute of Cardiovascular Science), assessed the new drug baxdrostat—which is taken as a tablet—with participation from nearly 800 patients across 214 clinics worldwide.
Results were presented at the European Society of Cardiology (ESC) Congress 2025 in Madrid and were simultaneously published in the New England Journal of Medicine.
The trial results showed that, after 12 weeks, patients taking baxdrostat (1 mg or 2 mg once daily in pill form) saw their blood pressure fall by around 9-10 mmHg more than placebo—a reduction large enough to cut cardiovascular risk. About four in 10 patients reached healthy blood pressure levels, compared with fewer than two in 10 on placebo.
Principal Investigator, Professor Williams, who is presenting the results at ESC, said, “Achieving a nearly 10 mmHg reduction in systolic blood pressure with baxdrostat in the BaxHTN Phase III trial is exciting, as this level of reduction is linked to substantially lower risk of heart attack, stroke, heart failure and kidney disease.”
How baxdrostat works
Blood pressure is strongly influenced by a hormone called aldosterone, which helps the kidneys regulate salt and water balance.
Some people produce too much aldosterone, causing the body to hold onto salt and water. This aldosterone dysregulation pushes blood pressure up and makes it very difficult to control.
Addressing aldosterone dysregulation has been a key effort in research over many decades, but it has been so far difficult to achieve.
Baxdrostat works by blocking aldosterone production, directly addressing this driver of high blood pressure (hypertension).
Professor Williams, Chair of Medicine at UCL, said, “These findings are an important advance in treatment and in our understanding of the cause of difficult-to-control blood pressure.
“Around half of people treated for hypertension do not have it controlled, however this is a conservative estimate and the number is likely higher, especially as the target blood pressure we try to reach is now much lower than it was previously.
“In patients with uncontrolled or resistant hypertension, the addition of baxdrostat 1mg or 2mg once daily to background antihypertensive therapy led to clinically meaningful reductions in systolic blood pressure, which persisted for up to 32 weeks with no unanticipated safety findings.
“This suggests that aldosterone is playing an important role in causing difficult to control blood pressure in millions of patients and offers hope for more effective treatment in the future.”
Historically, higher-income Western countries were reported to have far higher levels of hypertension. However, largely due to changing diets (adding less salt to food), the numbers of people living with the condition is now far higher in Eastern and lower-income countries. More than half of those affected live in Asia, including 226 million people in China and 199 million in India.
Professor Williams added, “The results suggest that this drug could potentially help up to half a billion people globally—and as many as 10 million people in the UK alone, especially at the new target level for optimal blood pressure control.”



AS ON JUNE2025 4.45 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

……
| Identifiers | |
|---|---|
| IUPAC name | |
| CAS Number | 1428652-17-8 |
| PubChem CID | 71535962 |
| IUPHAR/BPS | 12362 |
| ChemSpider | 76804781 |
| UNII | NF3P9Z8J5Y |
| KEGG | D12789 |
| ChEMBL | ChEMBL4113975 |
| Chemical and physical data | |
| Formula | C22H25N3O2 |
| Molar mass | 363.461 g·mol−1 |
| 3D model (JSmol) | Interactive image |
| SMILES | |
| InChI | |
PATENTS
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: TW-201319054-APriority Date: 2011-09-23
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: TW-I576342-BPriority Date: 2011-09-23Grant Date: 2017-04-01
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: US-2013079365-A1Priority Date: 2011-09-23
- Bicyclic dihydroquinoline-2-one derivativesPublication Number: US-9353081-B2Priority Date: 2011-09-23Grant Date: 2016-05-31
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: WO-2013041591-A1Priority Date: 2011-09-23
- Novel bicyclic dihydroquinolin-2-one derivativesPublication Number: JP-2014526539-APriority Date: 2011-09-23
- Novel bicyclic dihydroquinolin-2-one derivativesPublication Number: JP-6012737-B2Priority Date: 2011-09-23Grant Date: 2016-10-25
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: KR-101723276-B1Priority Date: 2011-09-23Grant Date: 2017-04-04
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: KR-20140076591-APriority Date: 2011-09-23
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: NZ-620652-APriority Date: 2011-09-23
- New bicyclic dihydroquinolin-2-one derivativesPublication Number: CN-103827101-APriority Date: 2011-09-23
- Bicyclic dihydroquinolin-2-one derivativesPublication Number: CN-103827101-BPriority Date: 2011-09-23Grant Date: 2016-12-07
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: EP-2758388-A1Priority Date: 2011-09-23
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: EP-2758388-B1Priority Date: 2011-09-23Grant Date: 2018-02-21
- NEW BICYCLIC DERIVATIVES OF DIHYDROKINOLIN-2-ONPublication Number: HR-P20180592-T1Priority Date: 2011-09-23
- Methods of using aldosterone synthase inhibitorsPublication Number: US-2024277698-A1Priority Date: 2021-06-24
- How to use aldosterone synthase inhibitorsPublication Number: CN-117545482-APriority Date: 2021-06-24
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: AU-2012311582-A1Priority Date: 2011-09-23
- New bicyclic dihydroquinoline-2-one derivativesPublication Number: AU-2012311582-B2Priority Date: 2011-09-23Grant Date: 2017-07-06
- Bicyclic dihydroquinoline-2-one derivativesPublication Number: CA-2845170-CPriority Date: 2011-09-23Grant Date: 2019-08-13
References
- “Baxdrostat – CinCor Pharma”. AdisInsight. Springer Nature Switzerland AG.
- Dogra S, Shah S, Gitzel L, Pusukur B, Sood A, Vyas AV, Gupta R (July 2023). “Baxdrostat: A Novel Aldosterone Synthase Inhibitor for Treatment Resistant Hypertension”. Current Problems in Cardiology. 48 (11): 101918. doi:10.1016/j.cpcardiol.2023.101918. PMID 37399857. S2CID 259320969.
- Awosika A, Cho Y, Bose U, Omole AE, Adabanya U (October 2023). “Evaluating phase II results of Baxdrostat, an aldosterone synthase inhibitor for hypertension”. Expert Opinion on Investigational Drugs. 32 (11): 985–995. doi:10.1080/13543784.2023.2276755. PMID 37883217. S2CID 264517675.
- The selective aldosterone synthase inhibitor Baxdrostat significantly lowers blood pressure in patients with resistant hypertensionPublication Name: Frontiers in EndocrinologyPublication Date: 2022-12-09PMCID: PMC9780529PMID: 36568122DOI: 10.3389/fendo.2022.1097968
- Results from a phase 1, randomized, double-blind, multiple ascending dose study characterizing the pharmacokinetics and demonstrating the safety and selectivity of the aldosterone synthase inhibitor baxdrostat in healthy volunteersPublication Name: Hypertension research : official journal of the Japanese Society of HypertensionPublication Date: 2022-10-20PMCID: PMC9747611PMID: 36266539DOI: 10.1038/s41440-022-01070-4
- Preclinical and Early Clinical Profile of a Highly Selective and Potent Oral Inhibitor of Aldosterone Synthase (CYP11B2)Publication Name: Hypertension (Dallas, Tex. : 1979)Publication Date: 2017-01PMCID: PMC5142369PMID: 27872236DOI: 10.1161/hypertensionaha.116.07716
/////Baxdrostat, PHASE 3, NF3P9Z8J5Y, CIN 107, RO 6836191,
DAZDOTUFTIDE


DAZDOTUFTIDE
- TRS-01
- CAS 2522933-44-2
- 4-((E)-(5-(2-(2-((S)-2-((S)-1-(L-Threonyl-L-lysyl)pyrrolidine-2-carboxamido)-5-guanidinopentanamido)acetamido)-2-carboxyethyl)-2-hydroxyphenyl)diazenyl)phenyl (2-(trimethylammonio)ethyl) phosphate
- L-Tyrosine, L-threonyl-L-lysyl-L-prolyl-L-arginylglycyl-3-((1E)-2-(4-((hydroxy(2-(trimethylammonio)ethoxy)phosphinyl)oxy)phenyl)diazenyl)-, inner salt
- [4-[[5-[(2S)-2-[[2-[[(2S)-2-[[(2S)-1-[(2S)-6-amino-2-[[(2S,3R)-2-amino-3-hydroxybutanoyl]amino]hexanoyl]pyrrolidine-2-carbonyl]amino]-5-(diaminomethylideneamino)pentanoyl]amino]acetyl]amino]-2-carboxyethyl]-2-hydroxyphenyl]diazenyl]phenyl] 2-(trimethylazaniumyl)ethyl phosphate
C43H68N13O13P
1006.1 g/mol
L-Tyrosine, L-threonyl-L-lysyl-L-prolyl-L-arginylglycyl-3-[(1E)-2-[4-[[hydroxy[2-(trimethylammonio)ethoxy]phosphinyl]oxy]phenyl]diazenyl]-, inner salt
SQ
| 1 | TKPRGY |
Protein/Peptide Sequence, Sequence Length: 6
modified (modifications unspecified)
- OriginatorTarsius Pharma
- DeveloperTarsier Pharma
- ClassAnti-inflammatories; Eye disorder therapies; Small molecules
- Mechanism of ActionImmunomodulators
- Orphan Drug StatusYes – Uveitis
- Phase IIIUveitis
- Phase I/IIOcular inflammation
- PreclinicalDiabetic macular oedema; Diabetic retinopathy; Dry age-related macular degeneration
- 16 Jan 2024Tarsier Pharma receives an agreement from the US FDA under Special Protocol Assessment for Tarsier-04 phase III trial for TR S01 eye drops for Uveitis
- 13 Nov 2023Tarsier Pharma announces successful outcome of a Type C meeting with the US FDA supporting the advancement of TRS 01 eye drop for Uveitis
- 13 Nov 2023Tarsier Pharma plans a Tarsier-04 phase III registrational trial of TR S01 for Uveitis in USA

| Molecular Formula | C43H68N13O13P.C2HF3O2 |
| Molecular Weight | 1120.0764 |
TRS-01 trifluoroacetate
I35XEI0JIK
CAS 2522933-45-3
4-((E)-(5-(2-(2-((S)-2-((S)-1-(L-Threonyl-L-lysyl)pyrrolidine-2-carboxamido)-5-guanidinopentanamido)acetamido)-2-carboxyethyl)-2-hydroxyphenyl)diazenyl)phenyl (2-(trimethylammonio)ethyl) phosphate, trifluoroacetate salt
Ocular inflammation, an inflammation of any part of the eye, is one of the most common ocular diseases. Ocular inflammation refers to a wide range of inflammatory disease of the eye, one of them is uveitis. These diseases are prevalent in all age groups and may be associated with systemic diseases such as Crohn’s disease, Behcet disease, Juvenile idiopathic arthritis and others. The inflammation can also be associated with other common eye symptoms such as dry eye and dry macular degeneration. Several drugs have the known side effect of causing uveitis and/or dry eye. The most common treatment for ocular inflammation, is steroids and specifically corticosteroids. However, these treatments have several known and sometimes severe side effects.
Phosphorylcholine (PC) is a small zwitterionic molecule secreted by helminths which permits helminths to survive in the host inducing a situation of immune tolerance as well as on the surface of some bacteria and apoptotic cells. Tuftsin-PhosphorylCholine (TRS) is bi-specific small molecule with immunomodulatory activities. TRS (Thr-Lys-Pro-Arg-Gly-Tyr-PC) is an immunomodulating peptide derivative.
Currently, TRS has been synthesized by post-synthesis modification of Thr-Lys-Pro-Arg-Gly-Tyr, so as to couple the PC moiety to the phenol ring of tyrosine. However, this synthetic approach results in very low yield, thus making the synthesis of TRS ineffective and costly. New simple and efficient methods of synthesizing TRS are highly required.
SCHEME

PATENT
WO2022224259
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2022224259&_cid=P11-MAOYY3-78105-1
EXAMPLES
EXAMPLE 1
CONJUGATION OF PHOSPHORYLCHOLINE TO BOC-TYR
[0151] 1) Preparation of diazonium salt
[0152] 4-Aminophenyl (2-(trimethylammonio)ethyl) phosphate (50 mg, 0.18 mmol)) was dissolved in 1M aqueous HC1 (1 mL), cooled in an ice-water bath and sodium nitrite (12.6 mg, 0.18 mmol) was added in a single batch. The resulting solution was stirred at 0°C for 30 min.
[0153] 2) Azo coupling
[0154] A new mixture was prepared with BOC-L-tyrosine (107 mg, 0.38 mmol) in NaHC03(lM)+NaOH buffer (pH 10) (3.3 mL) + acetonitrile (1.2 mL). The mixture was cooled in an ice-water bath. The diazonium salt mixture was added drop-wise. A red solution was formed. Stirring of this was continued at 0 °C for 6 minutes. The reaction mixture was acidified with IN aqueous HC1 to pH=~3.
[0155] The obtained solution was lyophilized overnight, and subsequently purified (e.g. by preparative MPLC), to obtain the compound:
, wherein R is Boc.
EXAMPLE 2
PREPARATION OF AN EXEMPLARY COMPOUND OF THE INVENITON
Preparation of diazonium salt:
Fmoc-Tyr-PPC
(compound 10)
[0156] 4-Aminophenyl (2-(trimethylammonio)ethyl) phosphate (250 mg, 0.912 mmol)) was dissolved in 1M aqueous HC1 (5 mL), cooled in an ice-water bath and sodium nitrite (62.9 mg, 0.912 mmol) was added in a single batch. The resulting solution was stirred at 0°C for 30 min. Azo coupling, a new mixture was prepared with Fmoc-Tyr-OH (739 mg, 1.832 mmol) in saturated NaHC03 (17 mL) + acetonitrile (12.5 mL). The resulting suspension/solution was cooled in an ice-water bath. The diazonium salt mixture was added drop-wise. Stirred at 0°C. The reaction mixture slowly turned yellow. After 5.5 h LCMS showed complete conversion. The reaction mixture was acidified with IN HC1 to pH~6, the yellowish suspension turned into a clear orange solution, which was lyophilized. This afforded 2.10 g. Dissolved in a mixture of DMSO/H20/MeCN (-1:1:1) and purified in 5 runs by acidic preparative MPLC. The fractions were combined and lyophilized overnight, to obtain the desired product (compound 10).
EXAMPLE 3
SPPS SYNTHESIS OF TRS
[0157] While facing difficulties with protection of the hydroxy group of compound 10, the inventors explored a novel strategy for SPPS synthesis of TRS :
[0158] The inventors initiated the SPPS synthesis by implementing the N-protected (Fmoc) phosphorylcholine modified tyrosine (e.g. compound 10) 200 mg of compound 10 were loaded onto the CTC resin. In brief, 2-Chlorotrityl chloride resin (1.0 – 1.2 mmol/g, 200 – 400 mesh) (450 mg, 1.441 mmol) was allowed to swell in dichloromethane (12 mL) by rocking for 30 min. The solvent was removed and a solution of (S,E)-4-((5-(2-((((9//-f1uoren-9-yl)methoxy)carbonyl)amino)-2-carboxyethyl)-2-hydroxyphenyl)diazenyl)phenyl(2-(trimethylammonio)-ethyl) phosphate (200 mg, 0.290 mmol) in dichloromethane (12 mL) containing DIPEA (0.177 mL, 1.016 mmol) (substrate did not dissolve in DCM, after addition of DIPEA a solution was obtained) was added.
[0159] After 17 h the solvent was removed and the resin was washed with dichloromethane (3×10 mL, each washing step > 2 minutes). The capping solution (CH2C12:MeOH: DIPEA 9: 1:0.5) was added (10.5 mL) and the resin was rocked for 1 hour. Then the resin was washed with dichloromethane (3×10 mL) and dried in vacuo.
[0160] This resin was then split into equal portions in order to investigate a number of conditions for the subsequent chemistry in parallel, aimed at preventing the formation of the previously found tyrosine O-acylation, as witnessed by the isolation of compound 13 (see Scheme 2). The different reaction conditions were outlined in Table 1 (see below).
Scheme 2: Solid phase peptide synthesis
Table 1: exemplary coupling conditions tested
[0161] As shown in Table 1, various coupling conditions have been tested. Entries a-c resulted in the formation of a substantial amount of the byproduct (13). An improvement was obtained by using Fmoc-Gly-OSu in DMF (entry d). In this case the formation of byproduct (13) was reduced to only 3% relative to the desired compound 12. Nonetheless, neither of these methods was capable of suppressing the formation of 13 completely, therewith still posing a risk for further peptide synthesis, as this may lead to the accumulation of byproducts (compound 13).
[0162] Surprisingly, the inventors found that the byproduct (or phenolic ester byproduct, represented by compound 13 in Scheme 3) can be cleaved under standard Fmoc deprotection conditions with piperidine or with DBU in DMF, affording compound 15 cleanly, as illustrated below:
/////////DAZDOTUFTIDE, PHASE 3, TRS-01, TRS 01
CEFILAVANCIN



