Home » Uncategorized (Page 8)
Category Archives: Uncategorized
ETRIPAMIL



ETRIPAMIL
CAS 1593673-23-4
AS ACETATE 512.64 CAS 2891832-59-8
HCL SALT 2560549-35-9
WeightAverage: 452.595
Monoisotopic: 452.267507647
Chemical FormulaC27H36N2O4
Benzoic acid, 3-[2-[[(4S)-4-cyano-4-(3,4-dimethoxyphenyl)-5-methylhexyl]methylamino]ethyl]-, methyl ester
methyl 3-[2-[[(4S)-4-cyano-4-(3,4-dimethoxyphenyl)-5-methylhexyl]-methylamino]ethyl]benzoate
- Methyl 3-[2-[[(4S)-4-cyano-4-(3,4-dimethoxyphenyl)-5-methylhexyl]methylamino]ethyl]benzoate
- (-)-MSP 2017
- MSP 2017
- OriginatorMilestone Pharmaceuticals
- DeveloperCorxel Pharmaceuticals; Milestone Pharmaceuticals
- ClassAmines; Antiarrhythmics; Benzoates; Esters; Ischaemic heart disorder therapies; Small molecules
- Mechanism of ActionCalcium channel antagonists
- PreregistrationParoxysmal supraventricular tachycardia
- Phase IIAtrial fibrillation
- Phase IUnspecified
- No development reportedAngina pectoris
- 14 May 2025Milestone Pharmaceuticals has patent protection for etripamil in the USA
- 28 Mar 2025Milestone pharmaceuticals plans to request a Type A meeting with USFDA to discuss the issues raised in the complete response letter
- 28 Mar 2025USFDA has issued a Complete Response Letter (CRL) regarding New Drug Application (NDA) for Etripamil for Paroxysmal supraventricular tachycardia
Etripamil has been used in trials studying the treatment of Paroxysmal Supraventricular Tachycardia (PSVT).
Etripamil (MSP-2017) is a short-acting, L-type calcium-channel antagonist. Etripamil inhibits calcium influx through slow calcium channels, thereby slowing AV node conduction and prolonging the AV node refractory period. Etripamil increases heart rate and decreases systolic blood pressure. Etripamil can be used in the study of paroxysmal supraventricular tachycardia (PSVT).
SCHEME
SIDE CHAIN

MAIN

SYN
US20180110752/ U.S. Patent No. 10,117,848,
EXAMPLES
Example 1: Synthesis methyl 3-(2-((4-cyano-4-(3,4-dimethoxyphenyl)-5-methylhexyl)(methyl)amino)ethyl)benzoate
Part I: Synthesis of 5-Bromo-2-(3,4-dimethoxyphenyl)-2-isopropylpentanenitrile
Part II: Synthesis of methyl 3-(2-(methylamino)ethyl)benzoate
Part III: Reaction of Compound II with Compound III Produced Compound I
| Analysis of the product by mass spectrometry revealed a peak with a mass-to-charge ratio (m/z) of 453, corresponding to the M+H molecular ion of compound I. |
Example 2: Concentrated Solution of Acetate Salt of Compound I
| A concentrated aqueous solution of the acetate salt of compound I is formed according to the following protocol: |
| This protocol readily can be adapted to provide a concentrated solution of the methanesulfonate salt of compound I. |
PRED BY CHIRAL SEPERATION
US20230065401
WO2016165014
EP4119137 chiral sepn done
[0034] In one embodiment the present invention is a kit for treating a cardiac arrhythmia (e.g., PSVT or atrial fibrillation), angina, or a migraine in a subject in need thereof wherein the kit comprises a nasal delivery system comprising two doses of a therapeutically effective amount of compound I having a structure according to the formula:
and instructions for nasally administering to the subject (i) a first dose, and, optionally, (ii) a second dose of an aqueous composition comprising a pharmaceutically acceptable acetate or methanesulfonate salt of compound I, or a racemate or enantiomer thereof, wherein the acetate or methanesulfonate salt of compound I, or the racemate or enantiomer thereof, is dissolved in the aqueous composition at a concentration of 350 mg/mL± 50 mg/mL, and wherein the second dose of the compound is to be administered between 5 minutes and 60 minutes after the first dose.
Cross ref U.S. Patent No. 10,117,848,
[0336]
- 1. A method of treating a cardiac arrhythmia in a subject in need thereof with a therapeutically effective amount of compound I having a structure according to the formula:
the method comprising nasally administering to the subject (i) a first dose, and (ii) a second dose of an aqueous composition comprising a pharmaceutically acceptable acetate or methanesulfonate salt of compound I, or a racemate or enantiomer thereof, wherein the acetate or methanesulfonate salt of compound I, or the racemate or enantiomer thereof, is dissolved in the aqueous composition at a concentration of 350 mg/mL ± 50 mg/mL, and wherein the second dose of the compound is administered between 5 minutes and 25 minutes after the first dose.
PATENT
Journal of the American College of Cardiology (2018), 72(5), 489-497
American Heart Journal (2022), 253, 20-29
Expert Opinion on Investigational Drugs (2020), 29(1), 1-4
EP4119137 WO2016165014
EP-2170050-B1
US-9737503-B2
US-4968717-A
EP-0231003-A2
- [1]. Stambler BS, et al. Etripamil Nasal Spray for Rapid Conversion of Supraventricular Tachycardia to Sinus Rhythm. J Am Coll Cardiol. 2018 Jul 31;72(5):489-497. [Content Brief][2]. Milestone Pharmaceuticals Announces USAN Approval of Generic Name “Etripamil” for its Phase 2 Clinical Development Product for the Treatment of Paroxysmal Supraventricular Tachycardia.[3]. Ascah A, et al. Cardiovascular and Pharmacokinetic Profiles of Intravenous Etripamil in Conscious Telemetered Cynomolgus Monkeys. Int J Toxicol. 2025 Apr 1:10915818251327963. [Content Brief][4]. Pion J, et al. Preclinical Safety Evaluation of Etripamil Nasal Spray in Cynomolgus Macaques (Macaca fascicularis) to Assess for Safety in Patients With Paroxysmal Supraventricular Tachycardia. Int J Toxicol. 2024 Sep-Oct;43(5):503-510. [Content Brief]
//////////ETRIPAMIL, (-)-MSP 2017, MSP 2017
ELUBIOL


ELUBIOL
Dichlorophenyl imidazoldioxolan
CAS 67914-69-6
- Elubiol
- 67914-69-6
- OristaR DCI
- Dichlorophenyl imidazoldioxolan
- (+/-)-Dichlorophenyl imidazoldioxolan
AMY 925
C27H30Cl2N4O5, 561.5 g/mol
ethyl 4-[4-[[(2R,4S)-2-(2,4-dichlorophenyl)-2-(imidazol-1-ylmethyl)-1,3-dioxolan-4-yl]methoxy]phenyl]piperazine-1-carboxylate
Elubiol (Dichlorophenyl imidazoldioxolan) has moderate sebum-inhibiting activity and can be used in the treatment of oily skin or dandruff.
SCHEME

