New Drug Approvals

Home » Articles posted by DR ANTHONY MELVIN CRASTO Ph.D (Page 45)

Author Archives: DR ANTHONY MELVIN CRASTO Ph.D

DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE

Blog Stats

  • 4,802,346 hits

Flag and hits

Flag Counter

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

Join 37.9K other subscribers
Follow New Drug Approvals on WordPress.com

Archives

Categories

Recent Posts

Flag Counter

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

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

Join 37.9K other subscribers
DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his Ph.D from ICT, 1991,Matunga, Mumbai, India, in Organic Chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with AFRICURE PHARMA, ROW2TECH, NIPER-G, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India as ADVISOR, earlier assignment was with GLENMARK LIFE SCIENCES LTD, as CONSUlTANT, Retired from GLENMARK in Jan2022 Research Centre as Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Total Industry exp 32 plus yrs, Prior to joining Glenmark, he has worked with major multinationals like Hoechst Marion Roussel, now Sanofi, Searle India Ltd, now RPG lifesciences, etc. He has worked with notable scientists like Dr K Nagarajan, Dr Ralph Stapel, Prof S Seshadri, etc, He did custom synthesis for major multinationals in his career like BASF, Novartis, Sanofi, etc., He has worked in Discovery, Natural products, Bulk drugs, Generics, Intermediates, Fine chemicals, Neutraceuticals, GMP, Scaleups, etc, he is now helping millions, has 9 million plus hits on Google on all Organic chemistry websites. His friends call him Open superstar worlddrugtracker. His New Drug Approvals, Green Chemistry International, All about drugs, Eurekamoments, Organic spectroscopy international, etc in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 32 PLUS year tenure till date Feb 2023, Around 35 plus products in his career. He has good knowledge of IPM, GMP, Regulatory aspects, he has several International patents published worldwide . He has good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, Polymorphism etc., He suffered a paralytic stroke/ Acute Transverse mylitis in Dec 2007 and is 90 %Paralysed, He is bound to a wheelchair, this seems to have injected feul in him to help chemists all around the world, he is more active than before and is pushing boundaries, He has 100 million plus hits on Google, 2.5 lakh plus connections on all networking sites, 100 Lakh plus views on dozen plus blogs, 227 countries, 7 continents, He makes himself available to all, contact him on +91 9323115463, email amcrasto@gmail.com, Twitter, @amcrasto , He lives and will die for his family, 90% paralysis cannot kill his soul., Notably he has 38 lakh plus views on New Drug Approvals Blog in 227 countries......https://newdrugapprovals.wordpress.com/ , He appreciates the help he gets from one and all, Friends, Family, Glenmark, Readers, Wellwishers, Doctors, Drug authorities, His Contacts, Physiotherapist, etc He has total of 32 International and Indian awards

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

Divalproex sodium


Divalproex sodium.png
Divalproex Sodium | ≥99%(HPLC) | Selleck | HDAC inhibitor

 Divalproex

  • 44089

WeightAverage: 144.2114
Chemical FormulaC8H16O2

UNII614OI1Z5WI, CAS number99-66-1, 76584-70-8

2-propylpentanoic acid, DIVALPROEX SODIUM76584-70-8Valproate semisodiumEpivalDepakoteSodium divalproateSemisodium ValproateAbbott 50711ValdisovalValproic Acid 
CAS Registry Number: 99-66-1 
CAS Name: 2-Propylpentanoic acid 
Additional Names: 2-propylvaleric acid; di-n-propylacetic acid 
Trademarks: Convulex (Pharmacia); Depakene (Abbott) 
Molecular Formula: C8H16O2 
Molecular Weight: 144.21 
Percent Composition: C 66.63%, H 11.18%, O 22.19% 
Literature References: Antiepileptic; increases levels of g-aminobutyric acid (GABA) in the brain. Prepn: B. S. Burton, Am. Chem. J.3, 385 (1882); E. Oberreit, Ber.29, 1998 (1896); M. Tiffeneau, Y. Deux, Compte Rend.212, 105 (1941). Anticonvulsant activity: H. Meunier et al.,Therapie18, 435 (1963). Toxicity data: Jenner et al.,Food Cosmet. Toxicol.2, 327 (1964). Comprehensive description: Z. L. Chang, Anal. Profiles Drug Subs.8, 529-556 (1979). Review of teratogenicity studies: H. Nau et al.,Pharmacol. Toxicol.69, 310-321 (1991); R. Alsdorf, D. F. Wyszynski, Expert Opin. Drug Safety4, 345-353 (2005). Review of pharmacology and clinical experience in epilepsy: E. M. Rimmer, A. Richens, Pharmacotherapy5, 171-184 (1985); in psychiatric disease: D. R. P. Guay, ibid.15, 631-647 (1995); in migraine prophylaxis: C. E. Shelton, J. F. Connelly, Ann. Pharmacother.30, 865-866 (1996). Review of pharmacodynamics and mechanisms of action: W. Löscher, Prog. Neurobiol.58, 31-59 (1999). 
Properties: Colorless liquid with characteristic odor. bp 219.5°. nD24.5 1.425. d40 0.9215. pKa 4.6. Very sol in organic solvents. Soly in water: 1.3 mg/ml. LD50 orally in rats: 670 mg/kg (Jenner). 
Boiling point: bp 219.5° 
pKa: pKa 4.6 
Index of refraction:nD24.5 1.425 
Density: d40 0.9215 
Toxicity data: LD50 orally in rats: 670 mg/kg (Jenner) 
Derivative Type: Sodium salt (1:1) 
CAS Registry Number: 1069-66-5 
Additional Names: Sodium valproate 
Trademarks: Depacon (Abbott); Depakin (Sanofi-Synthelabo); Dépakine (Sanofi-Aventis); Epilim (Sanofi-Aventis); Ergenyl (Sanofi-Synthelabo); Leptilan (Dolorgiet); Orfiril (Desitin) 
Molecular Formula: C8H15NaO2 
Molecular Weight: 166.19 
Percent Composition: C 57.82%, H 9.10%, Na 13.83%, O 19.25% 
Properties: White, odorless, crystalline, deliquescent powder. pKa 4.8. Hygroscopic. One gram is sol in 0.4 ml water; 1.5 ml ethanol; 5 ml methanol. Practically insol in common organic solvents. LD50 orally in mice: 1700 mg/kg (Meunier). 
pKa: pKa 4.8 
Toxicity data: LD50 orally in mice: 1700 mg/kg (Meunier) 
Derivative Type: Sodium salt (2:1) 
CAS Registry Number: 76584-70-8 
Additional Names: Sodium hydrogen bis(2-propylpentanoate); divalproex sodium; valproate semisodium 
Manufacturers’ Codes: Abbott 50711 
Trademarks: Depakote (Abbott); Valcote (Abbott) 
Molecular Formula: C16H31NaO4 
Molecular Weight: 310.40 
Percent Composition: C 61.91%, H 10.07%, Na 7.41%, O 20.62% 
Derivative Type: Magnesium salt 
Trademarks: Depamag (Sigma-Tau) 
Molecular Formula: C16H30MgO4 
Molecular Weight: 310.71 
Percent Composition: C 61.85%, H 9.73%, Mg 7.82%, O 20.60% 
Therap-Cat: Anticonvulsant; antimanic; antimigraine.Keywords: Anticonvulsant; Antimigraine; Antimanic.