CEFILAVANCIN, TD-1792
CAS 722454-12-8
C87H96Cl3N16O28S2, 1984.28
F76229E21M
Vancomycin, 26-[[[3-[[(Z)-[1-(2-amino-5-chloro-4-thiazolyl)-2-[[(6R,7R)-2-carboxy-8-oxo-3-(pyridiniomethyl)-5-thia-1-azabicyclo[4.2.0]oct-2-en-7-yl]amino]-2-oxoethylidene]amino]oxy]propyl]amino]carbonyl]-26-decarboxy-
1-{[(6R,7R)-7-[(2Z)-2-(2-amino-5-chloro-1,3-thiazol-4-yl)-2-[(3-{[(1S,2R,18R,19R,22S,25R,28R,40S)-48-{[(2S,3R,4S,5S,6R)-3-{[(2S,4S,5S,6S)-4-amino-5-hydroxy-4,6-dimethyloxan-2-yl]oxy}-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy}-22-(carbamoylmethyl)-5,47-dichloro-2,18,32,35,37-pentahydroxy-19-[(2R)-4-methyl-2-(methylamino)pentanamido]-20,23,26,42,44-pentaoxo-7,13-dioxa-21,24,27,41,43-pentaazaoctacyclo[26.14.2.2^{3,6}.2^{14,17}.1^{8,12}.1^{29,33}.0^{10,25}.0^{34,39}]pentaconta-3,5,8,10,12(48),14,16,29(45),30,32,34(39),35,37,46,49-pentadecaen-40-yl]formamido}propoxy)imino]acetamido]-2-carboxylato-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-en-3-yl]methyl}pyridin-1-ium
Phase III Skin and soft tissue infections
- OriginatorGlaxoSmithKline; Theravance
- DeveloperR-Pharm; Theravance Biopharma
- ClassAcetamides; Antibacterials; Azabicyclo compounds; Beta-lactams; Cephalosporins; Peptide antibiotics; Pyridines; Thiazoles
- Mechanism of ActionCell wall inhibitors
BUILDING BLOCK
Vancomycin,

| Formula | C66H75Cl2N9O24 |
| Molar mass | 1449.27 g·mol−1 |
Cefilavancin (TD-1792) is an experimental antibiotic medication developed for the treatment of bacterial infections such as drug-resistant strains of Staphylococcus aureus. It is a prodrug which is also a codrug, injected intravenously and then cleaved inside the body to two active components, one of which is a modified form of vancomycin and the other a cephalosporin antibiotic. In clinical trials cefilavancin has shown similar efficacy with reduced side effects compared to vancomycin itself.[1][2][3][4][5][6][7][8]
- 31 Jan 2020Cefilavancin is still in phase III trials for Skin and soft tissue infection in Russia and Georgia (R-Pharm pipeline, January 2020)
- 17 Jun 2015Phase II development is ongoing the USA
- 02 Jun 2014Theravance Biopharma is formed as a spin-off of Theravance
SCHEME

SYN
WO2003031449
https://patentscope.wipo.int/search/en/WO2003031449
cheme A
REF
Li, Huijuan; ET AL, Medicine (Philadelphia, PA, United States) (2022), 101(34), e30120
References
- ^ Long DD, Aggen JB, Chinn J, Choi SK, Christensen BG, Fatheree PR, et al. (October 2008). “Exploring the positional attachment of glycopeptide/beta-lactam heterodimers”. The Journal of Antibiotics. 61 (10): 603–614. doi:10.1038/ja.2008.80. PMID 19168974.
- ^ Tyrrell KL, Citron DM, Warren YA, Goldstein EJ (April 2012). “In vitro activity of TD-1792, a multivalent glycopeptide-cephalosporin antibiotic, against 377 strains of anaerobic bacteria and 34 strains of Corynebacterium species”. Antimicrobial Agents and Chemotherapy. 56 (4): 2194–2197. doi:10.1128/AAC.06274-11. PMC 3318369. PMID 22290981.
- ^ Stryjewski ME, Potgieter PD, Li YP, Barriere SL, Churukian A, Kingsley J, et al. (November 2012). “TD-1792 versus vancomycin for treatment of complicated skin and skin structure infections”. Antimicrobial Agents and Chemotherapy. 56 (11): 5476–5483. doi:10.1128/aac.00712-12. PMC 3486540. PMID 22869571.
- ^ Douglas EJ, Laabei M (September 2023). “Staph wars: the antibiotic pipeline strikes back”. Microbiology. 169 (9). Reading, England. doi:10.1099/mic.0.001387. PMC 10569064. PMID 37656158.
- ^ Surur AS, Sun D (2021). “Macrocycle-Antibiotic Hybrids: A Path to Clinical Candidates”. Frontiers in Chemistry. 9: 659845. Bibcode:2021FrCh….9..317S. doi:10.3389/fchem.2021.659845. PMC 8120311. PMID 33996753.
- ^ Saxena D, Maitra R, Bormon R, Czekanska M, Meiers J, Titz A, et al. (December 2023). “Tackling the outer membrane: facilitating compound entry into Gram-negative bacterial pathogens”. npj Antimicrobials and Resistance. 1 (1): 17. doi:10.1038/s44259-023-00016-1. PMC 11721184. PMID 39843585.
- ^ Koh AJ, Thombare V, Hussein M, Rao GG, Li J, Velkov T (2023). “Bifunctional antibiotic hybrids: A review of clinical candidates”. Frontiers in Pharmacology. 14: 1158152. doi:10.3389/fphar.2023.1158152. PMC 10313405. PMID 37397488.
- ^ Homer JA, Johnson RM, Koelln RA, Moorhouse AD, Moses JE (2024). “Strategic re-engineering of antibiotics”. Nature Reviews Bioengineering. doi:10.1038/s44222-024-00250-w.
| Clinical data | |
|---|---|
| Other names | TD-1792 |
| Routes of administration | Intravenous |
| Identifiers | |
| showIUPAC name | |
| CAS Number | 722454-12-8 |
| PubChem CID | 76960417 |
| DrugBank | DB05735 |
| ChemSpider | 34990483 |
| UNII | F76229E21M |
| ChEMBL | ChEMBL4297645 |
| Chemical and physical data | |
| Formula | C87H95Cl3N16O28S2 |
| Molar mass | 1983.27 g·mol−1 |
| 3D model (JSmol) | Interactive image |
| showSMILES | |
| showInChI | |
////////////CEFILAVANCIN, TD-1792, TD 1792, F76229E21M, цефилаванцин, 头孢拉凡星, سيفيلافانسين , GlaxoSmithKline, Theravance, PHASE 3
BOFUTRELVIR



BOFUTRELVIR
Cas 2103278-86-8
| Molecular Weight | 452.55 |
|---|---|
| Formula | C25H32N4O4 |
UNII-T5UX5SKK2S; Mpro inhibitor 11A; 2103278-86-8; T5UX5SKK2S, DC-402234, DC402234, MPI-10
IUPAC/Chemical Name: N-[(2S)-3-cyclohexyl-1-oxo-1-[[(2S)-1-oxo-3-[(3S)-2-oxopyrrolidin-3-yl]propan-2-yl]amino]propan-2-yl]-1H-indole-2-carboxamide
N-[(2S)-3-cyclohexyl-1-oxo-1-[[(2S)-1-oxo-3-[(3S)-2-oxopyrrolidin-3-yl]propan-2-yl]amino]propan-2-yl]-1H-indole-2-carboxamide
Bofutrelvir has an additive antiviral effect when combined with Remdesivir
FB2001
Bofutrelvir (FB2001) is a SARS-CoV-2 main protease Mpro inhibitor with an IC50 value of 53 nM and an EC50 value of 0.53 μM. Bofutrelvir exhibits potent antiviral efficacy against several current SARS-CoV-2 variants with EC50 values of 0.26-0.42 μM. Bofutrelvir has an additive antiviral effect when combined with Remdesivir.
Bofutrelvir is a small molecule inhibitor of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) main protease (Mpro; 3C-like protease; 3CL protease; 3CLpro; nsp5 protease), with potential antiviral activity against SARS-CoV-2. Upon intravenous administration or inhalation into the lungs, bofutrelvir selectively targets, binds to, and inhibits the activity of SARS-CoV-2 Mpro. This inhibits the proteolytic cleavage of viral polyproteins, thereby inhibiting the formation of viral proteins including helicase, single-stranded-RNA-binding protein, RNA-dependent RNA polymerase, 20-O-ribose methyltransferase, endoribonuclease and exoribonuclease. This prevents viral transcription and replication. Bofutrelvir may have antiviral activity in the brain.
- Originator Frontier Biotechnologies
- Class Amides; Antivirals; Indoles; Pyrrolidinones; Small molecules
- Mechanism of Action Coronavirus 3C-like proteinase inhibitors
Highest Development Phases
- Phase II/III COVID 2019 infections
Most Recent Events
- 28 Apr 2024No recent reports of development identified for phase-I development in COVID-2019-infections in USA (IV, Infusion)
- 04 Jan 2023Phase-II/III clinical trials in COVID-2019 infections in China (Inhalation) (NCT05675072)
- 30 Dec 2022Frontier Biotechnologies completes a phase I trial in COVID-2019 infections in China (Inhalation) (NCT05583812)
- N-[(2S)-3-cyclohexyl-1-oxo-1-({(2S)-1-oxo-3-[(3S)-2-oxopyrrolidin-3-yl]propan-2-yl}amino)propan-2-yl]-1H-indole-2-carboxamide is a secondary carboxamide resulting from the formal condensation of the carboxy group of 1H-indole-2-carboxylic acid with the primary amino group of 3-cyclohexyl-N-{(2S)-1-oxo-3-[(3S)-2-oxopyrrolidin-3-yl]propan-2-yl}-L-alaninamide. It is an inhibitor of SARS coronavirus main proteinase and inhibits SARS-CoV-2 replication in cell culture (EC50 = 0.53 muM). It has a role as an EC 3.4.22.69 (SARS coronavirus main proteinase) inhibitor and an anticoronaviral agent. It is an indolecarboxamide, a member of pyrrolidin-2-ones, an aldehyde, a secondary carboxamide and an oligopeptide.
SCHEME

PATENTS
CN110818691
https://patentscope.wipo.int/search/en/detail.jsf?docId=CN289596961&_cid=P11-M9Z1Y3-09353-1