PATENT
DE2804096
https://patentscope.wipo.int/search/en/detail.jsf?docId=DE102084041&_cid=P20-MB323Q-91006-1
PATENT
US4358449
https://patentscope.wipo.int/search/en/detail.jsf?docId=US37288536&_cid=P20-MB3265-92366-1
PATENT
CN102070620
https://patentscope.wipo.int/search/en/detail.jsf?docId=CN84648943&_cid=P20-MB329Q-94515-1
| Example 1 |
| According to the method of the present invention, bacteriostatic ester (±) cis-4-[4-[[2-(2,4-dichlorophenyl)-2-(1-H-imidazolemethyl)-1,3-dioxolane-4-yl]methoxy]phenyl]-1-piperazinecarboxylic acid ethyl ester is prepared, comprising the following steps: |
| 1. Condensation reaction |
| In a dry 500ml three-necked flask, add 473g of dimethyl sulfoxide, 130g of active lipid, 50g of N-(4-hydroxyphenyl)piperazine, and 21g of potassium hydroxide. Control the temperature at 30℃ and keep the reaction for 24 hours. After the reaction, add 520g of purified water. After the addition is completed, cool to 5℃, stir and keep warm for 2h, and filter to obtain the antibacterial ester condensate. The condensation yield is about 85%. |
| 2. Esterification reaction |
| In a three-necked flask, 322g of dichloromethane, 50g of antibacterial ester condensate, and 52g of potassium carbonate were added, and then 11.9g of ethyl chloroformate was slowly added. After the addition was completed, the temperature was controlled at 25°C and the reaction was kept warm for 4 hours. After the reaction was completed, 108g of purified water was slowly added. After the addition was completed, stirring was continued for 2h. The organic layer was washed three times with purified water until the pH reached 7. After washing, dichloromethane was evaporated under reduced pressure. After evaporation, 60ml of methyl isobutyl ketone was added and the temperature was kept at 0-5°C for 2-4h. The antibacterial ester was obtained by suction filtration, and the esterification yield was about 80%. |
| Heat the antibacterial ester and dissolve it in 8 times the amount of acetone, add 0.5 times the amount of activated carbon, reflux and keep warm for 0.5 hours, cool down to no reflux, filter and remove the activated carbon, concentrate the filtrate to 5 times the weight of the antibacterial ester, add water and cool down to 0-5°C after concentration, keep warm for 1-3 hours under stirring, and filter to obtain an off-white crystalline powder. After analysis, the antibacterial ester content is greater than 97%. |
PATENT
CN101665490
https://patentscope.wipo.int/search/en/detail.jsf?docId=CN83857361&_cid=P20-MB32BE-95479-1


| Synthesis of 4-(4-hydroxyphenyl)piperazine: |
| Example 1 |
| In a 1000ml reaction bottle, under nitrogen protection, add 500g of water, 178.5g of dichloroethylamine hydrochloride and 109g of p-hydroxyaniline, heat to 100°C, add 160g of 50% sodium hydroxide solution (80g of sodium hydroxide dissolved in 80g of water), reflux for 10 hours. Then cool to 35°C, add 400g of methanol, adjust the pH value to 8 with ammonia water, filter, and dry the filter cake in vacuum at 40°C to obtain 128g of 4-(4-hydroxyphenyl)piperazine (HPLC content greater than 98%), with a yield of 71.9%. |
| Example 2 |
| In a 1000ml reaction bottle, under nitrogen protection, add 500g of water, 178.5g of dichloroethylamine hydrochloride and 218g of p-hydroxyaniline, heat to 70°C, add 112g of 50% potassium hydroxide solution (56g of potassium hydroxide dissolved in 56g of water), and react for 5 hours. Then cool to 35°C, add 400g of methanol, adjust the pH value to 8 with ammonia water, filter, and dry the filter cake in vacuum at 40°C to obtain 112g of 4-(4-hydroxyphenyl)piperazine (HPLC content greater than 98%), with a yield of 62.9%. |
| Example 3 |
| In a 1000ml reaction bottle, under nitrogen protection, add 500g of water, 312g of dichloroethylamine hydrobromide and 150g of p-hydroxyaniline, stir at room temperature (25°C), add 200g of 50% potassium bicarbonate solution (100g of potassium bicarbonate dissolved in 100g of water), react for 1 hour, then cool to 35°C, add 400g of methanol, adjust the pH value to 8 with ammonia water, filter, and dry the filter cake in vacuum at 40°C to obtain 87g of 4-(4-hydroxyphenyl)piperazine (HPLC content greater than 98%), with a yield of 48.8%. |
| Example 4 |
| In a 1000ml reaction bottle, under nitrogen protection, add 500g of water, 452g of dichloroethylamine hydroiodide and 327g of p-hydroxyaniline, heat to 100°C, add 480g of 50% sodium hydroxide solution (240g of sodium hydroxide dissolved in 240g of water), reflux for 10 hours. Then cool to 35°C, add 600g of methanol, adjust the pH value to 8 with ammonia water, filter, and dry the filter cake in vacuum at 40°C to obtain 154g of 4-(4-hydroxyphenyl)piperazine (HPLC content greater than 98%), with a yield of 86.5%. |
| Synthesis of Ethyl [4-(4-Hydroxyphenyl)]-1-piperazinecarboxylate |
| Example 5 |
| In a 2000 ml reaction bottle, add 178 g of 4-(4-hydroxyphenyl)piperazine, 150 g of sodium bicarbonate and 500 g of acetone, cool to -20°C with ice brine, add 110 g of ethyl chloroformate dropwise, and keep the temperature in the bottle not higher than zero degrees. After the addition is complete, heat to room temperature and react for 5 hours; |
| Add 700g of water, stir for 1 hour and filter. Add the filter cake obtained by filtration to a 1000ml reaction bottle, add 300g of 75% ethanol solution by volume, heat to dissolve, cool to zero degrees with ice brine, filter, and dry the filter cake in vacuum at 40°C to obtain 146g of [4-(4-hydroxyphenyl)]-1-piperazinecarboxylic acid ethyl ester (HPLC content greater than 99%), 58.4%. |
| Example 6 |
| In a 2000ml reaction bottle, add 178g of 4-(4-hydroxyphenyl)piperazine, 180g of sodium carbonate and 500g of acetone, cool to -10°C with ice brine, add 165g of ethyl chloroformate dropwise, and keep the temperature in the bottle not higher than zero degrees. After the addition is complete, heat to 50 degrees and react for 1 hour; |
| Add 700g of water, stir for 1 hour and filter. Add the filter cake obtained by filtration to a 1000ml reaction bottle, add 300g of 75% ethanol solution by volume, heat to dissolve, cool to zero degrees with ice brine, filter, and dry the filter cake in vacuum at 40°C to obtain 156g of [4-(4-hydroxyphenyl)]-1-piperazinecarboxylic acid ethyl ester (HPLC content greater than 99%), 62.4%. |
| Example 7 |
| In a 2000ml reaction bottle, add 178g of 4-(4-hydroxyphenyl)piperazine, 400g of potassium bicarbonate and 1000g of acetone, cool to 0°C with ice brine, add 440g of ethyl chloroformate dropwise, and keep the temperature in the bottle not higher than zero degrees. After the addition is completed, react at about 0°C for 10 hours; |
| Add 1000g of water, stir for 1 hour and filter. Add the filter cake obtained by filtration to a 1000ml reaction bottle, add 500g of 75% ethanol solution by volume, heat to dissolve, cool to zero degrees with ice brine, filter, and dry the filter cake in vacuum at 40°C to obtain 216g of [4-(4-hydroxyphenyl)]-1-piperazinecarboxylic acid ethyl ester (HPLC content greater than 99%), 86.4%. |
| Example 8 |
| In a 2000ml reaction bottle, add 178g of 4-(4-hydroxyphenyl)piperazine, 140g of triethylamine, and 500g of acetone; cool to -10°C with ice brine, add 110g of ethyl chloroformate dropwise, and keep the temperature in the bottle not higher than zero degrees. After the addition is complete, react at -10°C for 10 hours; |
| Add 700g of water, stir for 1 hour and filter. Add the filter cake obtained by filtration to a 1000ml reaction bottle, add 300g of 75% ethanol solution by volume, heat to dissolve, cool to zero degrees with ice brine, filter, and dry the filter cake in vacuum at 40°C to obtain 126g of [4-(4-hydroxyphenyl)]-1-piperazinecarboxylic acid ethyl ester (HPLC content greater than 99%), 50.4%. |
| Synthesis of Ketoconazole Derivatives: |
| Example 9 |
| In a 1000ml reaction bottle, add 45g of cis-[2-(2,4-dichlorophenyl)-2(1H-imidazol-1-yl-methyl)-1,3-dioxopentyl]-4-methyl-p-toluenesulfonate, 25g of ethyl [4-(4-hydroxyphenyl)]-1-piperazinecarboxylate, 5.6g of potassium hydroxide and 180g of dimethyl sulfoxide; react at 25°C for 20 hours. After the reaction, add 450g of ice water to the reaction bottle to reduce the temperature in the reaction bottle to 10°C, and filter; wash the filter cake with water until it is neutral and dry; obtain 42g of crude ketoconazole derivative (HPLC content is 94%). |
| In a 1000ml reaction bottle, add 42g of crude ketoconazole derivative and 350g of ethyl acetate, heat to dissolve, add 0.5g of activated carbon, reflux for half an hour, filter, wash the filter cake with hot ethyl acetate, combine the ethyl acetate, and concentrate to 230g; cool naturally to room temperature, then continue to cool to 0°C with ice water, and keep warm for 1 hour, filter, and vacuum dry to obtain 39g of white powder (HPLC content greater than 99%), with a yield of 73.6%. |
| Example 10 |
| In a 1000 ml reaction bottle, add 45 g of cis-[2-(2,4-dichlorophenyl)-2(1H-imidazol-1-yl-methyl)-1,3-dioxolane]-4-methyl-p-toluenesulfonate, 50 g of ethyl [4-(4-hydroxyphenyl)]-1-piperazinecarboxylate, 11.2 g of sodium hydroxide and 200 g of dioxane; react at 50° C. for 10 hours. After the reaction, add 450 g of ice water to the reaction bottle to reduce the temperature in the reaction bottle to 10° C. and filter; wash the filter cake with water until it is neutral and dry; obtain 41 g of crude ketoconazole derivative (HPLC content is 94%). |
| In a 1000ml reaction bottle, add 41g of crude ketoconazole derivative and 340g of ethyl acetate, heat to dissolve, add 0.5g of activated carbon, reflux for half an hour; filter, wash the filter cake with hot ethyl acetate, combine ethyl acetate, and concentrate to 230g; cool naturally to room temperature, then continue to cool to 0°C with ice water, and keep warm for 1 hour, filter, and vacuum dry to obtain 37g of white powder (HPLC content greater than 99%), with a yield of 69.8%. |
| Embodiment 11 |
| In a 1000ml reaction bottle, add 45g of cis-[2-(2,4-dichlorophenyl)-2(1H-imidazol-1-yl-methyl)-1,3-dioxopentyl]-4-methyl-p-toluenesulfonate, 100g of ethyl [4-(4-hydroxyphenyl)]-1-piperazinecarboxylate, 22.4g of sodium methoxide and 300g of tetrahydrofuran; react at 0°C for 50 hours. After the reaction, add 500g of ice water to the reaction bottle to reduce the temperature in the reaction bottle to 10°C, filter; wash the filter cake with water until neutral and dry; obtain 49g of crude ketoconazole derivative (HPLC content is 94%). |
| In a 1000ml reaction bottle, add 49g of crude ketoconazole derivative and 350g of ethyl acetate, heat to dissolve, add 0.5g of activated carbon, reflux for half an hour; filter, wash the filter cake with hot ethyl acetate, combine ethyl acetate, and concentrate to 250g; cool naturally to room temperature, then continue to cool to 0°C with ice water, and keep warm for 1 hour, filter, and vacuum dry to obtain 43.9g of white powder (HPLC content greater than 99%), with a yield of 82.8%. |
| Example 12 |
| In a 1000ml reaction bottle, add 45g of cis-[2-(2,4-dichlorophenyl)-2(1H-imidazol-1-yl-methyl)-1,3-dioxopentyl]-4-methyl-p-toluenesulfonate, 42g of ethyl [4-(4-hydroxyphenyl)]-1-piperazinecarboxylate, 15g of sodium ethoxide and 300g of N,N-dimethylformamide; react at 10°C for 30 hours. After the reaction, add 500g of ice water to the reaction bottle to reduce the temperature in the reaction bottle to 10°C, and filter; wash the filter cake with water until it is neutral and dry; obtain 51.2g of crude ketoconazole derivative (HPLC content is 94%). |
| In a 1000ml reaction bottle, add 51.2g of crude ketoconazole derivative and 400g of ethyl acetate, heat to dissolve, add 0.5g of activated carbon, reflux for half an hour; filter, wash the filter cake with hot ethyl acetate, combine ethyl acetate, and concentrate to 250g; cool naturally to room temperature, then continue to cool to 0°C with ice water, and keep warm for 1 hour, filter, and vacuum dry to obtain 44.8g of white powder (HPLC content greater than 99%), with a yield of 84.5%. |
REF
[1]. Pierard GE, et al. Modulation of sebum excretion from the follicular reservoir by a dichlorophenyl-imidazoldioxolan. Int J Cosmet Sci. 1996 Oct;18(5):219-27. [Content Brief]
////////////ELUBIOL, AMY 925, Dichlorophenyl imidazoldioxolan, OristaR DCI
Elfucose