Synthesis Reference

Daniel Aubert, Francis Blanc, Henri Desmolin, Michel Morre, Lucette Sindely, “Valproic acid preparations.” U.S. Patent US5017613, issued January, 1965.

US5017613

Patent

https://patents.google.com/patent/WO2007004238A2/enDivalproex sodium is one of the most widely used epileptic agents presently available in the market. Both the constituents, valproic acid and sodium valproate themselves have also been used for the treatment of epileptic seizures and convulsions. But their utility has remained restricted since valproic acid is a liquid and is difficult to formulate for an oral dosage form whereas sodium valproate is a hygroscopic solid with poor stability characteristics. Divalproex sodium is an oligomer having 1:1 molar ratio of valproic acid and sodium valproate containing 4 to 6 units. The relevant prior art includes US 4,988,731 (’73I) relates to a non-hygroscopic stable sodium hydrogen divalproate oligomer. Its probable structure is shown in Fig 1

Figure imgf000002_0001

Fig 1 – Divalproex sodiumWhere M is a about 2.As can be seen from the displayed structure, one mole each of the valproic acid forms coordinate bonds with the sodium of the sodium valproate molecule, and the valproate ion is ionically bonded to the sodium atom. The structure is thus consistent with the unique characteristic of the compound. However the preferred mode of representing Divalproex sodium is by reference to single compound of the formula{(CH3CH2CH2)2CHCO2} {(CH3CH2CH2)2CHCO2}Na, HThe said patent also describes two alternative processes for the preparation of the oligomer. According to one aspect, the oligomer is produced by dissolving sodium valproate and valproic acid in equimolar amount in acetone and crystallizing from chilled acetone at around O0C. Alternatively Divalproex sodium can be isolated from a two component liquid medium, which includes acetone where in half equivalent of NaOH to the valproic acid present, preferable as a solution in an acetone miscible solvent eg. water. The new compound can be recovered from the liquid phase by evaporating the solvent(s) and, if desired, the new compound can be recrystallized, for instance from acetonitrile or others or the material may be spay-dried, lyophilized or purified by chromatography.US ‘731 claims yield of 90% of theory.Drawbacks of the above mentioned reported methods for the preparation of Divalproex sodium described in US 4988731 are difficult to reproduce on a large scale and provides inconsistent yields and the material obtained is not always free flowing in nature. The process involves the crystallization of a 1:1 mixture of valproic acid and sodium valproate from a chilled solution of acetone, followed by washing with chilled acetone. Divalproex sodium is as such fairly soluble in acetone at temperatures above 1O0C and extreme care has to be. taken while performing washing with chilled acetone as any rise in temperature would lead to the loss of yield. This problem actually comes to the fore while scaling up the process during commercialization since during centrifugation of the large volume the temperature of the mixture rises and acetone has to be cooled below O0C, which require large amount of liquid nitrogen or dry ice. Moreover it was also observed that due to the cooled nature of the solvent, the isolated Divalproex sodium absorbs considerable amount of moisture and therefore requires longer time to dry eventually leading to longer time cycle for the otherwise simple single step process. Also the high moisture content in the recovered acetone makes it unsuitable for reuse. Alternatively, to avoid absorption of water, the centrifugation had to be carried out under a blanket of dry nitrogen gas. These additional infrastructural loads add to input costs eventually making the otherwise single step low cost process becoming uncompetitive and economically unviable.Similarly the other process involves the addition of half molar equivalent of sodium hydroxide dissolved in water to valproic acid and the solvent has to be evaporated to obtain crude product, which has to be recrystallized to get Divalprox of the desired specification. The process is operationally tedious and requires the reduction in the level of water in the reaction mass via evaporation of the solvent followed by re- crystallization from acetonitrile making the process lengthy and economically unviable. There is therefore a need for operationally making this single step process more efficient and high yieldingExample I:To lOOg of Valproic acid with stirring at 20-300C, powdered NaOH ( 13g; half molar) is added & the resulting reaction mixture is stirred at 40-500C for 1 hr. Then acetonitrile(600ml) is added to obtain clear solution at 40-500C and the solution is charcoalized at 40-500C followed by filtration at 40-500C through hyflo-bed. The resultant reaction mixture was stirred at 10-200C for 2-3 hr. The solid , thus