| Synthesis of compound 1-2: |
| Under argon protection, N-tert-butyloxycarbonyl-L-glutamic acid dimethyl ester (1-1) (6g, 21.8mmol) was dissolved in 60mL of anhydrous tetrahydrofuran, and a tetrahydrofuran solution of LiHMDS (1M in THF) (47mL, 47mmol) was slowly dripped at -78℃, and the temperature was kept stable at -78℃ during the dripping process, which lasted for about 1 hour. After the dripping was completed, it was stirred at -78℃ for 1 hour. Bromoacetonitrile (2.79g, 23.3mmol) was dissolved in 20ml of tetrahydrofuran, and then the solution was slowly dripped into the reaction system, and the dripping process lasted for 1 to 2 hours. The temperature was controlled at -78℃ and the reaction was continued for 3 hours. After the reaction was completed, NH4Cl solution was added to the reaction solution to quench the reaction, and the mixture was stirred for 10min and then warmed to room temperature. 40mL of saturated sodium chloride solution was poured in and stirred thoroughly, and the reaction system was seen to be stratified. The organic layer was separated, and the aqueous phase was extracted with ethyl acetate (EA). The organic layers were combined, dried over anhydrous sodium sulfate, concentrated, and subjected to column chromatography (Flash, PE:EA=1:5) to obtain 3.9 g of a light yellow oil 1-2 with a yield of 58%. |
| Synthesis of compound 1-3: |
| Dissolve 1-2 (1 g, 3.15 mmol) in 25 mL of anhydrous methanol, stir to 0°C in an ice bath, and then add cobalt dichloride hexahydrate (450 mg, 1.89 mmol). After 10 min, add sodium borohydride (715 mg, 18.9 mmol) in small portions. The reaction solution continues to react in an ice bath for 1 h and then returns to room temperature. After 15 h, quench with 5 mL of saturated NH4Cl solution and continue stirring for 10 min. After filtering out the solid, evaporate the filtrate to dryness, extract with 20 mL of water and 30×3 mL of ethyl acetate, combine the organic phases, and add anhydrous Na 2 SO 4 After drying for 1 h, the residue was concentrated under reduced pressure and separated by column chromatography [PE:EA=1:2] to obtain 460 mg of a white powdery solid with a yield of 51%. |
| Synthesis of compound 1-4: |
| Compound 1-3 (2.6 g) was dissolved in a dichloromethane solution of trifluoroacetic acid (1/1, v/v), stirred at room temperature for 1 hour, concentrated, added with 100 ml of dichloromethane, washed with saturated sodium carbonate solution, and the organic layer was dried over anhydrous sodium sulfate and concentrated to obtain an oily substance 1-4 (2.7 g) with a yield of 99%. |
| Synthesis of compound 1-5: |
| Boc-cyclohexylalanine (1.26 g, 5 mmol), EDCI (1.36 g, 6 mmol), and HOBt (0.822 g, 6 mmol) were added to 80 ml of dichloromethane solution and stirred at room temperature for 30 min. Compound 1-4 (0.896 g, 5 mmol) was then added, and 1.2 equivalents of triethylamine were added dropwise, and stirred at room temperature. After TLC monitoring (ultraviolet), dichloromethane was used for extraction after the reaction was complete, and the mixture was washed with dilute hydrochloric acid, saturated sodium bicarbonate solution, and saturated sodium chloride. The organic layers were combined and dried over anhydrous sodium sulfate, and concentrated to obtain 1.2 g of a white viscous solid with a yield of 60%. |
| Synthesis of compound 1-6: |
| Compound 1-5 (2.5 g) was dissolved in a dichloromethane solution of trifluoroacetic acid (1/1, v/v), stirred at room temperature for 1 hour, concentrated, added with 100 ml of dichloromethane, washed with saturated sodium carbonate solution, and the organic layer was dried over anhydrous sodium sulfate and concentrated to obtain an oily substance 1-6 (2.61 g) with a yield of 99%. |
| Synthesis of compound 1-7: |
| Indole 2-carboxylic acid (0.795 g, 5 mmol), EDCI (1.36 g, 6 mmol), and HOBt (0.822 g, 6 mmol) were added to 80 ml of dichloromethane solution and stirred at room temperature for 30 min. Compound 1-6 (2.2 g, 5 mmol) was then added, and 1.2 equivalents of triethylamine were added dropwise, and stirred at room temperature. After TLC monitoring (ultraviolet), dichloromethane was used for extraction after the reaction was complete, and the mixture was washed with dilute hydrochloric acid, saturated sodium bicarbonate solution, and saturated sodium chloride. The organic layers were combined and dried over anhydrous sodium sulfate, and concentrated to obtain 1.3 g of a white viscous solid with a yield of 60%. |
| Synthesis of compound 1-8: |
| Dissolve 1-7 (243 mg, 0.51 mmol) in 20 ml of methanol, slowly add sodium borohydride (107 mg, 2.9 mmol) in batches, and stir at room temperature for about 2 hours to complete the reaction. After the reaction is completed, add about 20 ml of saturated brine to quench the reaction, concentrate the methanol in the reaction system, and add dichloromethane for extraction. The organic phase is washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to obtain a white solid substance 1-8, which can be directly used in the next step. |
| Synthesis of compound 1-9: |
| Dissolve the intermediate 1-8 (129 mg, 0.29 mmol) in 20 ml of dichloromethane, add Dess-Martin oxidant (147 mg, 0.35 mmol) and solid sodium bicarbonate (29 mg, 0.35 mmol), and stir at room temperature. After the reaction is complete by TLC monitoring (ultraviolet), filter the reaction system, extract the filtrate with saturated sodium bicarbonate, and the organic layer is purified by saturated sodium salt. |
| The product was washed with water, dried over anhydrous sodium sulfate and concentrated. The product was separated and purified by flash column chromatography (CH2Cl2:MeOH=20:1) to obtain 77 mg of compound 1 as a white solid powder with a yield of 60%. |
| Synthesis of compound 1-10: |
| Compound 1-9 (129 mg, 0.29 mmol) was dissolved in dichloromethane solvent, acetic acid (19.2 mg, 0.32 mmol) and benzyl isocyanate (37.6 mg, 0.32 mmol) were added to react to obtain compound 1-10. Flash column chromatography (CH 2 Cl 2 :MeOH=20:1) to separate and purify to obtain 126 mg of white solid powder compound 1-10 with a yield of 70%. |
| Synthesis of compound 1-11: |
| Compound 1-10 (187 mg, 0.3 mmol) was dissolved in methanol solvent, LiOH (0.6 mmol) was added and stirred to obtain compound 1-11. 2 Cl 2 :MeOH=20:1) to separate and purify to obtain 148 mg of white solid powder compound 1-11 with a yield of 85%. |
| Synthesis of compound 1-12: |
| Compound 1-11 (174 mg, 0.3 mmol) was dissolved in dichloromethane solvent, Dess-Martin oxidant (152 mg, 0.36 mmol) was added, sodium bicarbonate (30 mg, 0.36 mmol) was added, and stirred to obtain a white solid powder compound 1-12 of 140 mg in total, with a yield of 80%. |
| 1 H NMR(500MHz,Chloroform)δ9.76(s,1H),7.73(s,1H),7.39(s,1H),7.32–7.26(m,2H),7.22(s,1H),7 .20–7.10(m,3H),7.01(s,1H),6.82(s,1H),6.68(s,1H),6.14(s,1H),5.57(s,1H),5.43(s,1H),4.3 8(s,1H),4.32(d,J=19.2Hz,2H),3.45(s,1H),3.35(s,1H),3.06(s,1H),2.20(dd,J=15.4,2.3Hz,4H ),2.12–2.03(m,2H),1.92(s,1H),1.77(s,1H),1.73–1.67(m,3H),1.66–1.53(m,6H),1.37(s,1H).; |
PATENT
WO2020030143
bioRxiv (2020), 1-17
- [1]. Ullrich S, Nitsche C. The SARS-CoV-2 main protease as drug target. Bioorg Med Chem Lett. 2020 Sep 1;30(17):127377. [Content Brief]
- [2]. Shang W, et al. In vitro and in vivo evaluation of the main protease inhibitor FB2001 against SARS-CoV-2. Antiviral Res. 2022 Dec;208:105450. [Content Brief]
///BOFUTRELVIR, FB2001, FB 2001, Phase 3, COVID 2019, T5UX5SKK2S, Mpro inhibitor, DC-402234, DC402234, MPI-10
Bleximenib



Bleximenib
CAS 2654081-35-1
WeightAverage: 599.796
Monoisotopic: 599.395916661
Chemical FormulaC32H50FN7O3
- CS-0636752
- DA-55335
- HY-148669
- PHASE 3
- JNJ-75276617; Menin-MLL inhibitor 24
- Benzamide, N-ethyl-5-fluoro-2-[[5-[2-[(1R)-4-[(2-methoxyethyl)methylamino]-1-(1-methylethyl)butyl]-2,6-diazaspiro[3.4]oct-6-yl]-1,2,4-triazin-6-yl]oxy]-N-(1-methylethyl)-
- N-ethyl-5-fluoro-2-{[5-(2-{(3R)-6-[(2-methoxyethyl)(methyl)amino]-2-methylhexan-3-yl}-2,6-diazaspiro[3.4]octan-6-yl)-1,2,4-triazin-6-yl]oxy}-N-(propan-2-yl)benzamide

2866179-95-3 (oxalate)
(R)-N-ethyl-5-fluoro-N-isopropyl-2-((5-(2-(6-((2-methoxyethyl)(methyl)amino)-2-methylhexan-3-yl)-2,6-diazaspiro[3.4]octan-6-yl)-1,2,4-triazin-6-yl)oxy)benzamide oxalate
Chemical Formula: C34H52FN7O7
Exact Mass: 689.39
Molecular Weight: 689.830
Elemental Analysis: C, 59.20; H, 7.60; F, 2.75; N, 14.21; O, 16.23
\Bleximenib is under investigation in clinical trials NCT04811560 (A Phase 1/2 Study of Bleximenib in Participants With Acute Leukemia) and NCT05453903 (A Study of Bleximenib in Combination With Acute Myeloid Leukemia (AML) Directed Therapies)
Bleximenib (JNJ-75276617) is an orally active and selective menin-KMT2A inhibitor, with IC50 values of 0.1 nM, 0.045 nM, and ≤0.066 nM for humans, mice, and dogs, respectively. Bleximenib can inhibit the proliferation and induce apoptosis and differentiation of tumor cells. Bleximenib can be used in the research of tumors such as leukemia.
Bleximenib is an orally bioavailable protein-protein interaction (PPI) inhibitor of the menin-mixed lineage leukemia (MLL; mixed-lineage leukemia 1; MLL1; myeloid/lymphoid leukemia; histone-lysine N-methyltransferase 2A; KMT2A) proteins, with potential antineoplastic activity. Upon oral administration, bleximenib inhibits the interaction between the two proteins menin and MLL and the formation of the menin-MLL complex. This reduces the expression of downstream target genes and results in an inhibition of the proliferation of leukemic cells with either KMT2A alterations such as gene rearrangements (KMT2A-r), duplications, and amplification, or nucleophosmin 1 gene (NPM1) alterations. The menin-MLL complex plays a key role in the survival, growth, transformation and proliferation of certain kinds of leukemia cells.
SCHEME

SIDECHAIN

PATENTS
Janssen Pharmaceutica NV; Johnson & Johnson (China) Investment Ltd.
WO2021121327
WO2022237719
PATENT
WO2022237720

PATENTS
PATENT
Compound A—(R)-N-ethyl-5-fluoro-N-isopropyl-2-((5-(2-(6-((2-methoxyethyl) (methyl)amino)-2-methylhexan-3-yl)-2,6-diazaspiro[3.4]octan-6-yl)-1,2,4-triazin-6-yl)oxy) benzamide
PATENT
WO2022262796
The present invention is directed to (R) -N-ethyl-5-fluoro-N-isopropyl-2- ( (5- (2- (6- ( (2-methoxyethyl) (methyl) amino) -2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) benzamide besylate salt (benzenesulfonate salt) :
[0011]
[0140]
tert-butyl (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate
[0141]
[0142]
The mixture 2- ( (5- (2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) -N-ethyl-5-fluoro-N-isopropylbenzamide (intermediate 3) (1.0 g, 2.4 mmol) , tert-butyl (5-methyl-4-oxohexyl) carbamate (intermediate 1) (830 mg, 3.62 mmol) and ZnCl 2(660 mg, 4.84 mmol) in MeOH (15 mL) was stirred at 80 ℃ for 0.5 h. Then NaBH 3CN (310 mg, 4.93 mmol) was added and the resulting mixture was stirred at 80 ℃ for 6 h. After cooled to RT, the mixture was concentrated under reduced pressure to give the crude product, which was further purified by preparative HPLC using a Waters Xbridge Prep OBD (column: C18 150×40 mm 10 um; eluent: ACN/H 2O (0.05%ammonia) from 45%to 75%v/v) to afford the title compound (700 mg, 46%yield) as colorless oil.
reparation of Compounds 62 and 63
[0144]
tert-butyl (R) – (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate
[0145]
tert-butyl (S) – (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate
[0146]
[0147]
tert-butyl (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate (Compound 61) (200 mg, 0.319 mmol) was purified by SFC over DAICEL CHIRALPAK IG (column: 250×30 mm 10 um; isocratic elution: EtOH (containing 0.1%of 25%ammonia) : supercritical CO 2, 40%: 60% (v/v) ) to afford the title compounds (Compound 62) (85 mg, 42%yield) and (Compound 63) (80 mg, 40%yield) both as light yellow oil.
[0148]
[0149]
(R) -2- ( (5- (2- (6-amino-2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) -N-ethyl-5-fluoro-N-isopropylbenzamide
[0150]
[0151]
To the solution of tert-butyl (R) – (4- (6- (6- (2- (ethyl (isopropyl) carbamoyl) -4-fluorophenoxy) -1, 2, 4-triazin-5-yl) -2, 6-diazaspiro [3.4] octan-2-yl) -5-methylhexyl) carbamate (Compound 62) (550 mg, 0.876 mmol) in DCM (4 mL) was slowly added TFA (4 mL) , and the resulting mixture was stirred at 25 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was diluted in DCM (40 mL) and the pH value was adjusted to around 12 by aq. NaOH (2 M, 16 mL) solution. The aqueous layer was extracted with DCM (10 mL x 2) . The combined organic layers were dried over anhydrous Na 2SO 4, filtered and concentrated in vacuo to afford the title compound (460 mg, crude) as yellow solid, which was used directly in next step without further purification.
[0152]
[0153]
(R) -N-ethyl-5-fluoro-N-isopropyl-2- ( (5- (2- (6- ( (2-methoxyethyl) amino) -2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) benzamide
[0154]
[0155]
The mixture of (R) -2- ( (5- (2- (6-amino-2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) -N-ethyl-5-fluoro-N-isopropylbenzamide (Compound 64) (120 mg, crude) , 1-bromo-2-methoxyethane (32 mg, 0.23 mmol) , Cs 2CO 3(222 mg, 0.681 mmol) , NaI (102 mg, 0.680 mmol) in DMF (1 mL) was stirred at 80 ℃ via microwave irradiation for 1 h. After cooling to RT, the mixture was diluted with H 2O (10 mL) and extracted with EtOAc (3 x 10 mL) . The combined organic layers were washed with H 2O (10 mL) , dried over Na 2SO 4, filtered and concentrated under reduced pressure to afford the crude product which was further purified by HPLC over a Phenomenex Gemini-NX (column: 150×30 mm 5 μm; eluent: ACN/H 2O (10mM NH 4HCO 3) from 51%to 71% (v/v) ) and further purified by SFC over DAICEL CHIRALCEL OD-H (column: 250×30 mm 5 um; eluent: supercritical CO 2in EtOH (0.1%v/v ammonia) 25/25, v/v) to afford the title compound (5.13 mg, 96%purity) as yellow solid.
[0156]
LC-MS (ESI) (Method 1) : R t= 2.997 min, m/z found 586.3 [M+H] +.
[0157]
[0158]
(R) -N-ethyl-5-fluoro-N-isopropyl-2- ( (5- (2- (6- ( (2-methoxyethyl) (methyl) amino) -2-methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) benzamide
[0159]
[0160]
The mixture of (R) -N-ethyl-5-fluoro-N-isopropyl-2- ( (5- (2- (6- ( (2-methoxyethyl) amino) -2- methylhexan-3-yl) -2, 6-diazaspiro [3.4] octan-6-yl) -1, 2, 4-triazin-6-yl) oxy) benzamide (Compound 11) (40.0 mg, 0.068 mmol) , formaldehyde (55.4 mg, 0.683 mol, 37%in water) and AcOH (8.2 mg, 0.137 mmol) in anhydrous MeOH (2 mL) was stirred at 45 ℃ for 1 h. Then, NaBH 3CN (8.6 mg, 0.137 mmol) was added to the mixture and the resulting mixture was stirred at 45 ℃ for another 1 h. After cooling to RT, the reaction mixture was treated with sat. aq. NaHCO 3(40 mL) to adjust the pH value to about 8 and further extracted with DCM (20 mL x 3) . The combined organic layers were dried over anhydrous Na 2SO 4, filtered and concentrated under reduced pressure to give the crude which was purified by preparative HPLC over Boston Prime (column: C18 150x30mm 5um, Mobile Phase A: H 2O (0.04%ammonia+10mM NH 4HCO 3) , Mobile Phase B: ACN, Flow rate: 25 mL/min, gradient condition B/A from 50%to 80% (50%B to 80%B) ) to afford the title compound (9.62 mg, 99.10%purity, 23.3%yield) as yellow oil.
- [1]. Kwon MC, et al. Preclinical efficacy of the potent, selective menin-KMT2A inhibitor JNJ-75276617 (bleximenib) in KMT2A- and NPM1-altered leukemias. Blood. 2024 Sep 12;144(11):1206-1220. [Content Brief][2]. Hogeling SM, et al. Bleximenib, the novel menin-KMT2A inhibitor JNJ-75276617, impairs long-term proliferation and immune evasion in acute myeloid leukemia. Haematologica. 2024 Dec 19. [Content Brief]
////////Bleximenib, CS-0636752, DA-55335, HY-148669, JNJ-75276617, Menin-MLL inhibitor 24
Bezisterim, HE 3286; NE-3107




Bezisterim, HE 3286; NE-3107
CAS 1001100-69-1
(1R,3aS,3bR,4R,7S,9aR,9bS,11aS)-1-ethynyl-9a,11a-dimethyl-1H,2H,3H,3aH,3bH,4H,6H,7H,8H,9H,9aH,9bH,10H,11H,11aH-cyclopenta[a]phenanthrene-1,4,7-triol
- (3β,7β,17α)-Pregn-5-en-20-yne-3,7,17-triol
- 17α-Ethynyl-5-androstene-3β,7β,17β-triol
- 17α-Ethynyl-Δ5-androstene-3β,7β,17β-triol
- 17α-Ethynylandrost-5-ene-3β,7β,17β-triol
- 3β,7β,17β-Trihydroxy-17α-ethynylandrost-5-ene
- Bezisterim
- HE 3286
- NE 3107
- Triolex
(3S,7R,8R,9S,10R,13S,14S,17R)-17-ethynyl-10,13-dimethyl-1,2,3,4,7,8,9,11,12,14,15,16-dodecahydrocyclopenta[a]phenanthrene-3,7,17-triol
| Formula | C21H30O3 |
|---|---|
| Molar mass | 330.468 g·mol−1 |
Q27286562
(3beta,7beta,17alpha)-Pregn-5-en-20-yne-3,7,17-triol
17.ALPHA.-ETHYNYL-5-ANDROSTENE-3.BETA.,7.BETA.,17.BETA.-TRIOL
PREGN-5-EN-20-YNE-3,7,17-TRIOL, (3.BETA.,7.BETA.,17.ALPHA.)-
- (1R,3aS,3bR,4R,7S,9aR,9bS,11aS)-1-ethynyl-9a,11a-dimethyl-1H,2H,3H,3aH,3bH,4H,6H,7H,8H,9H,9aH,9bH,10H,11H,11aH-cyclopenta(a)phenanthrene-1,4,7-triol
- (1R,3aS,3bR,4R,7S,9aR,9bS,11aS)-1-ethynyl-9a,11a-dimethyl-1H,2H,3H,3aH,3bH,4H,6H,7H,8H,9H,9aH,9bH,10H,11H,11aH-cyclopenta[a]phenanthrene-1,4,7-triol
- 17-ethynyl-5-androstene-3, 7, 17-triol