Elfucose
Cas 87-96-7
Chemical Formula: C6H12O5
Exact Mass: 164.07
Molecular Weight: 164.157
L-fucopyranose (6-deoxy-L-galactopyranose)
(3S,4R,5S,6S)-6-methyloxane-2,3,4,5-tetrol
- 6-Deoxy-L-galactose (ACI)
- Fucose, L- (8CI)
- (-)-Fucose
- 46: PN: US20220380460 SEQID: 47 claimed sequence
- 6-Desoxygalactose
- L-(-)-Fucose
- L-Fucose
- L-Galactomethylose
- L-Galactopyranose, 6-deoxy-
- CERC 803
- Elfucose
- Fucose
- NSC 1219
- congenital glycosylation disorders
- 6-Deoxy-L-galactopyranose
- L-galactomethylose
- 87-96-7
- Fucose, L-
- 6-deoxy-galactose
Fucose is under investigation in clinical trial NCT03354533 (Study of ORL-1F (L-fucose) in Patients With Leukocyte Adhesion Deficiency Type II).
L-fucopyranose is the pyranose form of L-fucose. It has a role as an Escherichia coli metabolite and a mouse metabolite. It is a L-fucose and a fucopyranose.
SCHEME

PATENT
WO2016150629
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2016150629&_cid=P10-MB1MYE-34318-1
Examples
The invention will now be illustrated in more detail by the following non-limiting examples.
Example 1: Production of L-fucose by biocatalytic oxidation of L-fucitol with galactose oxidase in the presence of peroxidase and catalase
A solution of L-fucitol (6.0 mL aqueous solution containing 600 mg L-fucitol, CAS 13074-06-1, Santa Cruz Biotechnology) was added to a round-bottom three-neck bottle (50 mL), followed by the addition of 1.2 mL K2HPO4 / KH2PO4 ( 1000 mM , pH=7.0) and 0.095 mL catalase (from bovine liver, SIGMA, 21,300 U/mg, 34 mg/mL), 0.120 mL peroxidase (from horseradish, 173 U/mg solid, SIGMA ) and 2.218 mL galactose oxidase (38.4 mg/mL, 2,708 U/mL). The resulting solution was purged with O 2 at room temperature until all L-fucitol was converted to L-fucose. The reaction was monitored by HLPC. The final product was isolated and analyzed by 1 H and 13C NMR. The results are summarized in Table 1.
[Table 1]
Reaction time [h] Conversion [%]
0
3,5 54,0
24 95,8
29 96,0
PATENT
WO2010022244
WO2007021879
////////////Elfucose, 6-Deoxy-L-galactose, Fucose, L- , (-)-Fucose, 6-Desoxygalactose, L-(-)-Fucose, L-Fucose, L-Galactomethylose, L-Galactopyranose, 6-deoxy-, CERC 803, Elfucose, Fucose, NSC 1219, congenital glycosylation disorders, 6-Deoxy-L-galactopyranose, L-galactomethylose, 87-96-7, Fucose, L-, 6-deoxy-galactose
Deupsilocin