obtained, was filtered and product was dried at 40-450C for 10-12 hr. (102.25g, 95%)Example II;To lOOg of Valproic acid with stirring at 20-300C, powdered NaOH (13g; half molar) is added & the resulting reaction mixture is stirred at 30-400C for 1 hr. Then acetone (600ml) is added to obtain clear solution at 30-400C and the material is charcoalized at 30-400C followed by filtration through hyflo-bed. The resultant reaction solution was stirred at -5°C to -1O0C for 2-3 hr. The solid , thus obtained, was filtered and product was dried at 40-450C for 10-12 hr. ( 55g, 51.11%) Example III:To a solution of Valproic acid (10Og) in dichloromethane (200ml) at 20-300C, powdered caustic (13g ; half molar) is added & the reaction mixture is stirred at 30- 400C for 1 hr to get clear solution. Then acetonitrile (600ml) is added to it inorder to crystallize the product. The solid, thus obtained, is further stirred at 0-50C for 2-3 hr followed by filtration. The product was dried at 40-450C for 10-12 hr. (10Og; 93%)Example IV:To a solution of Valproic acid (10Og) in diisopropyl ether(200ml) at 20-300C, powdered caustic (13g ; half molar) is added & the reaction mixture is stirred at 40-500C for 1 hr to get clear solution. Then acetonitrile (800ml) is added to it inorder to crystallize the product. The solid, thus obtained, is further stirred at 0-50C for 2-3 hr followed by filtration. The product was dried at 40-450C for 10-12 hr. (104g; 96.65%)Example V:To a solution of Valproic acid (10Og) in methyl tertiary butyl ether(200ml) at 20- 300C, powdered caustic (13g ; half molar) is added & the reaction mixture is stirred at 40-500C for 1 hr to get clear solution. Then acetonitrile (800ml) is added to it inorder to crystallize the product. The solid, thus obtained, is further stirred at 0-50C for 2-3 hr followed by filtration. The product was dried at 40-450C for 10-12 hr. (102g;94.79%)Example VI:To a solution of Valproic acid (10Og) in toluene (200ml) at 20-300C, powdered caustic (13g ; half molar) is added & the reaction mixture is stirred at 40-500C for 1 hr to get clear solution. Then acetonitrile (800ml) is added to it inorder to crystallize the product. The solid, thus obtained, is further stirred at 0-50C for 2-3 hr followed by filtration. The product was dried at 40-450C for 10-12 hr. (101g; 93.87%)Example VII: A mixture of sodium valproate (6Og) and valproic acid (52.04g) was taken in acetonitrile (800ml) and heated at reflux to obtain a clear solution, which was filtered through hyflo-bed to remove suspended particles. Then the solution was stirred at 10- 200C for 2-3 hr. The solid, thus obtained, was filtered and washed with acetonitrile (100ml). The product was dried at 40-450C for 10-12 hr. (105g ; 93.75%)Example VIII;To a solution of valproic acid (10Og) in methanol (200ml) at 20-300C5 caustic (13g; half molar) is added & the reaction mixture is stirred at 20-300C for 1 hr. Then the methanol was recovered at reduced pressure and acetonitrile (600ml) is added to it with stirring. The reaction mixture was further stirred at 0-50C for 2-3 hr. The solid, thus obtained, is filtered, washed with acetonitrile (100ml) and product was dried at 40-45°C for 10-12 hr.(102g; ~ 95%) Example IX:To a solution of valproic acid (10Og) in methanol (200ml) at 20-300C, caustic (13g; half molar) is added & the reaction mixture is stirred at 20-300C for 1 hr. Then the methanol was recovered at reduced pressure and acetone (600ml) is added to it with stirring. The reaction mixture was further stirred at -5°C to -1O0C for 2-3 hr. The solid, thus obtained, is filtered, washed with chilled acetone (100ml) and product was dried at 40-450C for 10-12 hr.(54g; ~ 50.11%)Example X:To a solution of valproic acid (10Og) in ethanol (200ml) at 20-300C, caustic (13g; half molar) is added & the reaction mixture is stirred at 20-300C for 1 hr. Then the ethanol was recovered at reduced pressure and acetonitrile (600ml) is added to it with stirring.The reaction mixture was further stirred at 0-50C for 2-3 hr. The solid, thus obtained, is filtered, washed with acetonitrile (100ml) and product was dried at 40-450C for 10-12 hr.(101g; ~ 93.87%)Example XI: To a solution of valproic acid (10Og) in ethanol (200ml) at 20-30°C, caustic (13g; half molar) is added & the reaction mixture is stirred at 20-300C for 1 hr. Then the ethanol was recovered at reduced pressure and acetone (600ml) is added to it with stirring. The reaction mixture was further stirred at -5°C to -100C for 2-3 hr. The solid, thus obtained, is filtered, washed with chilled acetone (100ml) and product was dried at 40-450C for 10-12 hr.(55g; ~ 51%)ADVANTAGES:> The process is high yielding. > The process produces Divalproex sodium with improved flowability.> The process produces Divalproex sodium that is non-hygroscopic and more stable.> The process is industrially feasible, precise, reproducible and does not require sophisticated infrastructure.