Bezisterim (developmental code names NE3107, HE3286) is a synthetic analogue of androstenetriol that is believed to have anti-inflammatory and insulin-sensitizing effects in the brain.[1] The compound crosses the blood–brain barrier and does not activate any neurotransmitter receptors.[2] It has been tested as a treatment for Alzheimer’s disease,[3][4][5][6] Parkinson’s disease,[1] and traumatic brain injury.[7] The drug is under development for a variety of conditions and its highest developmental phase is phase 3 for Alzheimer’s disease.[1]
- Originator Hollis-Eden Pharmaceuticals
- Developer BioVie; Harbor Therapeutics; National Institutes of Health (USA); NeurMedix
- Class Anti-inflammatories; Antidementias; Antiepileptic drugs; Antifibrotics; Antiglaucomas; Antihyperglycaemics; Antimigraines; Antineoplastics; Antiparkinsonians; Antirheumatics; Hormones; Insulin sensitisers; Nootropics; Obesity therapies; Small molecules
- Mechanism of Action Adiponectin stimulants; Interleukin 23 inhibitors; Interleukin 6 inhibitors; Mitogen-activated protein kinase 1 inhibitors; Mitogen-activated protein kinase 3 inhibitors; NF-kappa B inhibitors; Tumour necrosis factor inhibitors
- Cystic fibrosis
- Phase III Alzheimer’s disease
- Phase II Parkinson’s disease; Traumatic brain injuries
- Preclinical Multiple myeloma; Prostate cancer
- No development reported Drug-induced dyskinesia
- Discontinued Amyotrophic lateral sclerosis; Cognition disorders; Cystic fibrosis; Epilepsy; Glaucoma; Huntington’s disease; Migraine; Myositis; Optic neuritis; Rheumatoid arthritis; Type 1 diabetes mellitus; Type 2 diabetes mellitus; Ulcerative colitis; Uveitis
28 Feb 2025BioVie plans the phase II ADdRESs-LC trial for Post-acute COVID-19 syndrome in USA (PO, Capsule), in February 2025 (NCT06847191)
- 18 Feb 2025Phase-II clinical trials in Parkinson’s disease (Early-stage disease, In the elderly) in USA (PO) (NCT06757010)
- 03 Jan 2025BioVie plans a phase II SUNRISE-PD trial for Parkinsons disease (Early stage disease) in February 2025 (PO) (NCT06757010)
SCHEME

US20100227841
https://patentscope.wipo.int/search/en/detail.jsf?docId=US43352763&_cid=P11-M9JSD6-84971-1
17α-Ethynylandrost-5-ene-3β,7β,17β-triol was prepared as follows
US20100222315 https://patentscope.wipo.int/search/en/detail.jsf?docId=US43344622&_cid=P11-M9JSIE-88638-1
WO2009149392
PATENT’
WO2009149392
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2009149392&_cid=P11-M9JSL7-90448-1


49] Example 7. Synthesis of 3β-acetoxy-androst-5-ene-17,17-ethylenedioxy: A 300L reactor was charged with 36 kg of triethylorthoformate, 20 kg of 3β-acetoxy-5-androsten-17-one, 12.6 kg of ethylene glycol and 400 g of p-toluenesulfonic acid. The mixture was heated to reflux under nitrogen until the reaction was complete (about 2-3 hours). The mixture was then cooled to 60 0C and 16 kg of anhydrous ethanol and 400 ml of pyridine were added. The resulting solution was transferred to a container and refrigerated overnight. The solids that formed were filtered and washed with 80 kg of 50% ethanol and dried at 40-50 0C to afford 18.5-21.0 kg (81.5-92.5%) of the title compound. [50] Example 8. Synthesis of 3β-acetoxy-androst-5-en-7-one-17,17-ethylenedioxy: A 500 L reactor was charged with 200 kg ethyl acetate and 25 kg of 3β-acetoxy-androst-5-en-17,17-ethylenedioxy. The mixture was stirred for 30 minutes whereupon 55 kg of 70% t-butyl peroxide and 9 kg of sodium bicarbonate were added. The reaction mixture was then cooled to 0 0C and 116 kg of 13% sodium perchlorate (aq.) was added over 10 hours so that a reaction temperature below 5 0C and pH between 7.5 and 8.5 were maintained. After the reaction was complete, the organic layer was separated and the aqueous phase was extracted with ethyl acetate (35 kg x 2). The combined organic phase was combined with a solution 33 kg of sodium sulfite in 167 kg of water, and the resulting mixture was stirred at 40 0C for 3 hours. The organic phase was washed with 50 kg of brine and concentrated to 55-60 kg whereupon 50 kg of methanol was added. After refrigeration overnight, a white solid was formed that was filtered and washed with 10 kg of methanol, and dried at 40-50 0C to yield 7.1-7.8 kg (27.4-30.1%) of the title compound.
[51] Example 9. Synthesis of 3β-acetoxy-androst-5-ene-17,17-ethylenedioxy-7β-ol. A 500 L reactor was charged with 48 kg of THF, 10 kg of 3β-acetoxy-androst-5-en-7-one-17,17-ethylenedioxy and a solution of 9.6 kg CeCI3-7H2O in 95 kg methanol. This mixture was cooled to 0 0C whereupon 2.0 kg of NaBH4 was added in batches over 3 hours in order to maintain the temperature below 5 0C. After stirring for 30 more minutes, 28 kg of acetone was added slowly in order to maintain the temperature below 5 0C, with stirring continued for another 30 minutes. To the mixture was added 240 kg water with stirring continued for 1 hour. The organic solvents were removed under vacuum and the residue was extracted with ethyl acetate (100 kg + 50 kg). The combined organic phase was washed with brine. Solvent was then removed to provide 8.6-8.9 kg (85.1-88.1 %) of the title compound. [52] Example 10. Synthesis of 3β-acetoxy-androst-5-en-17-one-7β-ol: A 500 L reactor was charged with 315 kg of acetone and 18 kg of 3β-acetoxy-androst-5-en-17,17-ethylenedioxy-7β-ol. The mixture was cooled to 5 0C and 2.34 kg of p-toluenesulfonic acid was added slowly to maintain the temperature below 10 0C. After stirring the mixture at 8-15 0C for 36-48 hours, 3.0 kg of sodium bicarbonate was added with stirring continued for 1 hour. Acetone was removed under vacuum, and to the residue was added 100 kg of water. The mixture was placed in a refrigerator overnight to give a white precipitate which was filtered to provide 33 kg (wet) of the title compound.
[53] Example 11. Synthesis of androst-5-en-17-one-3β,7β-diol: A 500 L reactor was charged 230 kg methanol, 33 kg (wet) 3β-acetoxy-7β-hydroxy-5-androsten-17-one, 108 kg water and 15 kg NaaCOβ. The mixture was heated to reflux for 3 hours. Methanol was removed under vacuum whereupon 250 kg of water was added to the residue. The mixture was put in refrigerator overnight to give a precipitate. The solids were collected by filtration, then washed with water and dried at 40-50 0C to yield 9.5-10.5 kg (67.9-75.0%) of the title compound as a white solid.
[54] Example 12. Purification of androst-5-en-17-one-3β,7β-diol: A 500 L reactor was charged with 20 kg crude 3β, 7β-dihydroxyandrost-5-en-17-one and 200 kg methanol and heated until all the solid dissolved. The solution was filtered while hot and after the filtrate cooled a white crystalline solid formed. The solids were collected by filtration, washed with small amount of methanol and dried at 40-50 0C. The solid was then refluxed in 50 kg of ethyl acetate for 20 minutes. After cooling the solid was filtered and dried at 40-50 0C under vacuum to provide 15.2 kg (76%) of purified title compound.
[55] Example 13. Synthesis of 3β,7β-bis-(trimethylsiloxy)-5-androsten-17-one: A mixture of 14.87 Kg of androst-5-en-17-one-3β,7β-diol, 23.8 Kg HMDS and 0.7 Kg saccharin catalyst in 100 L acetonitrile was heated to reflux for 8 hours with stirring under a nitrogen atmosphere. Liberated ammonia was purged under slight vacuum. The reaction volume was then reduced by distillation to collect 3OL of distillate (requires about 2 h). The reaction volume was further reduced to half of the original reaction volume by distillation under reduced pressure (700 mmHg), which requires about 2h of heating at 50 0C. The resulting uniform thick slurry is cooled to 5 0C (requires about 3 h), with additional acetonitrile added to maintain a minimum mixing volume, and held at that temperature for 1. The precipitated product was collected by filtration and dried at 45-50 0C under vacuum (29 mmHg) to a loss on drying (LOD) of not more than 1 % (requires 20 h) to provide 16 Kg (81 % yield) of the title compound (95% purity). [56] Example 14. Synthesis of 17α-ethynyl-5-androstene-3β,7β,17β-triol: To 11.02 Kg TMS-acetylene in 56.5 L tetrahydrofuran (THF) at -27 0C under a nitrogen atmosphere was added 8.51 L 10M n-BuLi. The n-butyl lithium was added very slowly to maintain a temperature at -7 to -27 0C (requires about 2 h) and the resulting reaction was stirred 10 min. at approximately 0°C to produce TMS-lithium-acetylide. To the TMS-lithium-acetylide solution was added a solution of 25.41 Kg of 3β,7β-bis-(trimethylsiloxy)-5-androsten-17-one in 95.3 L THF filtered through a 25 μm filter while allowing the reaction temperature to rise to 20-25 0C. After addition was completed, the reaction temperature was increased to 40-45 0C. To quench the reactor contents, 31.8 L of methanol was added over a period of about 1 h followed by 3.81 Kg KOH in 18.4 L of water giving a final reactor temperature of 50 0C. Liberated acetylene is purged under slight vacuum. The reactor contents were then concentrated by distillation at 80 0C for 1 h then under vacuum (175 mmHg) at about 70 0C (with an initial temperature of 25 0C to avoid bumping) to half of the original pot volume. The residue was cooled to about 10 0C and 35.0 Kg of deionized water was added, followed by 16.4 Kg 12N HCI while maintaining a pot temperature of about 10 0C and giving a final pH of 1. Additional 26.0 kg deionized water was added and the resulting mixture was stirred at about 5 0C for 1 h. The resulting slurry was filtered and washed with 75/25 mixture of methanol/water (16.9 L methanol, 5.6 L water). The collected solids were dried under vacuum (28 in Hg) at 45 0C for 12h for a loss on drying of no more than 0.5% to provide 9.6 Kg of the title compound (83% yield).
[57] Example 15. Recrystallization of 17α-ethynyl-5-androstene-3β,7β,17β-triol: Crude 9.6 Kg 17α-ethynyl-5-androstene-3β,7β,17β-triol prepared in
Example 14 was dissolved in refluxing 50/50 methanol/water (4.2 Kg methanol and 5.4 Kg water). To the solution was added 33.4 Kg methanol followed by 37.6 Kg of THF. The mixture was heated to reflux and stirring was continued until all solids have dissolved, whereupon 99.8 Kg of deionized water was added while maintaining a reactor temperature of 60-75 0C. The mixture was cooled to 0-5 0C over a period of 2 h and maintain at that temperature for 1 h while stirring was continued. The solids were recovered by filtration, washed with 9.6 Kg cold 50/50 methanol water and dried under vacuum (28 in Hg) at 50 0C for 8 h to provide 8.2 Kg of 17α-ethynyl-5-androstene-3β,7β,17β-triol. This first recrystallization is used to remove trace colored impurities from the initial product. A second recrystallization was conducted by heating the solid from the first recrystallization in ~10:1 methanohwater (145.8 Kg methanol and 18.2 Kg of water) to 80°C until all the solids have dissolved. The solution at 55-60 0C was filtered through a 25 μm filter to remove particulate impurities, whereupon 2.5 Kg of methanol at 55-60 0C (used to rinse the reactor) was added. Vacuum distillation at 125 mmHg at 70 0C was conducted until 0.9 to 1.2 times the volume of methanol that was added to the reactor was collected as distillate with water added as necessary to permit stirring (about 120-160 Kg water added). Final reaction volume was 200-225 L. The reactor mixture was cooled to 0-5 0C and maintained at that temperature for 1 h. The resulting slurry was filtered and the filter cake rinsed with 10 Kg deionized water and dried under vacuum (28 in Hg) at 50 0C for 12 h to a residual water content of less than 0.5%. This isolation procedure was used to reduce the THF content in the final product. The yield was 8.0 Kg of recrystallized title compound (83% yield).