Deupsilocin, Psilocin-d10
Psilocin-D10- Deupsilocin
- Psilocine-d10
| Molecular Formula | C12H16N2O |
| Molecular Weight | 214.3299 |
CAS 1435934-64-7
3-[2-[Di(methyl-d3)amino]ethyl-1,1,2,2–d4]-1H-indol-4-ol
3-[2-[bis(trideuteriomethyl)amino]-1,1,2,2-tetradeuterioethyl]-1H-indol-4-ol
| 1H-Indol-4-ol, 3-[2-[di(methyl-d3)amino]ethyl-1,1,2,2-d4]- |
Many mental health disorders, as well as neurological disorders, are impacted by alterations, dysfunction, degeneration, and/or damage to the brain’s serotonergic system, which may explain, in part, common endophenotypes and comorbidities among neuropsychiatric and neurological diseases. Many therapeutic agents that modulate serotonergic function are commercially available, including serotonin reuptake inhibitors, selective serotonin reuptake inhibitors, antidepressants, monoamine oxidase inhibitors, and, while primarily developed for depressive disorders, many of these therapeutics are used across multiple medical indications including, but not limited to, depression in Alzheimer’s disease and other neurodegenerative disease, chronic pain, existential pain, bipolar disorder, obsessive compulsive disorder, anxiety disorders and smoking cessation. However, in many cases, the marketed drugs show limited benefit compared to placebo, can take six weeks to work and for some patients, and are associated with several side effects including trouble sleeping, drowsiness, fatigue, weakness, changes in blood pressure, memory problems, digestive problems, weight gain and sexual problems.
The field of psychedelic neuroscience has witnessed a recent renaissance following decades of restricted research due to their legal status. Psychedelics are one of the oldest classes of psychopharmacological agents known to man and cannot be fully understood without reference to various fields of research, including anthropology, ethnopharmacology, psychiatry, psychology, sociology, and others. Psychedelics (serotonergic hallucinogens) are powerful psychoactive substances that alter perception and mood and affect numerous cognitive processes. They are generally considered physiologically safe and do not lead to dependence or addiction. Their origin predates written history, and they were employed by early cultures in many sociocultural and ritual contexts. After the virtually contemporaneous discovery of (5R,8R)-(+)-lysergic acid-N,N-diethylamide (LSD) and the identification of serotonin in the brain, early research focused intensively on the possibility that LSD and other psychedelics had a serotonergic basis for their action. Today there is a consensus that psychedelics are agonists or partial agonists at brain serotonin 5-hydroxytryptamine 2 A (5-HT2A) receptors, with particular importance on those expressed on apical dendrites of neocortical pyramidal cells in layer V, but also may bind with lower affinity to other receptors such as the sigma-1 receptor. Several useful rodent models have been developed over the years to help unravel the neurochemical correlates of serotonin 5-HT2A receptor activation in the brain, and a variety of imaging techniques have been employed to identify key brain areas that are directly affected by psychedelics.
Psychedelics have both rapid onset and persisting effects long after their acute effects, which includes changes in mood and brain function. Long lasting effects may result from their unique receptor affinities, which affect neurotransmission via neuromodulatory systems that serve to modulate brain activity, i.e., neuroplasticity, and promote cell survival, are neuroprotective, and modulate brain neuroimmune systems. The mechanisms which lead to these long-term neuromodulatory changes are linked to epigenetic modifications, gene expression changes and modulation of pre- and post-synaptic receptor densities. These, previously under-researched, psychedelic drugs may potentially provide the next-generation of neurotherapeutics, where treatment resistant psychiatric and neurological diseases, e.g., depression, post-traumatic stress disorder, dementia and addiction, may become treatable with attenuated pharmacological risk profiles.
Although there is a general perception that psychedelic drugs are dangerous, from a physiologic safety standpoint, they are one of the safest known classes of CNS drugs. They do not cause addiction, and no overdose deaths have occurred after ingestion of typical doses of classical psychotic agents, such as LSD, psilocybin, or mescaline (Scheme 1). Preliminary data show that psychedelic administration in humans results in a unique profile of effects and potential adverse reactions that need to be appropriately addressed to maximize safety. The primary safety concerns are largely psychologic, rather than physiologic, in nature. Somatic effects vary but are relatively insignificant, even at doses that elicit powerful psychologic effects. Psilocybin, when administered in a controlled setting, has frequently been reported to cause transient, delayed headache, with incidence, duration, and severity increased in a dose-related manner [Johnson et al., Drug Alcohol Depend, 2012, 123 (1-3):132-140]. It has been found that repeated administration of psychedelics leads to a very rapid development of tolerance known as tachyphylaxis, a phenomenon believed to be mediated, in part, by 5-HT2A receptors. In fact, several studies have shown that rapid tolerance to psychedelics correlates with downregulation of 5-HT2A receptors. For example, daily LSD administration selectively decreased 5-HT2 receptor density in the rat brain [Buckholtz et al., Eur. J. Pharmacol., 1990, 109:421-425. 1985; Buckholtz et al., Life Sci. 1985, 42:2439-2445].
SCHEME

PATENT
Mindset Pharma Inc., US11591353
https://patentscope.wipo.int/search/en/detail.jsf?docId=US376433397&_cid=P10-MARMO8-36145-1
PATENT
WO2021155470
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021155470&_cid=P10-MARMST-39096-1
PATENT
Cybin IRL Limited, WO2023247665
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2023247665&_cid=P10-MARMVV-41020-1
PATENT
WO2023078604
WO2022195011
| Classic psychedelics and dissociative psychedelics are known to have rapid onset antidepressant and anti-addictive effects, unlike any currently available treatment. Randomized clinical control studies have confirmed antidepressant and anxiolytic effects of classic psychedelics in humans. Ketamine also has well established antidepressant and anti-addictive effects in humans mainly through its action as an NMDA antagonist. Ibogaine has demonstrated potent anti-addictive potential in pre-clinical studies and is in the early stages of clinical trials to determine efficacy in robust human studies [Barsuglia et al., Prog Brain Res, 2018, 242:121-158; Corkery, Prog Brain Res, 2018, 242:217-257]. |
/////////Deupsilocin, Psilocin-d10, KXD3HS8D6X, Psilocin-D10, Deupsilocin, Psilocine-d10
Demannose


Demannose
CAS 530-26-7,
3458-28-4
180.16 g/mol
- D-Mannopyranose
- Carubinose
- Seminose
- mannopyranose
- (3S,4S,5S,6R)-6-(hydroxymethyl)oxane-2,3,4,5-tetrol
- C6H12O6
D-mannopyranose congenital glycosylation disorders
D-mannopyranose is d-Mannose in its six-membered ring form. It has a role as a metabolite. It is a D-aldohexose, a D-mannose and a mannopyranose.
SCHEME

LIT
Tetrahedron Letters (1987), 28(31), 3569-72
///////////Demannose, D-Mannopyranose, Carubinose, Seminose, mannopyranose
Dasminapant




Dasminapant
CAS 1570231-89-8
| Molecular Weight | 1157.40 |
|---|---|
| Formula | C60H72N10O10S2 |
| APG-1387, SM-1387, E53VN70K2X, INN 12430, APG-1387 UNII-E53VN70K2X APG-1387 (SMAC MIMETIC) SMAC-mimetic APG-1387 IAP Inhibitor APG-1387 |
| (5S,5’S,8S,8’S,10aR,10’aR)-3,3′-[1,3-phenylenebis(sulfonyl)]bis{N-(diphenylmethyl)-5-[(2S)-2-(methylamino)propanamido]-6-oxodecahydropyrrolo[1,2-a][1,5]diazocine-8-carboxamide} |
(5S,8S,10aR)-3-[3-[[(5S,8S,10aR)-8-(benzhydrylcarbamoyl)-5-[[(2S)-2-(methylamino)propanoyl]amino]-6-oxo-1,2,4,5,8,9,10,10a-octahydropyrrolo[1,2-a][1,5]diazocin-3-yl]sulfonyl]phenyl]sulfonyl-N-benzhydryl-5-[[(2S)-2-(methylamino)propanoyl]amino]-6-oxo-1,2,4,5,8,9,10,10a-octahydropyrrolo[1,2-a][1,5]diazocine-8-carboxamide
Dasminapant (APG-1387), a bivalent SMAC mimetic and an IAP antagonist, blocks the activity of IAPs family proteins (XIAP, cIAP-1, cIAP-2, and ML-IAP). Dasminapant induces degradation of cIAP-1 and XIAP proteins, as well as caspase-3 activation and PARP cleavage, which leads to apoptosis. Dasminapant can be used for the research of hepatocellular carcinoma, ovarian cancer, and nasopharyngeal carcinoma.
Dasminapant, also known as APG-1387 and SM-1387, is a IAP inhibitor. APG-1387 promotes the rapid degradation of cIAP1/2 and XIAP, and it exerts an antitumor effect on nasopharyngeal carcinoma cancer stem cells. Further studies show that APG-1387 enhances the chemosensitivity and promotes apoptosis in combination with CDDP and 5-FU of NPC in vitro and vivo.
PATENTS
WO2022012671
PATENT
WO2014031487 …
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2014031487&_cid=P11-MAJOJ5-33000-1
PATENT
US20140057924