Divalproex Sodium is a stable coordination compound comprised of sodium valproate and valproic acid with anticonvulsant and antiepileptic activities. Divalproex dissociates to the valproate ion in the gastrointestinal tract. This agent binds to and inhibits gamma-aminobutyric acid (GABA) transaminase and its anticonvulsant activity may be exerted by increasing brain concentration of GABA and by inhibiting enzymes that catabolize GABA or block the reuptake of GABA into glia and nerve endings. Divalproex may also work by suppressing repetitive neuronal firing through inhibition of voltage-sensitive sodium channels.

Valproate semisodium is a mixture of valproic acid and its sodium salt in a 1:1 molar ratio. It is used for the management and treatment of seizure disorders, mania, and prophylactic treatment of migraine headache. It has a role as an antimanic drug, an anticonvulsant and a GABA agent. It contains a valproic acid and a sodium valproate.

Divalproex sodium, valproate sodium, and valproic acid, are all similar medications that are used by the body as valproic acid. Therefore, the term valproic acid will be used to represent all of these medications in this discussion.

Valproate (VPA) and its valproic acidsodium valproate, and valproate semisodium forms are medications primarily used to treat epilepsy and bipolar disorder and prevent migraine headaches.[2] They are useful for the prevention of seizures in those with absence seizurespartial seizures, and generalized seizures.[2] They can be given intravenously or by mouth, and the tablet forms exist in both long- and short-acting formulations.[2]

Common side effects of valproate include nausea, vomiting, sleepiness, and dry mouth.[2] Serious side effects can include liver failure, and regular monitoring of liver function tests is therefore recommended.[2] Other serious risks include pancreatitis and an increased suicide risk.[2] Valproate is known to cause serious abnormalities in babies if taken during pregnancy,[2][3] and as such it is not typically recommended for women of childbearing age who have migraines.[2]

Valproate’s precise mechanism of action is unclear.[2][4] Proposed mechanisms include affecting GABA levels, blocking voltage-gated sodium channels, and inhibiting histone deacetylases.[5][6] Valproic acid is a branched short-chain fatty acid (SCFA) made from valeric acid.[5]

Valproate was first made in 1881 and came into medical use in 1962.[7] It is on the World Health Organization’s List of Essential Medicines[8] and is available as a generic medication.[2] It is marketed under the brand names Depakote, among others.[2] In 2018, it was the 131st most commonly prescribed medication in the United States, with more than 5 million prescriptions.[9][10]

Terminology

Valproic acid (VPA) is an organic weak acid. The conjugate base is valproate. The sodium salt of the acid is sodium valproate and a coordination complex of the two is known as valproate semisodium.[11]

Medical uses

It is used primarily to treat epilepsy and bipolar disorder. It is also used to prevent migraine headaches.[12]

Epilepsy

Valproate has a broad spectrum of anticonvulsant activity, although it is primarily used as a first-line treatment for tonic–clonic seizuresabsence seizures and myoclonic seizures and as a second-line treatment for partial seizures and infantile spasms.[12][13] It has also been successfully given intravenously to treat status epilepticus.[14][15]

Mental illness

Bipolar disorder

Valproate products are also used to treat manic or mixed episodes of bipolar disorder.[16][17]

Schizophrenia

A 2016 systematic review compared the efficacy of valproate as an add-on for people with schizophrenia:[18]

There is limited evidence that adding valproate to antipsychotics may be effective for overall response and also for specific symptoms, especially in terms of excitement and aggression. Valproate was associated with a number of adverse events among which sedation and dizziness appeared more frequently than in the control groups.[18]
showOutcomeFindings in wordsFindings in numbersQuality of evidence

Dopamine dysregulation syndrome

Based upon five case reports, valproic acid may have efficacy in controlling the symptoms of the dopamine dysregulation syndrome that arise from the treatment of Parkinson’s disease with levodopa.[19][20][21]

Migraines

Valproate is also used to prevent migraine headaches. Because this medication can be potentially harmful to the fetus, valproate should be considered for those able to become pregnant only after the risks have been discussed.[22]

Other

The medication has been tested in the treatment of AIDS and cancer, owing to its histone-deacetylase-inhibiting effects.[23]

Contraindications

Contraindications include:

Adverse effects

See also: List of adverse effects of valproic acid and List of adverse effects of valproate semisodium

Most common adverse effects include:[22]

Serious adverse effects include:[22]

Valproic acid has a black box warning for hepatotoxicitypancreatitis, and fetal abnormalities.[22]

There is evidence that valproic acid may cause premature growth plate ossification in children and adolescents, resulting in decreased height.[26][27][28][29] Valproic acid can also cause mydriasis, a dilation of the pupils.[30] There is evidence that shows valproic acid may increase the chance of polycystic ovary syndrome (PCOS) in women with epilepsy or bipolar disorder. Studies have shown this risk of PCOS is higher in women with epilepsy compared to those with bipolar disorder.[31] Weight gain is also possible.[32]

Pregnancy

Valproate causes birth defects;[33] exposure during pregnancy is associated with about three times as many major abnormalities as usual, mainly spina bifida with the risks being related to the strength of medication used and use of more than one drug.[34][35] More rarely, with several other defects, including a “valproate syndrome”.[36] Characteristics of this valproate syndrome include facial features that tend to evolve with age, including a triangle-shaped forehead, tall forehead with bifrontal narrowing, epicanthic folds, medial deficiency of eyebrows, flat nasal bridge, broad nasal root, anteverted nares, shallow philtrum, long upper lip and thin vermillion borders, thick lower lip and small downturned mouth.[37] While developmental delay is usually associated with altered physical characteristics (dysmorphic features), this is not always the case.[38]

Children of mothers taking valproate during pregnancy are at risk for lower IQs.[39][40][41] Maternal valproate use during pregnancy increased the probability of autism in the offspring compared to mothers not taking valproate from 1.5% to 4.4%.[42] A 2005 study found rates of autism among children exposed to sodium valproate before birth in the cohort studied were 8.9%.[43] The normal incidence for autism in the general population is estimated at less than one percent.[44] A 2009 study found that the 3-year-old children of pregnant women taking valproate had an IQ nine points lower than that of a well-matched control group. However, further research in older children and adults is needed.[45][46][47]

Sodium valproate has been associated with paroxysmal tonic upgaze of childhood, also known as Ouvrier–Billson syndrome, from childhood or fetal exposure. This condition resolved after discontinuing valproate therapy.[48][49]

Women who intend to become pregnant should switch to a different medication if possible or decrease their dose of valproate.[50] Women who become pregnant while taking valproate should be warned that it causes birth defects and cognitive impairment in the newborn, especially at high doses (although valproate is sometimes the only drug that can control seizures, and seizures in pregnancy could have worse outcomes for the fetus than exposure to valproate). Studies have shown that taking folic acid supplements can reduce the risk of congenital neural tube defects.[22] The use of valproate for migraine or bipolar disorder during pregnancy is contraindicated in the European Union, and the medicines are not recommended for epilepsy during pregnancy unless there is no other effective treatment available.[51]

Elderly

Valproate in elderly people with dementia caused increased sleepiness. More people stopped the medication for this reason. Additional side effects of weight loss and decreased food intake were also associated with one-half of people who become sleepy.[22]

Overdose and toxicity

FormLower limitUpper limitUnit
Total (including
protein bound)
50[52]125[52]µg/mL or mg/l
350[53]700[53]μmol/L
Free6[52]22[52]µg/mL or mg/l
35[53]70[53]μmol/L

Excessive amounts of valproic acid can result in sleepiness, tremorstuporrespiratory depressioncomametabolic acidosis, and death.[54] In general, serum or plasma valproic acid concentrations are in a range of 20–100 mg/l during controlled therapy, but may reach 150–1500 mg/l following acute poisoning. Monitoring of the serum level is often accomplished using commercial immunoassay techniques, although some laboratories employ gas or liquid chromatography.[55] In contrast to other antiepileptic drugs, at present there is little favorable evidence for salivary therapeutic drug monitoring. Salivary levels of valproic acid correlate poorly with serum levels, partly due to valproate’s weak acid property (pKa of 4.9).[56]

In severe intoxication, hemoperfusion or hemofiltration can be an effective means of hastening elimination of the drug from the body.[57][58] Supportive therapy should be given to all patients experiencing an overdose and urine output should be monitored.[22] Supplemental L-carnitine is indicated in patients having an acute overdose[59][60] and also prophylactically[59] in high risk patients. Acetyl-L-carnitine lowers hyperammonemia less markedly[61] than L-carnitine.

Interactions

Valproate inhibits CYP2C9glucuronyl transferase, and epoxide hydrolase and is highly protein bound and hence may interact with drugs that are substrates for any of these enzymes or are highly protein bound themselves.[24] It may also potentiate the CNS depressant effects of alcohol.[24] It should not be given in conjunction with other antiepileptics due to the potential for reduced clearance of other antiepileptics (including carbamazepinelamotriginephenytoin and phenobarbitone) and itself.[24] It may also interact with:[22][24][62]