[59] Example 16. Synthesis of 3β-acetoxy-androst-5-en-7-on-17-oxime: 3β-Acetoxy-androst-5-en-7,17-dione (45 g, 130 mmol) was dissolved in 800 ml_ methanol, 200 ml_ dichloromethane and 14.5g Et3N (144 mmol). To the solution at RT was added a solution of 10 g of hydroxylamine hydrochloride dissolved in 200 ml_ methanol. After stirring overnight, 200 ml_ of water was added followed by removal of volatile organics by evaporation under reduced pressure. To the resulting residue was added an additional 1 L of water to give a while solid that was filtered and washed well with water. Obtained was 45 g of crude title oxime in 95% purity by 1H-NMR, which was used in the next step without further purification.
[60] Example 17. Synthesis of 3β-acetoxy-androst-5-en-17-oxime-7β-ol: To a solution of 44 g of 3β-acetoxy-androst-5-en-7-on-17-oxime (100 mol%) in 800 ml_ methanol and 200 ml_ tetrahydrofuran was added 50 g of cerium chloride heptahydrate (110 mol%) in 20 ml_ of methanol. The resulting mixture was stirred until the solids were completely dissolved. To the solution cooled to about -5 0C was added 7 g sodium borohydride over 30 min. After stirring an additional 1.5 h at -5 0C, the reaction mixture was quenched with acetone (100 mL) and then allowed to warm to room temperature over a 30 min. period. The quenched reaction mixture was concentrated under vacuum to remove volatile organics. To the residue was added 800 mL of water followed by extraction with ethyl acetate (3 x 500 mL). The combined organic extracts were washed with brine, dried over Na2SO4, then concentrated to provide 42 g of the title compound as a white foam, which was used in the next step without further purification.. [61] Example 18. 3β-acetoxy-androst-5-en-17-one-7β-ol: To a solution of 42 g of 3β-acetoxy-androst-5-en-17-oxime-7β-ol (100 mol%) in 200 mL of ethanol was added 100 mL of water followed by 80 g (400 mol%) of sodium dithionite. The reaction was heated at 55 0C and stirred 16 h. After cooling, the reaction was concentrated under reduced pressure. The residue was diluted with 100 mL of water, and the resulting solid was collected by filtration and redissolved in 1 L dichloromethane. To the DCM solution was added 1 g activated carbon. After stirring overnight the mixture was filtered, and the resulting filtrate was washed with water, dried and concentrated to provide 25 g of crude product. Recrystallization from ethyl acetate gave 22g of the title compound. [62] Example 19. Estrogen receptor binding assay: A suitable example system is an estrogen receptor- kit manufactured by PanVera for ERβ, which contains recombinant estrogen receptor β ligand, FLUORMONE™ ES2 (ES2), a fluorescently labeled estrogen ligand, and appropriate buffer. The system was used in a fluorescence polarization competition assay in which a test article, such as a preparation of Compound 1 or a positive control displaces ES2 from its binding site. When bound to ERβ, ES2 tumbles slowly and has a high fluorescence polarization value. Unbound ES2 tumbles quickly and displays a low fluorescence polarization value. The change in polarization value in the presence of test compound then determines relative binding affinity of that test compound for ERβ as expressed by its IC50, which is the concentration of test compound that results in half-maximum shift in polarization. From IC50, K/ was calculated using the Cheng-Prusoff equation [Biochem. Pharmacol. 22: 3099-3108, (1973)]: K, = IC50Z(I + D/Kd) where D is the concentration of ES2 and Kd is the dissociation constant for binding of ES2 to ERβ (Kd = 4 ± 2 nM).
[63] The competition assay was conducted according to the manufacturer’s protocol (Lit. No. L0712, Rev. 10/03). Assay reagents used were bacculovirus expressed, full length human ERβ 4.5 pmol/μL in 50 mM Bis-Tris Propane (pH = 9), 400 mM KCI, 2 mM DTT, 1 mM EDTA, 10% glycerol, ES2 400 nM in methanol and E2 screening buffer consisting of 100 mM potassium phosphate (pH = 7.4), 100 μg/mL BGG, 0.02% NaN3. The ES2-ERβ complex was formed with 20 μL 20 nM ERβ (0.020 pmol/μL) and 20 μl_ 2 nM ES2 (0.002 pmol/μL). Positive control (estrogen) solution was prepared using 20 μL of a 1.0 mM stock solution in DMSO and 80 μL DMSO. In a first dilution, 50 μL of this solution is added to 50 μL of DMSO, which is followed by dilutions in 2-fold increments, to provide for a 14 point dilution curve. In a second dilution, to 4 μL of each DMSO solution from the first dilution is added 400 μL of ES2 screening buffer. To 20 μL of test compound, serially diluted in the manner described immediately above, in a 384 well black flat bottom microtiter plate, was added 20 μL of the ES2-ERβ complex (0.5% final DMSO concentration) followed by incubation in the dark at 20-30 0C for 1-4 h. Test compound was treated similarly except the starting concentration was 10 mM. Fluorescence polarization values are obtained using 485 nm excitation and 530 nm emission interference filters. Binding assay for ERa was conducted as for ERβ except bacculovirus expressed, full length human 2.8 pmol/μL ERa was used as reagent with the ERα-ES2 complex formed from 20 μL 30 nM (0.030 pmol/μL) and 20 μL 2 nM ES2 (0.002 pmol/μL). [64] Example 20. AR, GR and PR receptor binding assays. The AR competition assay was conducted according to the manufacturer’s protocol (Lit. No. L0844, Rev. 05/02) in the manner described for ERβ with the following exceptions. Reagents used were recombinant rat androgen receptor ligand binding domain tagged with His and GST [AR-LBD (His-GST)] 0.38 pmol/μL in buffer containing protein stabilizing agents and glycerol (pH = 7.5), 200 nM FLUORMONE™ AL Green, which is a fluorescently labeled androgen ligand, in 20 mM Tris, 90% methanol and AR screening buffer containing stabilizing agents and glycerol (pH = 7.5) with 2 μL of 1 mM DTT added per mL screening buffer (AR screening buffer 2 mM in added DTT) was used as the reagents. The AL Green-AR complex was formed with 20 μL 50 nM AR (0.050 pmol/μL) and 20 μL 2 nM AL Green (0.002 pmol/μL). K, was calculated using, for the dissociation constant for binding of the fluorophore to receptor, Kd = 20 ± 10 nM. [65] The PR competition assay was conducted according to the manufacturer’s protocol (Lit. No. L0503, Rev. 06/03) in the manner described for ERβ with the following exceptions. Reagents used were recombinant human progesterone receptor ligand binding domain tagged with GST [PR-LBD (GST)] 3.6 pmol/μL in 50 mM Tris (pH = 8.0), 500 mM KCI, 1 M urea, 5 mM DTT, 1 mM EDTA and 50% glycerol, 400 nM FLUORMONE™ PL Green, which is a fluorescently labeled progesterone ligand, in 20 mM Tris 90% methanol (pH = 6.8) and PR screening buffer containing protein stabilizing agents and glycerol (pH = 7.4) with 4 μL of 1 mM DTT added per mL screening buffer (PR screening buffer 4 mM in added DTT). The PL Green-PR complex was formed with 20 μL 80 nM PR (0.080 pmol/μL) and 20 μL 4 nM PL Green (0.004 pmol/μL). K, was calculated using, for the dissociation constant for binding of the fluorophore to receptor, Kd = 40 nM.
[66] The GR competition assay was conducted according to the manufacturer’s protocol (Lit. No. L0304, Rev. 12/01) in the manner described for ERβ with the following exceptions. Reagents used were recombinant full length human glucocorticoid receptor 0.240 pmol/μL in 10 mM phosphate buffer (pH = 7.4), 200 mM Na2MoO4, 0.1 mM EDTA, 5 mM DTT and 10% glycerol, 200 nM FLUORMONE™ GS1 , which is a fluorescently labeled glucocorticoid ligand, in 75% methanol, and GR screening buffer containing 100 mM potassium phosphate (pH = 7.4), 200 mM Na2MoO4, 1 mM EDTA, 20% DMSO with 5 μL of 1 mM DTT per mL screening buffer added (GR screening buffer 5 mM in added DTT), 1 mM GR stabilizing peptide, which is a co-activator related peptide [see Chang, CY. MoI. Cell Biol. 19: 8226-36 (1999)] in 10 mM phosphate buffer (pH = 7.4) and 1 M DTT in water were used as the reagents. To 2.5 mL of the GR screening buffer is added 2.5 mL GR stabilizing peptide solution and 125 μL of 1 M DTT to form the GR stabilizing peptide-glucocorticoid receptor complex. Order of addition to the microtiter plate was 20 μL test compound in 1 % DMSO, 10 μL of 16 nM GR (0.016 pmol/μL) and finally 10 μL of 4 nM GS1 , followed by incubation in the dark at 20-30 0C for 4 h (total experiment time should not exceed 7 h). K, was calculated using, for the dissociation constant for binding of the fluorophore to receptor, Kd = 0.3 ± 0.1 nM.
[67] Example 21. Impurity profiling of 17α-ethynyl-5-androstene-3β,7β,17β- triol (Compound 1) preparations.
[68] Process A: HPLC conditions for Impurity profiling of Compound 1 preparations form Process B are give in Table 1.
[69]
Table 1. HPLC Conditions for Impurity Profiling of Compound 1 Preparations form Process A
PATENT
Hollis-Eden Pharmaceuticals, Inc. WO2008039566
Zhejiang Xianju Junye Pharmaceutical Co., Ltd.; Jiangxi Junye Biopharmaceutical Co., Ltd.CN114478672
Harbor BioSciences, Inc.US20100227841
Harbor BioSciences, Inc. US20100222315 A1
Hollis-Eden Pharmaceuticals, Inc. US20100075937
Neurmedix Inc. US20080153792 A1
Hollis-Eden Pharmaceuticals, Inc.; Harbor Therapeutics, Inc. US20080146532 A1
Harbor Therapeutics, Inc.; Neurmedix, Inc. US20160045516 A1
Harbor Therapeutics, Inc. US8354396 B2
Hollis-Eden Pharmaceuticals, Inc. WO2009149392
| Clinical data | |
|---|---|
| Other names | NE3107; NE-3107; HE3286; HE-3286; 17α-Ethynyl-5-androstene-3β,7β,17β-triol; |
| Legal status | |
| Legal status | Investigational |
| Identifiers | |
| showIUPAC name | |
| CAS Number | 1001100-69-1 |
| PubChem CID | 16739648 |
| DrugBank | DB05212 |
| ChemSpider | 20571043 |
| UNII | PH8858757I |
| KEGG | D12932 |
| ChEMBL | ChEMBL4297284 |
| CompTox Dashboard (EPA) | DTXSID501267252 |
| Chemical and physical data | |
| Formula | C21H30O3 |
| Molar mass | 330.468 g·mol−1 |
| 3D model (JSmol) | Interactive image |
| showSMILES | |
| showInChI | |
References
- ^ Jump up to:a b c “Bezisterim”. AdisInsight. 5 September 2024. Retrieved 26 September 2024.
- ^ Reading, Chris L; Ahlem, Clarence N; Parameswaran, Narayanan (December 2021). “Rationale for an anti-inflammatory insulin sensitizer in a phase 3 Alzheimer’s disease trial”. Alzheimer’s & Dementia. 17 (S9). doi:10.1002/alz.057438.
- ^ Stoiljkovic, Milan; Horvath, Tamas L.; Hajós, Mihály (July 2021). “Therapy for Alzheimer’s disease: Missing targets and functional markers?”. Ageing Research Reviews. 68: 101318. doi:10.1016/j.arr.2021.101318. PMC 8131215. PMID 33711510.
- ^ Balzano, Tiziano; Esteban-García, Noelia; Blesa, Javier (2 January 2023). “Neuroinflammation, immune response and α-synuclein pathology: how animal models are helping us to connect dots”. Expert Opinion on Drug Discovery. 18 (1): 13–23. doi:10.1080/17460441.2023.2160440. PMID 36538833. S2CID 254959175.
- ^ Liu, Ping; Wang, Yunyun; Sun, Yan; Peng, Guoping (April 2022). “Neuroinflammation as a Potential Therapeutic Target in Alzheimer’s Disease”. Clinical Interventions in Aging. 17: 665–674. doi:10.2147/CIA.S357558. PMC 9064449. PMID 35520949.
- ^ Xi, Yilong; Chen, Yun; Jin, Yi; Han, Guochen; Song, Mingjie; Song, Tingting; Shi, Yang; Tao, Ling; Huang, Zewei; Zhou, Jianping; Ding, Yang; Zhang, Huaqing (May 2022). “Versatile nanomaterials for Alzheimer’s disease: Pathogenesis inspired disease-modifying therapy”. Journal of Controlled Release. 345: 38–61. doi:10.1016/j.jconrel.2022.02.034. PMID 35257810. S2CID 247285338.
- ^ “U.S. Clinical Trial: Neurological Associates of West Los Angeles Listed a New Clinical Trial to Study Insulin-sensitizing NE3107 in Improving Sleep and Fatigue in Subjects With Traumatic Brain Injury.” Contify Life Science News, 1 Aug. 2023, p. NA. Gale OneFile: Health and Medicine, link.gale.com/apps/doc/A759542006/HRCA?u=anon~bb46c85&sid=sitemap&xid=0c315c7e. Accessed 14 Dec. 2023.
/////Bezisterim, HE 3286, NE 3107, Triolex, NE3107, NE-3107, HE3286, HE-3286, PHASE 2
Tibremciclib


Tibremciclib
cas 2397678-18-9, GTPL12881
CRB7BT5JDQ
518.6 g/mol, C28H32F2N8
N-[5-[(4-ethylpiperazin-1-yl)methyl]pyridin-2-yl]-5-fluoro-4-[(1R)-6-fluoro-1-methyl-1,2,3,4-tetrahydropyrido[1,2-a]benzimidazol-8-yl]pyrimidin-2-amine
Tibremciclib is a CDK4 inhibitor with antineoplastic activity[1].
- Originator Betta Pharmaceuticals Co Ltd
- Class Antineoplastics; Small molecules
- Mechanism of Action Cyclin-dependent kinase 4 inhibitors; Cyclin-dependent kinase 6 inhibitors
- Phase III Breast cancer; Solid tumours
13 Sep 2024 Efficacy and adverse event data from a phase III trial in Breast cancer presented at the 49th European Society for Medical Oncology Congress 2024 (ESMO-2024)
- 30 Jun 2023Phase-III clinical trials in Breast cancer (Metastatic disease, Late-stage disease, Combination therapy, Second-line therapy or greater) in China (PO) (NCT05433480)
- 02 Jun 2023Efficacy, adverse events and PK data from a phase I trial in Solid tumours presented at the 59th Annual Meeting of the American Society of Clinical Oncology (ASCO-2023)
Cyclin-dependent kinases (CDKs) are a class of serine / threonine protein kinases that participate in the regulation of the cell cycle, transcription initiation, and control of certain specific metabolic cascades. Different CDKs and cyclins form CDK-cyclin complexes. If the CDK activity is dysregulated, it will directly or indirectly cause uncontrolled cell proliferation, genomic instability (increased DNA mutation, chromosome deletion, etc.) and chromosomal instability (change in chromosome number). )Wait.
The CDKs family has identified more than 20 subtypes. CDK1, CDK2, CDK4, and CDK6 are involved in cell cycle regulation; CDK7, CDK8, CDK9, and CDK11 are involved in transcription regulation; and other kinases include CDK3 and CDK5. Among them, CDK4 / 6 (cyclin-dependent kinases 4 and 6) is a key factor in regulating the cell cycle. Cancer-related cell cycle mutations mainly exist in the G1 and G1 / S phase transformation. CDK4 / 6 binds to CyclinD A complex with kinase activity is formed and phosphorylation of the tumor suppressor gene Rb product pRb releases the bound transcription factor E2F to initiate transcription of genes related to the S phase, prompting cells to pass the checkpoint and transfer from the G1 phase to the S phase. The specific activation of CDK4 / 6 is closely related to the proliferation of some tumors. About 80% of human tumors have abnormalities in the cyclin D-CDK4 / 6-INK4-Rb pathway. CDK4 / 6 inhibitors block the cell cycle in the G1 phase, thereby inhibiting tumor proliferation.
The development of drugs targeting CDK4 / 6 kinases is a significant area. The advantages of anti-tumor targets are: (1) Most proliferating cells rely on CDK2 or CDK4 / 6 to proliferate, but CDK4 / 6 inhibitors do not show Cytotoxicity of “pan-CDK inhibitors”, such as bone marrow suppression and intestinal response; (2) Preclinical experiments show that if the level of cyclin D or the inactivation of P16INK4a can increase the sensitivity of cells to drugs, due to tumors Compared with normal cells, cells have the above phenomenon, so the targeting of drugs is increased to a certain extent.
PCT International Application PCT / CN2017 / 117950 describes a class of benzimidazole derivatives that are used as CDK4 / 6 protein kinase inhibitors, and most of these compounds effectively inhibit CDK4 and CDK6. Because there are still unmet needs in the treatment options for kinase-mediated diseases, here we further screen the salt forms and crystal forms of benzimidazole derivatives to meet the medical needs of patients.
SCHEME
SIDE CHAIN

SIDE CHAIN

MAIN

Patent
Betta Pharmaceuticals Co., Ltd., WO2019242719
https://patents.google.com/patent/WO2019242719A1/en