SCHEME

///////////////Dasminapant, APG-1387, SM-1387, E53VN70K2X, INN 12430, APG 1387, UNII-E53VN70K2X, APG-1387 (SMAC MIMETIC), SMAC-mimetic APG-1387, IAP Inhibitor APG-1387, SM 1387
Civorebrutinib


Civorebrutinib
WS-413, 933NK55FMX
5-amino-3-[4-(5-chloropyridin-2-yl)oxyphenyl]-1-[(6R)-4-cyano-4-azaspiro[2.5]octan-6-yl]pyrazole-4-carboxamide
| Molecular Weight | 463.92 |
|---|---|
| Formula | C23H22ClN7O2 |
| CAS No. | 2155853-43-1 |
Civorebrutinib (WS-413) is a Bruton’s tyrosine kinase inhibitor with antineoplastic effect.
Scheme


Patent
Zhejiang Yukon Pharma Co., Ltd. WO2017198050
WO2019091440
WO2019091438
PATENT
WO2019091441
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019091441&_cid=P10-MADPL7-76599-1
[0116]Preparation of (R)-5-amino-3-(4-((5-chloropyridin-2-yl)oxy)phenyl)-1-(4-cyano-4-azaspiro[2.5]octan-6-yl)-1H-pyrazole-4-carboxamide (Compound 1)
[0119]
[0120]DIPEA (185 g, 1.44 mol, 250 mL, 3 eq) was added to a solution of intermediate compound 11 (167 g, 479 mmol, 1 eq) in EtOH (1670 mL) at 0 ° C. Intermediate compound 17 (187 g, 575 mmol, 1.2 eq) was added to the mixture. The mixture was then stirred at 25 ° C for 12 h under a N2 atmosphere. LCMS (ET14245-55-P1A2, product: RT = 1.723 min) showed that the reaction was complete. The reaction was filtered to obtain the product. The product was used directly in the next step without purification. Intermediate compound 18 (243 g, 407 mmol, yield 85%, purity 93.1%) was obtained as a white solid.
[0122]
[0123]Intermediate compound 18 (121 g, 218 mmol, 1 eq) was stirred in H
2 SO
4 (1200 mL) at 30° C. for 36 h. TLC (DCM: MeOH=10:1, Rf=0.9) showed that compound 18 was completely consumed and only one desired spot was formed (DCM: MeOH=10:1, Rf=0.2). Multiple batches of reaction mixtures were combined, and the combined mixture was poured into MTBE (20 L), solids were precipitated and the filtrate was collected by suction filtration. The pH of the filtrate was adjusted to 10 with aqueous ammonia, extracted with EtOAc (2 L x 10), dried with Na
2 SO
4 , filtered and concentrated under reduced pressure to give intermediate compound 19 (crude product 311 g, equivalent to 238 g product) as a yellow solid.
[0125]
[0126]To a solution of intermediate compound 19 (199 g, 453 mmol, 1 eq) in DMF (1400 mL) was added cesium carbonate (295 g, 907 mmol, 2 eq) and stirred at 15 ° C for 0.5 hours. Then BrCN (52.8 g, 499 mmol, 36.7 mL, 1.1 eq) was added and stirred at 15 ° C for 2 hours. TLC (DCM: MeOH = 10: 1, R
f = 0.2) showed that compound 19 was completely reacted and only one desired spot was generated (DCM: MeOH = 10: 1, R
f = 0.6). Multiple batches of reaction mixtures were combined and the resulting mixture was filtered to remove cesium carbonate. The filtrate was then concentrated under reduced pressure to remove DMF. The residue was diluted with water (2 L) and extracted with ethyl acetate (1 L × 4). The organic phases were combined and washed with water (2 L × 2) and brine (2 L), dried over sodium sulfate, filtered and concentrated under reduced pressure. Acetonitrile (1 L) was added to the residue to precipitate a white solid, which was filtered and the filter cake was washed with acetonitrile (200 mL×2) to give Compound 1 (140 g, 302 mmol, yield 55%, purity 97.0%).
[0127]
1H NMR:CDCl 3400MHzδ8.05(d,J=2.4Hz,1H),7.60(dd,J=2.4,8.8Hz,1H),7.51(d,J=8.8Hz,2H),7.15(d,J=8.8Hz,2H),6.86(d,J=8.8Hz,1H),5.60(s,2H),5.23(br.s.,2H),4.22-4.16(m,1H),3.59-3.41(m,2H),2.39-2.24(m,2H),2.12-2.09(m,1H),1.23-1.10(m,2H),0.80-0.74(m,2H),0.62-0.61(m,1H).
////////Civorebrutinib, WS-413, WS 413, 933NK55FMX
Canlitinib



Canlitinib
Cas 2222730-78-9
| Molecular Weight | 619.61 |
|---|---|
| Formula | C33H31F2N3O7 |
6-[4-[2-fluoro-4-[[1-[(4-fluorophenyl)carbamoyl]cyclopropanecarbonyl]amino]phenoxy]-6-methoxyquinolin-7-yl]oxyhexanoic acid
CANLITINIB is a small molecule drug with a maximum clinical trial phase of II and has 1 investigational indication.
Canlitinib is a tyrosine kinase inhibitor, extracted from patent WO2018072614 (IV-2). Canlitinib has the potential for cancer study.
Kanitinib is a tyrosine kinase inhibitor targeting the oncoprotein c-Met (hepatocyte growth factor receptor; HGFR; MET) and vascular endothelial growth factor receptor 2 (VEGFR2), with potential anti-angiogenic and antineoplastic activities. Upon oral administration, kanitinib targets and binds to c-Met and VEGFR2, thereby disrupting c-Met- and VEGFR2-dependent signal transduction pathways. This may induce cell death in tumor cells overexpressing c-Met and/or VEGFR2 protein. c-Met and VEGFR2 are both overexpressed in many tumor cell types and play key roles in tumor cell proliferation, survival, invasion, metastasis, and tumor angiogenesis
SCHEME