  • Aspirin: may increase valproate concentrations. May also interfere with valproate’s metabolism.
  • Benzodiazepines: may cause CNS depression and there are possible pharmacokinetic interactions.
  • Carbapenem antibiotics: reduce valproate levels, potentially leading to seizures.
  • Cimetidine: inhibits valproate’s metabolism in the liver, leading to increased valproate concentrations.
  • Erythromycin: inhibits valproate’s metabolism in the liver, leading to increased valproate concentrations.
  • Ethosuximide: valproate may increase ethosuximide concentrations and lead to toxicity.
  • Felbamate: may increase plasma concentrations of valproate.
  • Mefloquine: may increase valproate metabolism combined with the direct epileptogenic effects of mefloquine.
  • Oral contraceptives: may reduce plasma concentrations of valproate.
  • Primidone: may accelerate metabolism of valproate, leading to a decline of serum levels and potential breakthrough seizure.
  • Rifampicin: increases the clearance of valproate, leading to decreased valproate concentrations
  • Warfarin: valproate may increase free warfarin concentration and prolong bleeding time.
  • Zidovudine: valproate may increase zidovudine serum concentration and lead to toxicity.

Pharmacology

Pharmacodynamics

Although the mechanism of action of valproate is not fully understood,[24] traditionally, its anticonvulsant effect has been attributed to the blockade of voltage-gated sodium channels and increased brain levels of gamma-aminobutyric acid (GABA).[24] The GABAergic effect is also believed to contribute towards the anti-manic properties of valproate.[24] In animals, sodium valproate raises cerebral and cerebellar levels of the inhibitory synaptic neurotransmitter, GABA, possibly by inhibiting GABA degradative enzymes, such as GABA transaminasesuccinate-semialdehyde dehydrogenase and by inhibiting the re-uptake of GABA by neuronal cells.[24]

Prevention of neurotransmitter-induced hyperexcitability of nerve cells, via Kv7.2 channel and AKAP5, may also contribute to its mechanism.[63] Also, it has been shown to protect against a seizure-induced reduction in phosphatidylinositol (3,4,5)-trisphosphate (PIP3) as a potential therapeutic mechanism.[64]

It also has histone-deacetylase-inhibiting effects. The inhibition of histone deacetylase, by promoting more transcriptionally active chromatin structures, likely presents the epigenetic mechanism for regulation of many of the neuroprotective effects attributed to valproic acid. Intermediate molecules mediating these effects include VEGFBDNF, and GDNF.[65][66]

Endocrine actions

Valproic acid has been found to be an antagonist of the androgen and progesterone receptors, and hence as a nonsteroidal antiandrogen and antiprogestogen, at concentrations much lower than therapeutic serum levels.[67] In addition, the drug has been identified as a potent aromatase inhibitor, and suppresses estrogen concentrations.[68] These actions are likely to be involved in the reproductive endocrine disturbances seen with valproic acid treatment.[67][68]

Valproic acid has been found to directly stimulate androgen biosynthesis in the gonads via inhibition of histone deacetylases and has been associated with hyperandrogenism in women and increased 4-androstenedione levels in men.[69][70] High rates of polycystic ovary syndrome and menstrual disorders have also been observed in women treated with valproic acid.[70]

Pharmacokinetics

Some metabolites of valproic acid. Glucuronidation and β-oxidation are the main metabolic pathways; ω-oxidation metabolites are considered hepatotoxic.[71][72] Details see text.

Taken by mouth, valproate is rapidly and virtually completely absorbed from the gut.[71] When in the bloodstream, 80–90% of the substance are bound to plasma proteins, mainly albumin. Protein binding is saturable: it decreases with increasing valproate concentration, low albumin concentrations, the patient’s age, additional use of other drugs such as aspirin, as well as liver and kidney impairment.[73][74] Concentrations in the cerebrospinal fluid and in breast milk are 1 to 10% of blood plasma concentrations.[71]

The vast majority of valproate metabolism occurs in the liver.[75] Valproate is known to be metabolized by the cytochrome P450 enzymes CYP2A6CYP2B6CYP2C9, and CYP3A5.[75] It is also known to be metabolized by the UDP-glucuronosyltransferase enzymes UGT1A3UGT1A4UGT1A6UGT1A8UGT1A9UGT1A10UGT2B7, and UGT2B15.[75] Some of the known metabolites of valproate by these enzymes and uncharacterized enzymes include (see image):[75]

  • via glucuronidation (30–50%): valproic acid β-O-glucuronide
  • via beta oxidation (>40%): 2E-ene-valproic acid, 2Z-ene-valproic acid, 3-hydroxyvalproic acid, 3-oxovalproic acid
  • via omega oxidation: 5-hydroxyvalproic acid, 2-propyl-glutaric acid
  • some others: 3E-ene-valproic acid, 3Z-ene-valproic acid, 4-ene-valproic acid, 4-hydroxyvalproic acid

All in all, over 20 metabolites are known.[71]

In adult patients taking valproate alone, 30–50% of an administered dose is excreted in urine as the glucuronide conjugate.[75] The other major pathway in the metabolism of valproate is mitochondrial beta oxidation, which typically accounts for over 40% of an administered dose.[75] Typically, less than 20% of an administered dose is eliminated by other oxidative mechanisms.[75] Less than 3% of an administered dose of valproate is excreted unchanged (i.e., as valproate) in urine.[75] Only a small amount is excreted via the faeces.[71] Elimination half-life is 16±3 hours and can decrease to 4–9 hours when combined with enzyme inducers.[71][74]

Chemistry

Valproic acid is a branched short-chain fatty acid and the 2-npropyl derivative of valeric acid.[5]