Synthesis of 1-A1-01 (Step 1)
In a 50L reactor, add 20L of dichloromethane (DCM), 1-A1-S1 (300g), and triethylamine (390g). While stirring, lower the temperature to below -5 ° C, and add benzyl chloroformate / Cbz- Cl (570 g) was added dropwise for 5 hours, and the temperature was naturally raised to room temperature. TLC (ethyl acetate: n-hexane = 1: 3) was monitored until the reaction was completed. Water (1.5 L) was added, and concentrated hydrochloric acid (80 mL) was slowly added dropwise to control the pH to 1-2. The solution was allowed to stand and separate. The organic phase was washed with 15 L of water, dried over anhydrous sodium sulfate for 0.5 hours, filtered to remove the desiccant, and collected the filtrate. And concentrated to obtain 730 g of light yellow oily liquid, which is crude 1-A1-01, yield 95.4%
Synthesis of 1-A1-02 (Step 2)
720mL of DCM, N, N-dimethylsulfoxide (90g) was added to a 20L reaction flask, protected by nitrogen, and the temperature was lowered below -65 ° C under stirring, and oxalyl chloride (106g) was added dropwise. The addition was completed in 2 hours. Stir for 20 minutes under heat preservation; add 1-A1-01’s dichloromethane solution (143g / 500mL DCM) dropwise. After 40 minutes, the addition is complete and the reaction is held for 15 minutes. Controlled at this temperature, TEA was added dropwise. After the addition was completed for 2 hours, the temperature was naturally raised to -20 ° C. 250 L of water was added to the system. The pH of the system was adjusted to 1-2 with hydrochloric acid. × 2) Washed, dried over anhydrous sodium sulfate, filtered to remove the desiccant, collected the filtrate and concentrated to obtain 432 g of a yellow oily liquid, which is the crude product 1-A1-02, which was directly used in the next reaction.
Synthesis of 1-A1-03 (Step 3)
In a stirred state, 400 mL of tetrahydrofuran (THF) and potassium tert-butoxide (215 g) were sequentially added to a 1 L reaction kettle, the temperature was lowered to 5-15 ° C., and triethyl phosphoryl acetate (430 g) was added dropwise. The dropwise addition was completed in 50 minutes. At a controlled temperature of 15 ° C, a tetrahydrofuran solution of 1-A1-02 (431 g / 100 mL of THF) was added dropwise. After the dropwise addition was completed for 1 hour, TLC (ethyl acetate: n-hexane = 1: 3) was monitored to complete the reaction, and the system was added. Saturated aqueous sodium chloride solution (1.5L), allowed to stand and separate, and collected the tetrahydrofuran phase; the aqueous phase was extracted with dichloromethane (2L), and the organic phases were combined and dried over anhydrous sodium sulfate for 0.5 hours, and the drying agent was removed by filtration. The filtrate was collected and concentrated, and the concentrate was purified by column chromatography to obtain 390 g of a pale yellow oily liquid, which was 1-A1-03 product.
Synthesis of 1-A1-041 (step 4)
In a 5L reactor, an aqueous solution of sodium hydroxide (301 g / 1.5 L of water) was added to a tetrahydrofuran (601 g / 2.3 L of THF) solution of 1-A1-03, and the mixture was heated to reflux for 3-4 hours to stop the reaction. The temperature was lowered to 40-50 ° C, and the layers were left to stand. The organic phase (THF) was collected and concentrated to a large amount of solids; the solids were dissolved by adding water (20L), and the aqueous phase was sequentially treated with methyl tert-butyl ether (2L) and ethyl acetate. Ester (2L), methyl tert-butyl ether (2L) washing; the aqueous phase was adjusted to pH 1-2 with concentrated hydrochloric acid, extracted twice with ethyl acetate (1.5L, 3L), the organic phases were combined, and anhydrous sulfuric acid was used Sodium was dried for 0.5 hours; the desiccant was removed by filtration, and the filtrate was collected and concentrated to a large amount of solids. The solids were added with isopropyl ether (3L) and slurried for 2 hours. The solids were collected by filtration and the solids were rinsed with isopropyl ether (1L). The solid was air-dried at 50 ° C for 3-4 hours to obtain 331 g of a pale yellow solid, which is a 1-A1-041 product with a yield of 52.7%.
Synthesis of 1-051 (step 5)
In a stirred state, 1-A1-041 (600g), methanol (25L), and concentrated sulfuric acid were added to a 50L reactor, and the reaction was heated under reflux for 3-4 hours. After the reaction was completed, the temperature was reduced to room temperature. Dichloromethane (15L) was added to the concentrate, and the pH was adjusted to 9-10 with an aqueous solution of potassium carbonate. The organic phase was collected by stirring, standing, and separating. The organic phase was dried over anhydrous sodium sulfate for 0.5 hours. The desiccant was removed by filtration and the filtrate was collected. And concentrated to obtain 6.37 kg of off-white solid, which is 1-A1-051 product, with a yield of 97.3%.
Synthesis of 1-A1 (step 6)
In a 2L hydrogenation kettle, add 1-A1-051 (500g), methanol (1.8L), and palladium on carbon. The system replaces nitrogen 3 times and hydrogen 3 times in sequence. The system maintains a hydrogen atmosphere, and the temperature is increased to 85 ° C and the pressure is 3.0. The reaction was carried out at Mpa for 3 hours, and the reaction was completed. The temperature was lowered to room temperature, the palladium on carbon was removed by filtration, and the organic phase was collected and concentrated until a large amount of light yellow solid appeared. Isopropyl ether (3L) was added to freeze (-20 ° C) for crystallization, and the solid product was collected by filtration. Ether (500 mL) was rinsed to obtain 234 g of a pale yellow solid, which was a 1-A1 product with a yield of 90.5%.
Synthesis of 1-A2 (Step 7)
In a stirred state, 1-A1 (200g), 4-bromo-2,6-difluoroaniline (410g), and toluene (1.2L) were added to a 50L reactor, and phosphorus oxychloride (413g) was added dropwise to the system. The addition was completed in 1 hour. Triethylamine was added dropwise under an ice bath, and the addition was completed in 1 hour. The temperature was raised to 110 ° C, and the reaction was performed for 1 hour. Reduce the temperature of the system to 2-10 ° C, add 1L of water, adjust the pH = 9-10 with saturated potassium carbonate aqueous solution, extract twice with ethyl acetate (1.5L, 1L), and combine the organic phases with 2L saturated sodium chloride aqueous solution. Wash, dry with anhydrous sodium sulfate for 0.5 hours, remove the desiccant by filtration, collect the filtrate and concentrate to the appearance of a solid product, add isopropyl ether (1L) to beat the solid for 10 minutes, filter, and collect 460 g of a yellow solid as a 1-A2 product.
Synthesis of 1-A3 (step eight)
Under stirring, 1-A2 (450g), N, N-dimethylformamide (2L), and cesium carbonate (700g) were added to the reaction kettle, and the reaction was heated to 110 ° C for 24 hours, and the reaction was detected by TLC. Ethyl acetate (3L) was added to the system, and solid impurities were removed by filtration. The filtrate was washed with a saturated sodium chloride aqueous solution (1L × 5), and the organic phase was dried over anhydrous sodium sulfate for 0.5 hours, concentrated to the appearance of a large amount of solid, Butyl ether (1L × 2) was beaten for 30 minutes, and 382 g of pale yellow solid product was obtained by filtration, that is, 1-A3, and the yield was 90.10%.
Synthesis of 1-01 (step 9)
With stirring, 1-A3 (380 g), pinacol diborate (400 g), potassium acetate (340 g), palladium acetate (6 g), tricyclohexyl phosphorus (7 g), and 1,4-dioxane were sequentially added. The ring was added to the reaction kettle, protected by nitrogen, and heated to 90 ° C for 2 hours. TLC was monitored until the reaction was complete. The temperature was reduced to room temperature, and the filtrate was concentrated to remove a large amount of 1,4-dioxane. The concentrate was purified by n-hexane and dichloromethane column chromatography, and n-hexane (1.2 L) was slurried for 1 hour to obtain 334 g of a gray solid. That is 1-01, and the yield is 70.10%.
Synthesis of 1-02 (step 10)
Under stirring, take 1-01 (128g), 1,4-dioxane (1L), 1-S3 (85g), potassium carbonate (110g), and purified water and add them to a 2L three-necked flask in sequence. [1,1′-Bis (diphenylphosphine) ferrocene] palladium dichloromethane complex (Pd (dppf) Cl 2 .DCM) was added. The temperature was raised to 60 ° C. After 4 hours of reaction, the reaction was complete. The reaction solution was cooled to room temperature, and concentrated under reduced pressure to remove most of 1,4-dioxane. Dichloromethane (1.5 L) and purified water (1.1 L) were added, stirred, and allowed to stand and separate. The layers were separated, and water was added. The phases were extracted with dichloromethane (10 L), the organic phases were combined, washed with 0.5% dilute hydrochloric acid (1 L x 2), saturated aqueous sodium chloride solution (1 L), and the layers were separated. The organic phase was dried over anhydrous sodium sulfate (500 g), filtered to remove the drying agent, and the filtrate was concentrated under reduced pressure. Ethyl acetate (0.5 L) was added to the concentrate and the mixture was stirred for 30 minutes to precipitate a solid. After filtration, the obtained solid was rinsed with ethyl acetate (0.5 L) and dried under vacuum at 45 ° C for 3 hours to obtain 120 g of a yellow solid.
Synthesis of 1-03 (step 11)
Under stirring, take 1-02 (100g), 1,4-dioxane (1L), 1-C2 (80g), and cesium carbonate (163g) into a 2L three-necked bottle in sequence, protected by nitrogen, and add palladium acetate ( 2g) and 4,5-bisdiphenylphosphine-9,9-dimethylxanthracene (Xantphos) (4g), heated to 85 ° C. until the reaction was complete. The reaction solution was cooled to room temperature and filtered to obtain a solid product. The solid was rinsed with ethyl acetate, and then added to a mixed system of dichloromethane (1.5 L) and purified water (1.1 L), stirred, allowed to stand, and separated into layers. The aqueous phase was extracted with dichloromethane (700 mL). The organic phases were combined and washed with purified water (700 mL x 2). The organic phase was dried by adding anhydrous sodium sulfate (700 g), filtered to remove the desiccant, and the filtrate was concentrated. Methanol (0.5 L) was added, heated to 55-65 ° C. and stirred for 0.5 hours, lowered to room temperature, and filtered. The solid product was filtered and rinsed with 500 mL of ethyl acetate. The solid was dried under vacuum at 45 ° C for 8 hours to obtain 111.79 g of a pale yellow solid 1-03.
Synthesis of compound II (step twelve)
Under stirring, take 1-03 (500g) and anhydrous methanol (3.8L), add them to a 10L reactor in sequence, and heat to 65 ° C. After the reaction system is clarified for 0.5 hours, add L-tartaric acid in methanol (150.89) dropwise. g of tartaric acid is dissolved in 500mL of anhydrous methanol), and the dropping time is controlled to be 45 to 60 minutes. After the addition is complete, the reaction is kept at 65 ° C for 4 hours. ), Control the dropwise addition time to 30 to 45 minutes. After the dropwise addition is complete, hold the reaction at 65 ° C for 1 hour. Continue to dropwise add L-tartaric acid in methanol (36.55g of tartaric acid dissolved in 250mL of anhydrous methanol) and control the dropwise addition time to 30. To 45 minutes, the dropwise addition was completed. The temperature was kept at 65 ° C for 1.5 hours, and the heating was stopped. The temperature was naturally lowered to 20-30 ° C, filtered, the filter cake was rinsed with methanol (400mL × 2), and dried at 45 ° C under vacuum for 36 hours. 530.64 g of crystalline powder was Compound II, which was identified by X-ray powder diffraction, and showed that the crystal form was Form A of Compound II.
WO2022199656
WO2023131179
///Tibremciclib, GTPL12881, BETTA, PHASE 3, CANCER
ATICAPRANT


ATICAPRANT
1174130-61-0
BENZAMIDE, 4-(4-(((2S)-2-(3,5-DIMETHYLPHENYL)-1-PYRROLIDINYL)METHYL)PHENOXY)-3-FLUORO-
C26H27FN2O2, 418.512
- 4-[4-[[(2S)-2-(3,5-Dimethylphenyl)-1-pyrrolidinyl]methyl]phenoxy]-3-fluorobenzamide (ACI)
- (S)-4-(4-((2-(3,5-Dimethylphenyl)pyrrolidin-1-yl)methyl)phenoxy)-3-fluorobenzamide
- 4-(4-{[(2S)-2-(3,5-dimethylphenyl)pyrrolidin-1-yl]methyl}phenoxy)-3-fluorobenzamide
- Aticaprant
- CERC 501
- JNJ 67953964
- JNJ 67953964AAA
- LY 2456302
- S-Aticaprant
- CERC-501
- JSPA 0658 JSPA-0658 JSPA0658
- LY 2456302 LY-2456302 , LY2456302
- OriginatorEli Lilly and Company
- DeveloperAvalo Therapeutics; Eli Lilly and Company; Johnson & Johnson Innovative Medicine
- ClassAntidepressants; Benzamides; Benzene derivatives; Drug withdrawal therapies; Fluorinated hydrocarbons; Pyrrolidines; Smoking cessation therapies
- Mechanism of ActionOpioid kappa receptor antagonists
- Phase III Major depressive disorder
- DiscontinuedAlcoholism; Cocaine-related disorders; Smoking withdrawal
- 26 Jun 2024Janssen Research & Development initiates a phase III VENTURA-7 trial for Major depressive disorder (Adjunctive treatment) in USA (PO, Tablet) (NCT06514742) (EudraCT2024-511557-21-00)
- 01 Oct 2023Janssen Pharmaceuticals is now called Johnson & Johnson Innovative Medicine (Janssen Pharmaceuticals website, October 2023)
- 19 May 2023Chemical structure information added
Aticaprant, also known by its developmental codes JNJ-67953964, CERC-501, and LY-2456302, is a κ-opioid receptor (KOR) antagonist which is under development for the treatment of major depressive disorder.[2][3][4] A regulatory application for approval of the medication is expected to be submitted by 2025.[2] Aticaprant is taken by mouth.[1]
Side effects of aticaprant include itching, among others.[4][5] Aticaprant acts as a selective antagonist of the KOR, the biological target of the endogenous opioid peptide dynorphin.[3] The medication has decent selectivity for the KOR over the μ-opioid receptor (MOR) and other targets, a relatively long half-life of 30 to 40 hours, and readily crosses the blood–brain barrier to produce central effects.[4][6]
Aticaprant was originally developed by Eli Lilly, was under development by Cerecor for a time, and is now under development by Janssen Pharmaceuticals.[2] As of July 2022, it is in phase 3 clinical trials for major depressive disorder.[2] Like other kappa opioid antagonists currently under clinical investigation for the treatment of major depression, its efficacy may be compromised by the countervailing activation of pro-inflammatory cytokines in microglia within the CNS.[7]
Aticaprant was also under development for the treatment of alcoholism, cocaine use disorder, and smoking withdrawal, but development for these indications was discontinued.[2]
Pharmacology
Pharmacodynamics
Aticaprant is a potent, selective, short-acting (i.e., non-“inactivating”) antagonist of the KOR (Ki = 0.81 nM vs. 24.0 nM and 155 nM for the μ-opioid receptor (MOR) and δ-opioid receptor (DOR), respectively; approximately 30-fold selectivity for the KOR).[8][9][10] The drug has been found to dose-dependently block fentanyl-induced miosis at 25 mg and 60 mg in humans (with minimal to no blockade at doses of 4 to 10 mg), suggesting that the drug significantly occupies and antagonizes the MOR at a dose of at least 25 mg but not of 10 mg or less.[10] However, a more recent study assessing neuroendocrine effects of the drug in normal volunteers and subjects with a history of cocaine dependence reported observations consistent with modest MOR antagonism at the 10 mg dose.[11] In animal models of depression, aticaprant has been found to have potent synergistic efficacy in combination with other antidepressants such as citalopram and imipramine.[12]
Positron emission tomography imaging revealed that brain KORs were almost completely saturated by the drug 2.5 hours following a single dose of 10 mg, which supported the 4 mg to 25 mg dosages that aticaprant is being explored at in clinical trials.[13][14] Occupancy was 35% for a 0.5 mg dose and 94% for a 10 mg dose.[15][14] At 24 hours post-dose, receptor occupancy was 19% for 0.5 mg and 82% for 25 mg.[15][14] No serious side effects were observed, and all side effects seen were mild to moderate and were not thought to be due to aticaprant.[14]
Pharmacokinetics
The oral bioavailability of aticaprant is 25%.[1] The drug is rapidly absorbed, with maximal concentrations occurring 1 to 2 hours after administration.[1] It has an elimination half-life of 30 to 40 hours in healthy subjects.[1] The circulating levels of aticaprant increase proportionally with increasing doses.[1] Steady-state concentrations are reached after 6 to 8 days of once-daily dosing.[1] Aticaprant has been shown to reproducibly penetrate the blood–brain barrier.[13][14]
History
Aticaprant was originally developed by Eli Lilly under the code name LY-2456302.[2] It first appeared in the scientific literature in 2010 or 2011.[16][17] The compound was first patented in 2009.[18]
In February 2015, Cerecor Inc. announced that they had acquired the rights from Eli Lilly to develop and commercialize LY-2456302 (under the new developmental code CERC-501).[19]
As of 2016, aticaprant has reached phase II clinical trials as an augmentation to antidepressant therapy for treatment-resistant depression.[20][12] A phase II study of aticaprant in heavy smokers was commenced in early 2016 and results of the study were expected before the end of 2016.[14] Aticaprant failed to meet its main endpoint for nicotine withdrawal in the study.[21]
In August 2017, it was announced that Cerecor had sold its rights to aticaprant to Janssen Pharmaceuticals.[22][21] Janssen was also experimenting with esketamine for the treatment of depression as of 2017.[21]
Research
In addition to major depressive disorder, aticaprant was under development for the treatment of alcoholism, cocaine use disorder, and smoking withdrawal.[2] However, development for these indications was discontinued.[2]
See also
κ-Opioid receptor § Antagonists
SCHEME