INT

PATENT
WO2020216188
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2020216188&_cid=P20-MA3XXD-35471-1
[0064]The preparation method of compound 1 is shown in Example 9 of compound patent WO 2018/072614 A1. Specifically, the preparation method of compound 1 is as follows.
[0065]
[0066]Under stirring, NaOH (4.4 g, 110 mmol) was added dropwise to a solution of methyl 6-[[4-[2-fluoro-4-[[1-[(4-fluorophenyl)carbamoyl]cyclopropanecarbonyl]amino]phenoxy]-6-methoxy-7-quinolyl]oxy]hexanoate (IV-1, 35.0 g, 55.2 mmol, prepared according to the method described in WO2013/040801A1) in ethanol (350 mL). After the addition was complete, water (50 mL) was added. The resulting mixture was stirred at 20-25°C for 18 h, the reaction solution was diluted with water (100 mL), stirred for 20 min, and the pH was adjusted to 3-4 with 1N HCl. The reaction mixture was concentrated under reduced pressure to distill off about 300 mL of ethanol. The solid product was collected by filtration to give 28.4 g of crude product, which was purified by silica gel column chromatography (eluent: ethyl acetate:methanol = 1:1, v/v) to give 6-[[4-[2-fluoro-4-[[1-[(4-fluorophenyl)carbamoyl]cyclopropanecarbonyl]amino]phenoxy]-6-methoxy-7-quinolyl]oxy]hexanoic acid (Compound 1), 9.6 g (yield: 28.1%).
[0067]Analytical data of compound 1: molecular weight 619.61; NMR hydrogen spectrum is shown in Figure 1, and NMR hydrogen spectrum data are as follows:
[0068]
1H-NMR(δ,DMSO-d6,400MHz):12.03(s,1H,OH),10.40(s,1H,NH),10.02(s,1H,NH),8.47~8.46(d,J=4,1H,CH),7.89-7.92(d,J=12,1H,CH),7.63-7.67(d,J=16,2H,2CH),7.51-7.52(d,J=4,2H2CH),7.39-7.43(t,2H,2CH),7.13-7.17(t,2H,2CH),6.41-6.42(d,J=4,1H,CH),4.12-4.15(t,2H,CH 2),3.95(s,3H,CH 3),2.24-2.28(t,2H,CH 2),1.78-1.85(m,2H,CH 2),1.57-1.64(m,2H,CH 2),1.43-1.51(m,6H,3CH 2)。
PATENT
CN111825609
PATENT
WO2018072614
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018072614&_cid=P20-MA3XZQ-37082-1
[0438]Preparation of 6-[[4-[2-fluoro-4-[[1-[(4-fluorophenyl)carbamoyl]cyclopropanecarbonyl]amino]phenoxy]-6-methoxy-7-quinolyl]oxy]hexanoic acid (IV-2), the reaction formula is as follows:
[0439]
[0440]Under stirring, NaOH (4.4 g, 110 mmol) was added dropwise to a solution of methyl 6-[[4-[2-fluoro-4-[[1-[(4-fluorophenyl)carbamoyl]cyclopropanecarbonyl]amino]phenoxy]-6-methoxy-7-quinolyl]oxy]hexanoate (IV-1, 35.0 g, 55.2 mmol, prepared according to the method described in WO2013/040801A1) in ethanol (350 mL). After the addition was complete, water (50 mL) was added. The resulting mixture was stirred at 20-25°C for 18 h, the reaction solution was diluted with water (100 mL), stirred for 20 min, and the pH was adjusted to 3-4 with 1N HCl. The reaction mixture was concentrated under reduced pressure to distill off about 300 mL of ethanol. The solid product was collected by filtration to give 28.4 g of crude product, which was purified by silica gel column chromatography (eluent: ethyl acetate:methanol = 1:1, v/v) to give 6-[[4-[2-fluoro-4-[[1-[(4-fluorophenyl)carbamoyl]cyclopropanecarbonyl]amino]phenoxy]-6-methoxy-7-quinolyl]oxy]hexanoic acid (IV-2), 9.6 g (yield: 28.1%). Analytical data:
1 H-NMR (400 MHz, DMSO-d
6 ): δ=8.17 (d, J=8.0 Hz, 1H), 7.81 (dd, J=2.8, 13.4 Hz, 1H) 7.62 (m, 2H), 7.51 (m, 4H), 7.39 (t, J=2.4 Hz, 2H), 6.44 (d, J=20.0 Hz, 1H), 4.13 (t, J=8.5 Hz, 2H), 3.85 (s, 3H), 2.27 (t, J=4.0 Hz, 2H), 1.83 (m, 2H), 1.68-1.46 (m, 8H). Mass spectrum (ESI) m/z: 620.2 [M+H]
+ .
PATENT
WO2013/040801
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2013040801&_cid=P20-MA3Y3E-39505-1
BRIGIMADLIN



BRIGIMADLIN
Cas 2095116-40-6
WeightAverage: 591.46
Monoisotopic: 590.1287742
Chemical FormulaC31H25Cl2FN4O3
Spiro[3H-indole-3,2′(1′H)-pyrrolo[2′,3′:4,5]pyrrolo[1,2-b]indazole]-7′-carboxylic acid, 6-chloro-3′-(3-chloro-2-fluorophenyl)-1′-(cyclopropylmethyl)-1,2,3′,3′a,10′,10′a-hexahydro-6′-methyl-2-oxo-, (2′S,3′S,3′aS,10′aS)-
(2′S,3′S,3′aS,10′aS)-6-Chloro-3′-(3-chloro-2-fluorophenyl)-1′-(cyclopropylmethyl)-1,2,3′,3′a,10′,10′a-hexahydro-6′-methyl-2-oxospiro[3H-indole-3,2′(1′H)-pyrrolo[2′,3′:4,5]pyrrolo[1,2-b]indazole]-7′-carboxylic acid (
- (3S,10’S,11’S,14’S)-6-chloro-11′-(3-chloro-2-fluorophenyl)-13′-(cyclopropylmethyl)-6′-methyl-2-oxospiro[1H-indole-3,12′-8,9,13-triazatetracyclo[7.6.0.02,7.010,14]pentadeca-1,3,5,7-tetraene]-5′-carboxylic acid
- (3S,3’S,3a’S,10a’S)-6-chloro-3′-(3-chloro-2-fluorophenyl)-1′-(cyclopropylmethyl)-6′-methyl-2-oxo-1,2,3′,3a’,10′,10a’-hexahydro- 1’H-spiro[indole-3,2′-pyrrolo[2′,3′:4,5]pyrrolo[1,2-b]indazole]-7′- carboxylic acid
- (3S,3’S,3a’S,10a’S)-6-Chloro-3′-(3-chloro-2-fluorophenyl)-1′-(cyclopropylmethyl)-6′-methyl-2-oxo-1,2,3′,3a’,10′,10a’-hexahydro1’H-spiro[indole-3,2′-pyrrolo[2′,3′:4,5]pyrrolo[1,2-b]indazole]-7′-carboxylic acid
- Spiro[3H-indole-3,2′(1’H)-pyrrolo[2′,3′:4,5]pyrrolo[1,2-b]indazole]-7′-carboxylic acid, 6-chloro-3′-(3-chloro-2-fluorophenyl)-1′-(cyclopropylmethyl)-1,2,3′,3’a,10′,10’a-hexahydro-6′-methyl-2-oxo-, (2’S,3’S,3’aS,10’aS)-
Brigimadlin (BI-907828) is a small molecule MDM2–TP53 inhibitor developed for liposarcoma.[2][3][4][5][6]
Brigimadlin is an orally available inhibitor of murine double minute 2 (MDM2), with potential antineoplastic activity. Upon oral administration, brigimadlin binds to MDM2 protein and prevents its binding to the transcriptional activation domain of the tumor suppressor protein p53. By preventing MDM2-p53 interaction, the transcriptional activity of p53 is restored. This leads to p53-mediated induction of tumor cell apoptosis. Compared to currently available MDM2 inhibitors, the pharmacokinetic properties of BI 907828 allow for more optimal dosing and dose schedules that may reduce myelosuppression, an on-target, dose-limiting toxicity for this class of inhibitors.
SCHEME


PATENT
https://patentscope.wipo.int/search/en/detail.jsf?docId=US231206177&_cid=P10-MA0ULZ-04263-1
PATENT
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017060431&_cid=P10-MA0TY5-76812-1