SYNTHESIS
WO/2024/178082COMPOSITION OF OPIOID RECEPTOR MODULATOR AND MDMA FOR USE THEREOF
WO/2024/173843QUINOLINE DERIVATIVES WHICH ACT AS KAPPA-OPIOID RECEPTOR ANTAGONISTS
20240238245COMPOSITIONS AND METHODS FOR THE TREATMENT OF DEPRESSION
20240189274Compositions And Methods For The Treatment Of Depression
WO/2024/102802ZELATRIAZIN FOR THE TREATMENT OF DEPRESSION
WO/2024/100285TREATMENT OF A COGNITIVE DISORDER WITH AN AGENT THAT INCREASES THE..
117615757Compositions and methods for treating depression
117142999Racemization method of drug intermediate
20230348377PURE FORMS OF CRYSTALLINE ATICAPRANT
WO/2023/170550POLYMORPH FORMS OF ATICAPRANT FOR USE IN TREATING MAJOR DEPRESSIVE DISORDER
WO/2023/170547PURE FORMS OF CRYSTALLINE ATICAPRANT
20230277499Forms of aticaprant
20230277500COMPOSITIONS COMPRISING ATICAPRANT
WO/2023/164385NEUROACTIVE STEROIDS FOR TREATMENT OF GASTROINTESTINAL DISEASES OR CONDITIONS
20090186873Kappa selective opioid receptor antagonist
WO/2009/094260KAPPA SELECTIVE OPIOID RECEPTOR ANTAGONIST
20100197669Kappa selective opioid receptor antagonist
2252581KAPPA SELECTIVE OPIOID RECEPTOR ANTAGONIST
201500053151-substituted 4-arylpiperazine as kappa opioid receptor antagonists
WO/2013/0864961-SUBSTITUTED 4-ARYLPIPERAZINE AS KAPPA OPIOID RECEPTOR ANTAGONISTS
101925576Kappa selective opioid receptor antagonist
PAPERS
ACS Omega (2020), 5(41), 26938-26945 https://pubs.acs.org/doi/full/10.1021/acsomega.0c04329


REF https://pubs.acs.org/doi/suppl/10.1021/acsomega.0c04329/suppl_file/ao0c04329_si_001.pdf
N-Methoxy-N-methyl-4-chlorobutyramide (S1). To a mixture of N,O-dimethylhydroxylamine hydrochloride (95.0 mmol, 9.27 g) in CH2Cl2 (150 mL) was
added 2 M NaOH (300 mmol, 150 mL) and 4-chlorobutyryl chloride (100 mmol,
11.2 mL) at 0 ˚C. The mixture was stirred for 42 h at room temperature. The
organic phase was separated, and the aqueous phase was extracted with CH2Cl2 (2 × 50 mL). The combined organic phase was washed with 2 M NaOH (100 mL), dried over Na2SO4, filtered, and concentrated
to afford the title comlund in 75% yield as a colorless liquid.
1H NMR (400 MHz, CDCl3) : 2.08-2.15
(m, 2H), 2.63 (t, J = 7.0 Hz, 2H), 3.19 (s, 3H), 3.64 (t, J = 6.3 Hz, 2H), 3.71 (s, 3H).
13C{
1H} NMR (100
MHz, CDCl3) : 27.1, 28.6, 32.1, 44.6, 61.1. IR (max/cm-1
): 2965, 2940, 2821, 1656, 14421, 1417, 1387,
1178, 1107, 997. HRMS (ESI+): calculated for [M+Na]+
: 188.0449, found: 188.0450.
4-Chloro-1-(3,5-dimethylphenyl)butan-1-one (S2). To a mixture of N-methoxy-N-methyl-4-chlorobutyramide (S1, 65.0 mmol, 10.8 g) in anhydrous Et2O
(100 mL) was added dropwise 3,5-dimethylphenylmagnesium bromide (ca. 1 M
in Et2O, ca. 130 mmol, prepared from 1-bromo-3,5-dimethylbenzene (130 mmol,
17.7 mL) and Mg turnings (169 mmol, 4.11 g) in anhydrous Et2O (130 mL)) over 1 h at -40 ˚C under Ar.
The reaction mixture was stirred at room temperature for 20 h. After cooling to 0 ˚C, saturated NH4Cl
solution (200 mL) was added. The organic phase was separated, washed with water (100 mL) and brine
(100 mL), dried over Na2SO4, and filtered. After concentration, the residue was purified by column chromatography (silica gel, hexane/EtOAc as eluent) to afford the title compound in 91% yield as a greenish
yellow liquid.
1H NMR (400 MHz, CDCl3) : 2.18-2.25 (m, 2H), 2.38 (s, 6H), 3.15 (t, J = 7.0 Hz, 2H),
3.67 (t, J = 6.3 Hz, 2H), 7.21 (s, 1H), 7.58 (s, 2H). 13C{
1H} NMR (100 MHz, CDCl3) : 21.2, 26.8, 35.4,
44.7, 125.8, 134.8, 136.8, 138.3, 199.4. IR (max/cm-1
): 3047, 3006, 2961, 2920, 2868, 1443, 1411, 1322,
1303, 1181, 1159, 844, 785, 687. HRMS (APCI+): calculated for [M+H]+
: 211.0884, found: 211.0884.
(RS)-N-(4-Chloro-1-(3,5-dimethylphenyl)butylidene)-tertbutanesulfinamide (S3). Ti(OEt)4 (100 mol, 21.0 mL) was added to a mixture
of (RS)-tert-butanesulfinamide (1.0 M in THF, 50 mmol, 50 mL) and 4-chloro1-(3,5-dimethylphenyl)butan-1-one (S2, 50.0 mmol, 10.5 g) under N2. The mixture was refluxed for 48 h. After cooling to room temperature, brine (100 mL)
was added, and the resulting mixture was filtered over Celite using EtOAc (ca.
300 mL). The organic was separated, dried over Na2SO4, and filtered. After concentration under reduced
pressure, the residue was purified by column chromatography (silica gel, hexane/EtOAc as eluent) to
afford the title compound in 57% yield as a brown viscous liquid.
1H NMR (400 MHz, CDCl3) : 1.33
(s, 9H), 2.10-2.22 (m, 2H), 2.36 (s, 6H), 3.27 (s, 1H), 3.43 (s, 1H), 3.64 (t, J = 6.5 Hz, 2H), 7.13 (s, 1H),
7.47 (s, 2H).
13C{
1H} NMR (100 MHz, CDCl3) : 21.3, 22.7, 30.2, 31.6, 44.7, 57.7, 125.2, 133.4, 137.6,
138.2, 178.6. IR (max/cm-1
): 3046, 2958, 2922, 2866, 1599, 1577, 1455, 1361, 1320, 1308, 1069, 856.
HRMS (ESI+): calculated for [M+H]
+
: 314.1340, found: 314.1344. []D
20 +11.0 (c = 1.01, CH2Cl2).
(RS,S)-1-tert-Butylsulfinyl-2-(3,5-dimethylphenyl)pyrrolidine (S4). To a solution of (RS)-N-(4-chloro-1-(3,5-dimethylphenyl)butylidene)-tert-butanesulfinamide
(S3, 25.6 mmol, 8.06 g) in anhydrous THF (100 mL) at -78 °C was added LiBEt3H
(28 mmol, 0.5 M in THF, 28.2 mL) under Ar. The reaction was stirred at -78 °C for
1 h, subsequently allowed to warm up to room temperature and stirred for additional
20 h. Saturated NaHCO3 solution (80 mL) was slowly added. The mixture was filtered and extracted
with EtOAc (3 × 100 mL). The combined organic phase was dried over Na2SO4 and filtered. After
concentration, the residue was purified by column chromatography (silica gel, hexane/EtOAc as eluent)
to afford the title compound in 72% yield as pale yellow solid. mp.: 56 ˚C. 1H NMR (400 MHz, CDCl3)
: 1.12 (s, 9H), 1.74-1.90 (m, 3H), 1.93-2.02 (m, 1H), 2.18-2.27 (m, 1H), 2.30 (s, 6H), 2.94-3.02 (m, 1H),
3.85-3.91 (m, 1H), 4.55-4.59 (m, 1H), 6.88 (s, 1H), 6.90 (s, 2H).
13C{
1H} NMR (100 MHz, CDCl3) :
21.3, 23.8, 26.3, 36.0, 42.1, 57.2, 69.2, 125.0, 128.7, 137.7, 143.2. IR (max/cm-1
): 3023, 2957, 2920,
2866, 1607, 1471, 1360, 1061, 957, 847. HRMS (ESI+): calculated for [M+Na]+
: 302.1549, found:
302.1548. []D
20
-137 (c = 0.49, CH2Cl2)
(S)-2-(3,5-Dimethylphenyl)pyrrolidine hydrochloride (1j•HCl). To a solution
of (RS,S)-1-tert-butylsulfinyl-2-(3,5-dimethylphenyl)pyrrolidine (S4, 14.7 mmol,
4.12 g) in dioxane (250 mL) was added dropwise HCl (ca. 150 mmol, 4 M in dioxane, 38 mL). The mixture was stirred for 1 h at room temperature under N2, and
then the mixture was concentrated under reduced pressure. Then, Et2O (200 mL) was added to the residue
and the mixture was cooled to 0 ˚C. The precipitate was collected by filtration, washed with Et2O (40
mL), and dried under reduced pressure to afford the title compound in 94% yield as white solid. mp.: 198
˚C. 1H NMR (400 MHz, D2O) : 2.00-2.15 (m, 3H), 2.18 (s, 6H), 2.27-2.35 (m, 1H), 3.27-3.36 (m, 2H),
4.45 (t, J = 8.0 Hz, 1H), 6.97 (s, 2H), 7.01 (s, 1H). 13C{
1H} NMR (100 MHz, D2O) : 20.9, 24.19, 30.9,
46.0, 63.8, 119.79, 125.6, 131.4, 135.3, 140.1. IR (max/cm-1
): 3033, 3012, 2970, 2855, 2743, 2571, 2480,
1608, 1590, 1414, 850. HRMS (ESI+): calculated for [M-Cl]
+
: 176.1434, found: 176.1435. []D
20 +7.1
(c = 1.01, MeOH).
(S)-2-(3,5-Dimethylphenyl)pyrrolidine (1j). To a suspension of (S)-2-(3,5-dimethylphenyl)pyrrolidine hydrochloride (1j•HCl, 13.5 mmol, 2.86 g) in anhydrous Et2O
(200 mL) was added a saturated solution of NaHCO3 (200 mL). The resulting mixture
was stirred for 20 min at room temperature. The organic was separated and the aqueous
phase was extracted with Et2O (2 × 100 mL). The combined organic phase was dried over MgSO4 and
filtered. The solvent was removed under reduced pressure to afford the title compound as a pale yellow
liquid in 99% yield.
1H NMR (400 MHz, CDCl3) : 1.60-1.71 (m, 1H), 1.78-1.96 (m, 2H), 1.98 (s, 1H),
2.11-2.19 (m, 1H), 2.30 (s, 6H), 2.95-3.02 (m, 1H), 3.17-3.23 (m, 1H), 4.03 (t, J = 7.7 Hz, 1H), 6.87 (s,
1H), 6.97 (s, 2H). 13C{
1H} NMR (100 MHz, CDCl3) : 21.3, 25.5, 34.2, 46.9, 62.6, 124.2, 128.4, 137.8,
144.7. IR (max/cm-1
): 3332, 3010, 2960, 2915, 2869, 1605, 1458, 1101, 845. HRMS (ESI+): calculated
for [M+H]+
: 176.1434, found: 176.1436. []D
20
-30.5 (c = 1.01, MeOH). Chiral HPLC (ChiralPak ODH, 4.6 mm × L 250 mm, hexane:2-propanol = 90:10, 0.5 mL/min, = 254 nm): tR/min = 18.7 (1%),
19.8 (99%).