intermediates B-7
Experimental procedure for the synthesis of B-7 a (method E)
To a solution of cyclopropanecarbaldehyde (1.7 mL, 22.7 mmol) in AcOH (19.5 mL) is added intermediate B-6a (1.60 g, 3.8 mmol) and the reaction mixture is stirred for 15 min. Sodium triacetoxyborohydride (1.34 g, 6.3 mmol) is added and the reaction mixture is stirred overnight. Water is added to the reaction mixture and it is extracted with EtOAc. The combined organic layer is dried (MgSO4), filtered, concentrated in vacuo and the crude product B-7a is purified by chromatography if necessary.
Experimental procedure for the synthesis of B-3a (method A)
6-Chloroisatin S-1a (5 g, 27,0 mmol), 1-(3-chloro-2-fluoro-phenyl)-2-nitroethene B-2a (5.5 g, 27.0 mmol) and amino acid B-1a (4.4 g, 27.0 mmol) are refluxed in MeOH for 4 h. The reaction mixture is concentrated in vacuo and purified by crystallization or chromatography if necessary.
Synthesis of compounds (la) according to the invention
Experimental procedure for the synthesis of la-1 (method J)
To a solution of intermediate B-12a (329 mg, 0.65 mmol) in DCM (7 mL) is added a solution of Oxone® (793 mg, 1.29 mmol) in H2O (7 mL) at 0 °C dropwise. The biphasic reaction mixture is stirred vigorously for 20 min at 0 °C and for additional 2 h at rt. The reaction mixture is diluted with H2O and is extracted with DCM. The combined organic layer is dried (MgSO4), filtered, concentrated in vacuo and the crude product is purified by chromatography which gives compound la-1.
Experimental procedure for the synthesis of la-20 (method J + method K)
* The location of overoxidation/N-oxid formation is not entirely clear. B-13a as depicted seems to be probable.
To a solution of intermediate B-12j (417 mg, 0.68 mmol) in DCM (10 mL) is added a solution of Oxone® (841 mg, 1.37 mmol) in H2O (7 mL) at 0 °C dropwise. The biphasic reaction mixture is stirred vigorously for 20 min at 0 °C and for additional 6 h at rt. The reaction mixture is diluted with H2O and extracted with DCM. The combined organic layer is dried (MgSO4), filtered, concentrated in vacuo which gives a crude mixture of la-20 and an oxidized form B-13a (M+H = 621). This mixture is dissolved in MeCN (4.2 mL) and bis(pinacolato)diborone (326 mg, 1.28 mmol) is added. The reaction mixture is heated under microwave irradiation to 100 °C for 30 min. The reaction mixture is diluted with H2O and extracted with DCM. The combined organic layer is dried (MgSO4), filtered, concentrated in vacuo and the crude product is purified by chromatography which gives compound la-20.
References
^ “Brigimadlin”. pubchem.ncbi.nlm.nih.gov.
- ^ Rinnenthal, Joerg; Rudolph, Dorothea; Blake, Sophia; Gollner, Andreas; Wernitznig, Andreas; Weyer-Czernilofsky, Ulrike; Haslinger, Christian; Garin-Chesa, Pilar; Moll, Jürgen; Kraut, Norbert; McConnell, Darryl; Quant, Jens (1 July 2018). “Abstract 4865: BI 907828: A highly potent MDM2 inhibitor with low human dose estimation, designed for high-dose intermittent schedules in the clinic”. Cancer Research. 78 (13_Supplement): 4865. doi:10.1158/1538-7445.AM2018-4865. S2CID 56768874.
- ^ Rudolph, Dorothea; Reschke, Markus; Blake, Sophia; Rinnenthal, Jörg; Wernitznig, Andreas; Weyer-Czernilofsky, Ulrike; Gollner, Andreas; Haslinger, Christian; Garin-Chesa, Pilar; Quant, Jens; McConnell, Darryl B.; Norbert, Kraut; Moll, Jürgen (1 July 2018). “Abstract 4866: BI 907828: A novel, potent MDM2 inhibitor that induces antitumor immunologic memory and acts synergistically with an anti-PD-1 antibody in syngeneic mouse models of cancer”. Cancer Research. 78 (13_Supplement): 4866. doi:10.1158/1538-7445.AM2018-4866. S2CID 80770832.
- ^ Cornillie, J.; Wozniak, A.; Li, H.; Gebreyohannes, Y. K.; Wellens, J.; Hompes, D.; Debiec-Rychter, M.; Sciot, R.; Schöffski, P. (April 2020). “Anti-tumor activity of the MDM2-TP53 inhibitor BI-907828 in dedifferentiated liposarcoma patient-derived xenograft models harboring MDM2 amplification”. Clinical and Translational Oncology. 22 (4): 546–554. doi:10.1007/s12094-019-02158-z. PMID 31201607. S2CID 189862528.
- ^ Schöffski, Patrick; Lahmar, Mehdi; Lucarelli, Anthony; Maki, Robert G (March 2023). “Brightline-1: phase II/III trial of the MDM2–p53 antagonist BI 907828 versus doxorubicin in patients with advanced DDLPS”. Future Oncology. 19 (9): 621–629. doi:10.2217/fon-2022-1291. PMID 36987836. S2CID 257802972.
- ^ Schoeffski, P.; Lorusso, P.; Yamamoto, N.; Lugowska, I.; Moreno Garcia, V.; Lauer, U.; Hu, C.; Jayadeva, G.; Lahmar, M.; Gounder, M. (October 2023). “673P A phase I dose-escalation and expansion study evaluating the safety and efficacy of the MDM2–p53 antagonist brigimadlin (BI 907828) in patients (pts) with solid tumours”. Annals of Oncology. 34: S472 – S473. doi:10.1016/j.annonc.2023.09.1859. S2CID 264392338.
| Names | |
|---|---|
| IUPAC name(3S,10’S,11’S,14’S)-6-chloro-11′-(3-chloro-2-fluorophenyl)-13′-(cyclopropylmethyl)-6′-methyl-2-oxospiro[1H-indole-3,12′-8,9,13-triazatetracyclo[7.6.0.02,7.010,14]pentadeca-1,3,5,7-tetraene]-5′-carboxylic acid | |
| Identifiers | |
| CAS Number | 2095116-40-6 |
| 3D model (JSmol) | Interactive image |
| ChemSpider | 128922236 |
| DrugBank | DB18578 |
| EC Number | 826-645-5 |
| KEGG | D12842 |
| PubChem CID | 129264140 |
| UNII | 9A934ZAN94 |
| showInChI | |
| showSMILES | |
| Properties | |
| Chemical formula | C31H25Cl2FN4O3 |
| Molar mass | 591.46 g·mol−1 |
| Hazards | |
| GHS labelling:[1] | |
| Pictograms | |
| Signal word | Danger |
| Hazard statements | H300, H360Df, H372, H413 |
| Precautionary statements | P203, P260, P264, P270, P273, P280, P301+P316, P318, P319, P321, P330, P405, P501 |
| Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa). | |
/////////////BRIGIMADLIN, BI-907828, BI 907828, 9A934ZAN94
Bocodepsin



Bocodepsin, OKI-179
CAS 1834513-65-3
S-((3E)-4-((6S,9S)-12,12-DIMETHYL-4,8,11,14-TETRAOXO-9-(PROPAN-2-YL)-7-OXA -3,10,13-TRIAZA-1(2,4)-(1,3)THIAZOLACYCLOTETRADECAPHAN-6-YL)BUT-3-EN-1-YL) (2S)-2-AMINO-3-METHYLBUTANETHIOATE
S-(4-((7S,10S)-4,4-DIMETHYL-2,5,8,12-TETRAOXO-7-(PROPAN-2-YL)-9-OXA-16-THIA- 3,6,13,18-TETRAAZABICYCLO(13.2.1)OCTADECA-15(18),17-DIEN-10-YL)BUT-3-EN-1-YL) (2S)-2-AMINO-3-METHYLBUTANETHIOATE
| Molecular Weight | 581.75 |
|---|---|
| Formula | C26H39N5O6S2 |
- L-Valine, S-L-valyl-(3S,4E)-3-hydroxy-7-mercapto-4-heptenonyl-2-(aminomethyl)-4-thiazolecarbonyl-2-methylalanyl-, (5–>2)-lactone
- S-{(3E)-4-[(6S,9S)-12,12-dimethyl-4,8,11,14-tetraoxo-9-(propan-2-yl)-7-oxa-3,10,13-triaza-1(2,4)-[1,3]thiazolacyclotetradecaphan-6-yl]but-3-en-1-yl} (2S)-2-amino-3-methylbutanethioate
- S-{4-[(7S,10S)-4,4-dimethyl-2,5,8,12-tetraoxo-7-(propan-2-yl)-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-15(18),17-dien-10-yl]but-3-en-1-yl} (2S)-2-amino-3-methylbutanethioate
- Originator OnKure Therapeutics
- Class Antineoplastics; Small molecules
- Mechanism of ActionHDAC1 protein inhibitors
Phase I/II Malignant melanoma; Solid tumours
- No development reportedHaematological malignancies
29 Jan 2025 OnKure Therapeutics completes the phase-I/II Nautilus trial in Malignant melanoma (Late-stage disease, Metastatic disease, Second-line therapy or greater, Combination therapy) in USA (PO) (NCT05340621),
- 11 Oct 2023Pharmacodynamics data from a preclinical studies in Solid tumours presented at the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics 2023 (AACR-NCI-EORTC-2023 2023)
- 11 Oct 2023Initial efficacy and adverse events data from a phase Ib/II NAUTILUS trial in Melanoma presented at the International Conference on Molecular Targets and Cancer Therapeutics 2023 (AACR-NCI-EORTC-2023)
Bocodepsin (OKI-179) is an orally active and selective HDAC inhibitor, with antitumor activity. Bocodepsin can be used for suppression on solid tumor and hematologic malignancies.
SCHEME


PATENT
WO2017201278
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017201278&_cid=P12-M9WKU5-87067-1

Examples
[00127] The following examples are provided for illustrative purposes only and are not intended to limit the scope of the invention.
[00128] Example 1: Preparation of (R)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12- tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10- yl)but-3-en-l-yl) 2-amino-3-methylbutanethioate hydrochloride.
Step 1 : Preparation of (R)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12- tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10- yl)but-3-en-l-yl) 2-((tert-butoxycarbonyl)amino)-3-methylbutanethioate. (7S,10S)-10-((E)- 4- chlorobut-l-en-l-yl)-7-isopropyl-4,4-dimethyl-9-oxa-16-thia-3,6,13,18- tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-diene-2,5,8,12-tetraone (15 g, 0.03 mol), (R)-2- ((tert-butoxycarbonyl)amino)-3-methylbutanethioic S-acid (12.5 g, 0.06 mol), K2CO3 (11.2 g, 0.09 mol), and KI (0.89 g, 0.006 mol) were dissolved in 150mL of acetonitrile and the resulting mixture was warmed to 60-65°C and stirred under nitrogen. After 16 hours, the mixture was cooled to 20°C, 300 mL of water was added, and the resulting suspension was extracted with ethyl acetate (2X200 mL). The combined organic phases were dried with anhydrous sodium sulfate, filtered and concentrated. The residue was purified by flash column chromatography (elution with ethyl acetate/petroleum ether = 1/1 to 4/1) to give (R)- 5- ((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18- tetraazabicyclo[13.2.1] octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-((tert- butoxycarbonyl)amino)-3-methylbutanethioate (17.0 g, 80% yield).
Step 2: Preparation of (R)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12- tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10- yl)but-3-en-l-yl) 2-amino-3-methylbutanethioate hydrochloride. (R)-S-((E)-4-((7S,10S)-7- isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18- tetraazabicyclo[l 3.2.1] octadeca- 1 ( 17), 15 (18)-dien- 10-y l)but-3-en- 1 -y 1) 2-((tert-butoxy carbony l)amino)-3-methylbutanethioate (1.7 g, 0.025 mol) was dissolved in 150 mL of dichloromethane and trifluoroacetic acid (22.5 mL) was added at 10°C. After stirring at 10°C for 4 hours under nitrogen, the mixture was concentrated to dryness and the residue was dissolved in 100 mL of ethyl acetate and treated with 10 mL of 4M HCl/ethyl acetate solution. The mixture was then treated with petroleum ether (100 mL) and the resulting white solid was collected by filtration and dried to give (R)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18- tetraazabicyclo[13.2.1] octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-amino-3-methylbutanethioate hydrochloride (0.40 g, 26% yield). Mass Spec(m/z): 582.8 (M+l).
129] Example 2: Preparation of (S)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-amino-3-methylbutanethioate hydrochloride.
Step 1 : (S)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-((tert-butoxy carbony l)amino)-3 -methy lbutanethioate. (7S,10S)-10-((E)-4-chlorobut- 1 -en- 1 -yl)-7-isopropyl-4,4-dimethyl-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-diene-2,5,8,12-tetraone (40 g, 0.0825 mol), (S)-2-((tert-butoxycarbonyl)amino)-3-methylbutanethioic S-acid (38.5 g, 0.165 mol), K2C03 (34.1 g, 0.247 mol), and KI (2.7 g, 0.0163 mol) were dissolved in 400 mL of acetonitrile and stirred at 60-65°C under nitrogen for 20 hours. The mixture was cooled to 20°C, water (300 mL) was added and the resulting suspension was extracted with ethyl acetate (2X200 mL). The organic phases were combined, dried with anhydrous sodium sulfate, filtered and concentrated. The residue was purified by flash column chromatography (elution with ethyl acetate/petroleum ether = 1/1 to 4/1) to give (S)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-((tert-butoxycarbonyl)amino)-3-methylbutanethioate (49.8 g, 89% yield).
Step 2: (S)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-amino-3-methylbutanethioate hydrochloride. (S)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-1 ( 17), 15( 18)-dien- 10-y l)but-3-en- 1 -y 1) 2-((tert-butoxy carbony l)amino)-3 -methylbutanethioate (47.8 g, 0.07 mol) was dissolved in dichloromethane (400 mL) and trifluoroacetic acid (65 mL) was added dropwise at 10 to 20°C while stirring under nitrogen. After the addition, the mixture was stirred at 15 to 20°C for 3 hours at which time an additional aliquot of trifluoroacetic acid (20 mL) was added and stirring at 15 to 20°C was continued for an additional 1.5 hours. The solution was then concentrated under vacuum to near dryness and the residue dissolved in ethyl acetate (250 mL). 20 mL of 4M HCl/ethyl acetate solution was then added while stirring at a temperature between 10 to 15°C resulting in the formation of a slurry. 250 mL n-heptane was then added and the solids were filtered, rinsed with n-heptane and dried in vacuo to give (S)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18- tetraazabicyclo[13.2.1] octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-amino-3 -methylbutanethioate hydrochloride as a white solid which contained some residual heptane. (49.0 g, 100% yield). Mass Spec(m/z): 582.8 (M+l)
130] Example 3: Preparation of (S)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-amino-3 -methylbutanethioate benzenesulfonate.
The product of Example 2, step 1 ((S)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-((tert-butoxycarbonyl)amino)-3-methylbutanethioate) (1 eq.) was dissolved in acetonitrile (10 vol) at 20-25°C and the mixture was treated with
benzenesulfonic acid (3 eq.). After stirring at room temperature for 5 hours, the solvent was removed by decanting, the residual oil was treated with THF (5vol), and the resulting mixture was stirred over night at room temperature. The resulting white solid was collected by filtration and dried in vacuo to give (S)-S-((E)-4-((7S,10S)-7-isopropyl-4,4-dimethyl-2,5,8,12-tetraoxo-9-oxa-16-thia-3,6,13,18-tetraazabicyclo[13.2.1]octadeca-l(17),15(18)-dien-10-yl)but-3-en-l-yl) 2-amino-3-methylbutanethioate benzenesulfonate (90% yield; 98% purity). 1HNMR (d6-DMSO) δ: 0.56 to 0.57 (m, 3H), 0.76 to 0.78 (m, 3H), 0.92 to 0.94 (m, 3H), 0.96 to 0.98 (m, 3H), 1.45 to 1.48 (m, 3H), 1.70 to 1.72 (m, 3H), 2.07 to 2.16 (m, 2H), 2.27 to 2.28 (m, 2H), 2.93 to 2.95 (m, 1H), 2.94 to 2.95 (m, 1H), 2.97 to 3.1 (m, 1H), 4.13 to 4.15 (m, 1H), 4.28 to 4.33 (1H), 4.92 to 5.0 (m, 1H), 5.61 to 5.64 (m, 3H), 7.29 to 7.32 (m, 3H), 7.57 to 7.60 (m, 2H), 7.88 to 7.92 (m, 1H), 8.17 (s, 1H), 8.32 (s, 3H), 8.48 to 8.50 (m, 1H).
[1]. Diamond JR, et al. Preclinical Development of the Class-I-Selective Histone Deacetylase Inhibitor OKI-179 for the Treatment of Solid Tumors. Mol Cancer Ther. 2022 Mar 1;21(3):397-406. [Content Brief]
///////////Bocodepsin, K5D067O1SW, OKI-179, Malignant melanoma, Solid tumours, OnKure Therapeutics, OKI 006
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO
.....