3-Fluoro-4-(4-formylphenoxy)benzonitrile2
(S5). A mixture of 3,4-
difluorobenzonitrile (35.0 mmol, 4.87 g), 4-hydroxybenzaldehyde (35.0
mmol, 4.27 g), and K2CO3 (70.0 mmol, 9.67 g) in N,N-dimethylacetamide
(90 mL) was stirred at 100 ˚C for 2 h under N2. After cooling, the reaction
mixture was poured into ice water. White precipitate was collected by filtration, washed with water, and dried under reduced pressure to afford the title compound as pale yellow
solid in 82% yield. mp.: 101 ˚C. 1H NMR (400 MHz, CDCl3) : 7.11-7.15 (m, 2H), 7.20 (t, J = 8.2 Hz,
1H), 7.49-7.51 (m, 1H), 7.54 (dd, J = 9.7, 1.9 Hz, 1H), 7.91-7.94 (m, 2H), 9.98 (s, 1H).
13C{
1H} NMR
(100 MHz, CDCl3) : 109.1 (d, 3
JC-F = 8.2 Hz), 117.1 (d, 4
JC-F = 2.5 Hz), 117.9, 121.3 (d, 2
JC-F = 21.3 Hz),
122.5 (d, 4
JC-F = 1.6 Hz), 129.6 (d, 3
JC-F = 4.1 Hz), 132.1, 132.7, 147.0 (d, 2
JC-F = 11.5 Hz), 153.6 (d, 1
JCF = 254.8 Hz), 160.7, 190.4. IR (max/cm-1
): 3100, 3060, 2846, 2812, 2761, 2232, 1697, 1687, 1585, 1497,
1277, 1216, 1166, 1114, 836. HRMS (APCI+): calculated for [M+H]+
: 242.0612, found: 242.0616.
3-Fluoro-4-(4-formylphenoxy)benzamide2
(2f). To a mixture of 3-
fluoro-4-(4-formylphenoxy)benzonitrile (S5, 26.0 mmol, 6.27 g) and
K2CO3 (13.0 mmol, 1.80 g) in DMSO (24 mL) was added dropwise 35%
H2O2 (ca. 29 mmol, 3.1 mL) at 10 ˚C over 5 min. The reaction mixture
was stirred at room temperature for 2 h. The reaction mixture was
poured into ice water. White precipitate was collected by filtration, washed with water, and dried under
reduced pressure to afford the title compound as white solid in 92% yield. mp. 129 ˚C. 1H NMR (400
MHz, (D3C)2SO) : 9.96 (s, 1H), 8.12 (s, 1H), 7.96 (d, J = 8.2 Hz, 2H), 7.93 (dd, J = 1.9, 10.0 Hz, 1H),
7.85-7.82 (m, 1H), 7.58 (s, 1H), 7.42 (t, J = 8.2 Hz, 1 H), 7.20 (d, J = 8.2 Hz, 2H).
13C{
1H} NMR (100
MHz, (D3C)2SO) : 116.6 (d, 2
JC-F = 19.7 Hz), 116.9, 122.6, 125.1 (d, 4
JC-F = 3.3 Hz), 131.9 (d, 2
JC-F =
21.3 Hz), 132.1, 132.7 (d, 3
JC-F = 5.7 Hz), 143.7 (d, 3
JC-F = 12.3 Hz), 153.1 (d, 1
JC-F = 248.2 Hz), 161.3,
165.8, 191.5. IR (max/cm-1
): 3356, 3185, 2844, 1668, 1598, 1504, 1433, 1382, 1269, 1218, 1156, 1128,
- HRMS (ESI+): calculated for [M+Na]
+
: 282.0537, found: 282.0541. HRMS (APCI+): calculated
for [M+H]+
: 260.0717, found: 260.0716.
NEXT
Reaction Chemistry & Engineering (2022), 7(8), 1779-1785
Journal of Medicinal Chemistry (2011), 54(23), 8000-8012
| Clinical data | |
|---|---|
| Other names | JNJ-67953964; CERC-501; LY-2456302 |
| Routes of administration | By mouth[1] |
| Pharmacokinetic data | |
| Bioavailability | 25%[1] |
| Elimination half-life | 30–40 hours[1] |
| Identifiers | |
| showIUPAC name | |
| CAS Number | 1174130-61-0 |
| PubChem CID | 44129648 |
| IUPHAR/BPS | 9194 |
| DrugBank | DB12341 |
| ChemSpider | 28424203 |
| UNII | DE4G8X55F5 |
| KEGG | D11831 |
| ChEMBL | ChEMBL1921847 |
| CompTox Dashboard (EPA) | DTXSID90151777 |
| Chemical and physical data | |
| Formula | C26H27FN2O2 |
| Molar mass | 418.512 g·mol−1 |
| 3D model (JSmol) | Interactive image |
| showSMILES | |
| showInChI | |
References
^ Jump up to:a b c d e f g h i Li W, Sun H, Chen H, Yang X, Xiao L, Liu R, et al. (2016). “Major Depressive Disorder and Kappa Opioid Receptor Antagonists”. Translational Perioperative and Pain Medicine. 1 (2): 4–16. PMC 4871611. PMID 27213169.
- ^ Jump up to:a b c d e f g h “CERC 501”. Adis Insight. 30 January 2018.
- ^ Jump up to:a b Browne CA, Wulf H, Lucki I (2022). “Kappa Opioid Receptors in the Pathology and Treatment of Major Depressive Disorder”. In Liu-Chen LY, Inan S (eds.). The Kappa Opioid Receptor. Handbook of Experimental Pharmacology. Vol. 271. pp. 493–524. doi:10.1007/164_2020_432. ISBN 978-3-030-89073-5. PMID 33580854. S2CID 231908782.
- ^ Jump up to:a b c Reed B, Butelman ER, Kreek MJ (2022). “Kappa Opioid Receptor Antagonists as Potential Therapeutics for Mood and Substance Use Disorders”. In Liu-Chen LY, Inan S (eds.). The Kappa Opioid Receptor. Handbook of Experimental Pharmacology. Vol. 271. pp. 473–491. doi:10.1007/164_2020_401. ISBN 978-3-030-89073-5. PMID 33174064. S2CID 226305229.
- ^ Krystal AD, Pizzagalli DA, Smoski M, Mathew SJ, Nurnberger J, Lisanby SH, et al. (May 2020). “A randomized proof-of-mechanism trial applying the ‘fast-fail’ approach to evaluating κ-opioid antagonism as a treatment for anhedonia”. Nature Medicine. 26 (5): 760–768. doi:10.1038/s41591-020-0806-7. PMC 9949770. PMID 32231295. S2CID 256839849.
- ^ Dhir A (January 2017). “Investigational drugs for treating major depressive disorder”. Expert Opinion on Investigational Drugs. 26 (1): 9–24. doi:10.1080/13543784.2017.1267727. PMID 27960559. S2CID 45232796.
- ^ Missig G, Fritsch EL, Mehta N, Damon ME, Jarrell EM, Bartlett AA, et al. (January 2022). “Blockade of kappa-opioid receptors amplifies microglia-mediated inflammatory responses”. Pharmacology, Biochemistry, and Behavior. 212: 173301. doi:10.1016/j.pbb.2021.173301. PMC 8748402. PMID 34826432.
- ^ Rorick-Kehn LM, Witkin JM, Statnick MA, Eberle EL, McKinzie JH, Kahl SD, et al. (February 2014). “LY2456302 is a novel, potent, orally-bioavailable small molecule kappa-selective antagonist with activity in animal models predictive of efficacy in mood and addictive disorders”. Neuropharmacology. 77: 131–144. doi:10.1016/j.neuropharm.2013.09.021. PMID 24071566. S2CID 3230414.
- ^ Lowe SL, Wong CJ, Witcher J, Gonzales CR, Dickinson GL, Bell RL, et al. (September 2014). “Safety, tolerability, and pharmacokinetic evaluation of single- and multiple-ascending doses of a novel kappa opioid receptor antagonist LY2456302 and drug interaction with ethanol in healthy subjects”. Journal of Clinical Pharmacology. 54 (9): 968–978. doi:10.1002/jcph.286. PMID 24619932. S2CID 14814449.
- ^ Jump up to:a b Rorick-Kehn LM, Witcher JW, Lowe SL, Gonzales CR, Weller MA, Bell RL, et al. (October 2014). “Determining pharmacological selectivity of the kappa opioid receptor antagonist LY2456302 using pupillometry as a translational biomarker in rat and human”. The International Journal of Neuropsychopharmacology. 18 (2): pyu036. doi:10.1093/ijnp/pyu036. PMC 4368892. PMID 25637376.
- ^ Reed B, Butelman ER, Fry RS, Kimani R, Kreek MJ (March 2018). “Repeated Administration of Opra Kappa (LY2456302), a Novel, Short-Acting, Selective KOP-r Antagonist, in Persons with and without Cocaine Dependence”. Neuropsychopharmacology. 43 (4): 928. doi:10.1038/npp.2017.245. PMC 5809790. PMID 29422497.
- ^ Jump up to:a b Urbano M, Guerrero M, Rosen H, Roberts E (May 2014). “Antagonists of the kappa opioid receptor”. Bioorganic & Medicinal Chemistry Letters. 24 (9): 2021–2032. doi:10.1016/j.bmcl.2014.03.040. PMID 24690494.
- ^ Jump up to:a b “Publication Reports Human Brain Penetration and Target Engagement of Cerecor’s Oral Kappa Opioid Receptor Antagonist, CERC-501”. BusinessWire. 11 December 2015.
- ^ Jump up to:a b c d e f Naganawa M, Dickinson GL, Zheng MQ, Henry S, Vandenhende F, Witcher J, et al. (February 2016). “Receptor Occupancy of the κ-Opioid Antagonist LY2456302 Measured with Positron Emission Tomography and the Novel Radiotracer 11C-LY2795050”. The Journal of Pharmacology and Experimental Therapeutics. 356 (2): 260–266. doi:10.1124/jpet.115.229278. PMC 4727157. PMID 26628406.
- ^ Jump up to:a b Placzek MS (August 2021). “Imaging Kappa Opioid Receptors in the Living Brain with Positron Emission Tomography”. In Liu-Chen LY, Inan S (eds.). The Kappa Opioid Receptor. Handbook of Experimental Pharmacology. Vol. 271. pp. 547–577. doi:10.1007/164_2021_498. ISBN 978-3-030-89073-5. PMID 34363128. S2CID 236947969.
- ^ Zheng MQ, Nabulsi N, Kim SJ, Tomasi G, Lin SF, Mitch C, et al. (March 2013). “Synthesis and evaluation of 11C-LY2795050 as a κ-opioid receptor antagonist radiotracer for PET imaging”. Journal of Nuclear Medicine. 54 (3): 455–463. doi:10.2967/jnumed.112.109512. PMC 3775344. PMID 23353688.
- ^ Mitch CH, Quimby SJ, Diaz N, Pedregal C, de la Torre MG, Jimenez A, et al. (December 2011). “Discovery of aminobenzyloxyarylamides as κ opioid receptor selective antagonists: application to preclinical development of a κ opioid receptor antagonist receptor occupancy tracer”. Journal of Medicinal Chemistry. 54 (23): 8000–8012. doi:10.1021/jm200789r. PMID 21958337.
- ^ “WO2009094260A1 – Kappa selective opioid receptor antagonist”. Google Patents. 13 January 2009. Retrieved 29 August 2022.
- ^ “Cerecor Bolsters Clinical Pipeline with Acquisition of Phase 2-ready Kappa Opioid Receptor Antagonist from Eli Lilly and Company”. cerecor.com. February 20, 2015. Archived from the original on 2015-02-23. Retrieved March 18, 2015.
- ^ Rankovic Z, Hargreaves R, Bingham M (2012). Drug Discovery for Psychiatric Disorders. Royal Society of Chemistry. pp. 314–317. ISBN 978-1-84973-365-6.
- ^ Jump up to:a b c Bushey R (August 2017). “J&J Adds New Depression Drug to Portfolio”. Drug Discovery and Development Magazine.
- ^ “Cerecor Announces Divestiture of CERC-501 to Janssen Pharmaceuticals, Inc”. Marketwired. August 2017. Archived from the original on 2017-09-01. Retrieved 2017-09-01.
Further reading
- Carlezon WA, Krystal AD (October 2016). “Kappa-Opioid Antagonists for Psychiatric Disorders: From Bench to Clinical Trials”. Depression and Anxiety. 33 (10): 895–906. doi:10.1002/da.22500. PMC 5288841. PMID 27699938.
- Li W, Sun H, Chen H, Yang X, Xiao L, Liu R, et al. (2016). “Major Depressive Disorder and Kappa Opioid Receptor Antagonists”. Translational Perioperative and Pain Medicine. 1 (2): 4–16. PMC 4871611. PMID 27213169.
- Dhir A (January 2017). “Investigational drugs for treating major depressive disorder”. Expert Opinion on Investigational Drugs. 26 (1): 9–24. doi:10.1080/13543784.2017.1267727. PMID 27960559. S2CID 45232796.
- Reed B, Butelman ER, Kreek MJ (2017). “Endogenous opioid system in addiction and addiction-related behaviors”. Current Opinion in Behavioral Sciences. 13: 196–202. doi:10.1016/j.cobeha.2016.12.002. ISSN 2352-1546. S2CID 53149180.
- Rakesh G, Pae CU, Masand PS (August 2017). “Beyond serotonin: newer antidepressants in the future”. Expert Review of Neurotherapeutics. 17 (8): 777–790. doi:10.1080/14737175.2017.1341310. PMID 28598698. S2CID 205823807.
- Helal MA, Habib ES, Chittiboyina AG (December 2017). “Selective kappa opioid antagonists for treatment of addiction, are we there yet?”. European Journal of Medicinal Chemistry. 141: 632–647. doi:10.1016/j.ejmech.2017.10.012. PMID 29107424.
- McHugh KL, Kelly JP (2018). “Modulation of the central opioid system as an antidepressant target in rodent models”. The Opioid System as the Interface between the Brain’s Cognitive and Motivational Systems. Progress in Brain Research. Vol. 239. pp. 49–87. doi:10.1016/bs.pbr.2018.07.003. ISBN 9780444641670. PMID 30314569.
- Bailey SJ, Husbands SM (June 2018). “Targeting opioid receptor signaling in depression: do we need selective κ opioid receptor antagonists?”. Neuronal Signaling. 2 (2): NS20170145. doi:10.1042/NS20170145. PMC 7373229. PMID 32714584.
- Chavkin C (August 2018). “Kappa-opioid antagonists as stress resilience medications for the treatment of alcohol use disorders”. Neuropsychopharmacology. 43 (9): 1803–1804. doi:10.1038/s41386-018-0046-4. PMC 6046055. PMID 29752444.
- Krystal AD, Pizzagalli DA, Mathew SJ, Sanacora G, Keefe R, Song A, et al. (December 2018). “The first implementation of the NIMH FAST-FAIL approach to psychiatric drug development”. Nature Reviews. Drug Discovery. 18 (1): 82–84. doi:10.1038/nrd.2018.222. PMC 6816017. PMID 30591715.
- Lazar MA, McIntyre RS (2019). “Novel Therapeutic Targets for Major Depressive Disorder”. Neurobiology of Depression. pp. 383–400. doi:10.1016/B978-0-12-813333-0.00034-2. ISBN 9780128133330. S2CID 86782597.
- Browne CA, Lucki I (September 2019). “Targeting opioid dysregulation in depression for the development of novel therapeutics”. Pharmacology & Therapeutics. 201: 51–76. doi:10.1016/j.pharmthera.2019.04.009. PMC 6859062. PMID 31051197.
- Banks ML (2020). “The Rise and Fall of Kappa-Opioid Receptors in Drug Abuse Research”. In Nader MA, Hurd YL (eds.). Substance Use Disorders. Handbook of Experimental Pharmacology. Vol. 258. pp. 147–165. doi:10.1007/164_2019_268. ISBN 978-3-030-33678-3. PMC 7756963. PMID 31463605.
- Browne CA, Jacobson ML, Lucki I (2020). “Novel Targets to Treat Depression: Opioid-Based Therapeutics”. Harvard Review of Psychiatry. 28 (1): 40–59. doi:10.1097/HRP.0000000000000242. PMID 31913981. S2CID 210120636.
- Jacobson ML, Browne CA, Lucki I (January 2020). “Kappa Opioid Receptor Antagonists as Potential Therapeutics for Stress-Related Disorders”. Annual Review of Pharmacology and Toxicology. 60: 615–636. doi:10.1146/annurev-pharmtox-010919-023317. PMID 31914893. S2CID 210121357.
- Mercadante S, Romualdi P (2020). “The Therapeutic Potential of Novel Kappa Opioid Receptor-based Treatments”. Current Medicinal Chemistry. 27 (12): 2012–2020. doi:10.2174/0929867326666190121142459. PMID 30666905. S2CID 58558833.
External links
Aticaprant – Eli Lilly and Company/Janssen Pharmaceuticals – AdisInsight
//////ATICAPRANT, CERC-501, JSPA 0658, JSPA-0658, JSPA0658, LY 2456302, LY-2456302, LY2456302, Phase 3, ELI LILLY, Major depressive disorder, JNJ-67953964, WHO 10582
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO
.....