History

Valproic acid was first synthesized in 1882 by Beverly S. Burton as an analogue of valeric acid, found naturally in valerian.[76] Valproic acid is a carboxylic acid, a clear liquid at room temperature. For many decades, its only use was in laboratories as a “metabolically inert” solvent for organic compounds. In 1962, the French researcher Pierre Eymard serendipitously discovered the anticonvulsant properties of valproic acid while using it as a vehicle for a number of other compounds that were being screened for antiseizure activity. He found it prevented pentylenetetrazol-induced convulsions in laboratory rats.[77] It was approved as an antiepileptic drug in 1967 in France and has become the most widely prescribed antiepileptic drug worldwide.[78] Valproic acid has also been used for migraine prophylaxis and bipolar disorder.[79]

Society and culture

Valproate is available as a generic medication.[2]

Approval status

Indications
FDA-labelled indication?[1]

TGA-labelled indication?[12]

MHRA-labelled indication?[80]
Literature support
EpilepsyYesYesYesLimited (depends on the seizure type; it can help with certain kinds of seizures: drug-resistant epilepsy, partial and absence seizures, can be used against glioblastoma and other tumors both to improve survival and treat seizures, and against tonic–clonic seizures and status epilepticus).[81][82][83][84]
Bipolar maniaYesYesYesLimited.[85]
Bipolar depressionNoNoNoModerate.[86]
Bipolar maintenanceNoNoNoLimited.[87]
Migraine prophylaxisYesYes (accepted)NoLimited.
Acute migraine managementNoNoNoOnly negative results.[88]
SchizophreniaNoNoNoWeak evidence.[89]
Agitation in dementiaNoNoNoWeak evidence. Not recommended for agitation in people with dementia.[90] Increased rate of adverse effects, including a risk of serious adverse effects.[90]
Fragile X syndromeYes (orphan)NoNoLimited.[66]
Familial adenomatous polyposisYes (orphan)NoNoLimited.
Chronic pain & fibromyalgiaNoNoNoLimited.[91]
Alcohol hallucinosisNoNoNoOne randomised double-blind placebo-controlled trial.[92]
Intractable hiccupsNoNoNoLimited, five case reports support its efficacy, however.[93]
Non-epileptic myoclonusNoNoNoLimited, three case reports support its efficacy, however.[94]
Cluster headachesNoNoNoLimited, two case reports support its efficacy.[95]
West syndromeNoNoNoA prospective clinical trial supported its efficacy in treating infantile spasms.[96]
HIV infection eradicationNoNoNoDouble-blind placebo-controlled trials have been negative.[97][98][99]
Myelodysplastic syndromeNoNoNoSeveral clinical trials have confirmed its efficacy as a monotherapy,[100] as an adjunct to tretinoin[100] and as an adjunct to hydralazine.[101]
Acute myeloid leukaemiaNoNoNoTwo clinical trials have confirmed its efficacy in this indication as both a monotherapy and as an adjunct to tretinoin.[102][103][104]
Cervical cancerNoNoNoOne clinical trial supports its use here.[105]
Malignant melanomaNoNoNoOne phase II study has seemed to discount its efficacy.[106]
Breast cancerNoNoNoA phase II study has supported its efficacy.[107]
Impulse control disorderNoNoNoLimited.[108][109]

Off-label uses

In 2012, pharmaceutical company Abbott paid $1.6 billion in fines to US federal and state governments for illegal promotion of off-label uses for Depakote, including the sedation of elderly nursing home residents.[110][111]

Some studies have suggested that valproate may reopen the critical period for learning absolute pitch and possibly other skills such as language.[112][113]

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

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

Formulations

Clinical data
Other namesvalproate sodium (USAN US)
License dataUS DailyMedValproate_sodium
Identifiers
showIUPAC name
CAS Number1069-66-5 
PubChem CID16760703
DrugBankDBSALT001257 
ChemSpider13428 
UNII5VOM6GYJ0D
KEGGD00710 
ChEBICHEBI:9925 
ChEMBLChEMBL433 
CompTox Dashboard (EPA)DTXSID6023733 
ECHA InfoCard100.002.525 
Chemical and physical data
FormulaC8H15NaO2
Molar mass166.196 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI
  (verify)
Clinical data
Trade namesDepakote, others
Other namessemisodium valproate, divalproex sodium (USAN US)
License dataUS DailyMedDivalproex_sodium
Identifiers
showIUPAC name
CAS Number76584-70-8 
PubChem CID23663956
DrugBankDBSALT000185 
ChemSpider48337 
UNII644VL95AO6
KEGGD00304 
ChEBICHEBI:4667 
ChEMBLChEMBL2105613 
CompTox Dashboard (EPA)DTXSID6023733 
ECHA InfoCard100.002.525 
Chemical and physical data
FormulaC16H31NaO4
Molar mass310.410 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI

Valproate exists in two main molecular variants: sodium valproate and valproic acid without sodium (often implied by simply valproate). A mixture between these two is termed semisodium valproate. It is unclear whether there is any difference in efficacy between these variants, except from the fact that about 10% more mass of sodium valproate is needed than valproic acid without sodium to compensate for the sodium itself.[114]

Brand names of valproic acid

Branded products include:

Brand names of sodium valproate

Portugal
  • Tablets – Diplexil-R by Bial.
United States
  • Intravenous injection – Depacon by Abbott Laboratories.
  • Syrup – Depakene by Abbott Laboratories. (Note Depakene capsules are valproic acid).
  • Depakote tablets are a mixture of sodium valproate and valproic acid.
  • Tablets – Eliaxim by Bial.
Australia
  • Epilim Crushable Tablets Sanofi[116]
  • Epilim Sugar Free Liquid Sanofi[116]
  • Epilim Syrup Sanofi[116]
  • Epilim Tablets Sanofi[116]
  • Sodium Valproate Sandoz Tablets Sanofi
  • Valpro Tablets Alphapharm
  • Valproate Winthrop Tablets Sanofi
  • Valprease tablets Sigma
New Zealand
  • Epilim by Sanofi-Aventis

All the above formulations are Pharmac-subsidised.[117]

UK
  • Depakote Tablets (as in USA)
  • Tablets – Orlept by Wockhardt and Epilim by Sanofi
  • Oral solution – Orlept Sugar Free by Wockhardt and Epilim by Sanofi
  • Syrup – Epilim by Sanofi-Aventis
  • Intravenous injection – Epilim Intravenous by Sanofi
  • Extended release tablets – Epilim Chrono by Sanofi is a combination of sodium valproate and valproic acid in a 2.3:1 ratio.
  • Enteric-coated tablets – Epilim EC200 by Sanofi is a 200-mg sodium valproate enteric-coated tablet.
UK only
  • Capsules – Episenta prolonged release by Beacon
  • Sachets – Episenta prolonged release by Beacon
  • Intravenous solution for injection – Episenta solution for injection by Beacon
Germany, Switzerland, Norway, Finland, Sweden
  • Tablets – Orfiril by Desitin Pharmaceuticals
  • Intravenous injection – Orfiril IV by Desitin Pharmaceuticals
South Africa
  • Syrup – Convulex by Byk Madaus[118]
  • Tablets – Epilim by Sanofi-synthelabo
Malaysia
  • Tablets – Epilim by Sanofi-Aventis
Romania
  • Companies are SANOFI-AVENTIS FRANCE, GEROT PHARMAZEUTIKA GMBH and DESITIN ARZNEIMITTEL GMBH
  • Types are Syrup, Extended release mini tablets, Gastric resistant coated tablets, Gastric resistant soft capsules, Extended release capsules, Extended release tablets and Extended release coated tablets
Canada
Japan
  • Tablets – Depakene by Kyowa Hakko Kirin
  • Extended release tablets – Depakene-R by Kyowa Hakko Kogyo and Selenica-R by Kowa
  • Syrup – Depakene by Kyowa Hakko Kogyo
Europe

In much of Europe, Dépakine and Depakine Chrono (tablets) are equivalent to Epilim and Epilim Chrono above.

Taiwan
Iran
  • Tablets – Epival 200 (enteric coated tablet) and Epival 500 (extended release tablet) by Iran Najo
  • Slow release tablets – Depakine Chrono by Sanofi Winthrop Industrie (France)
Israel

Depalept and Depalept Chrono (extended release tablets) are equivalent to Epilim and Epilim Chrono above. Manufactured and distributed by Sanofi-Aventis.

India, Russia and CIS countries
  • Valparin Chrono by Torrent Pharmaceuticals India
  • Valprol CR by Intas Pharmaceutical (India)
  • Encorate Chrono by Sun Pharmaceutical (India)
  • Serven Chrono by Leeven APL Biotech (India)

Brand names of valproate semisodium

  • Brazil – Depakote by Abbott Laboratories and Torval CR by Torrent do Brasil
  • Canada – Epival by Abbott Laboratories
  • Mexico – Epival and Epival ER (extended release) by Abbott Laboratories
  • United Kingdom – Depakote (for psychiatric conditions) and Epilim (for epilepsy) by Sanofi-Aventis and generics
  • United States – Depakote and Depakote ER (extended release) by Abbott Laboratories and generics[22]
  • India – Valance and Valance OD by Abbott Healthcare Pvt Ltd, Divalid ER by Linux laboratories Pvt Ltd, Valex ER by Sigmund Promedica, Dicorate by Sun Pharma
  • Germany – Ergenyl Chrono by Sanofi-Aventis and generics
  • Chile – Valcote and Valcote ER by Abbott Laboratories
  • France and other European countries — Depakote
  • Peru – Divalprax by AC Farma Laboratories
  • China – Diprate OD

References

  1. Jump up to:a b c d “Depakene, Stavzor (valproic acid) dosing, indications, interactions, adverse effects, and more”Medscape Reference. WebMD. Archived from the original on 21 February 2014. Retrieved 13 February 2014.
  2. Jump up to:a b c d e f g h i j k l “Valproic Acid”. The American Society of Health-System Pharmacists. Archived from the original on 2017-07-31. Retrieved Oct 23, 2015.
  3. ^ “Valproate banned without the pregnancy prevention programme”GOV.UK. Retrieved 26 April 2018.
  4. ^ Owens MJ, Nemeroff CB (2003). “Pharmacology of valproate”. Psychopharmacol Bull. 37 Suppl 2: 17–24. PMID 14624230.
  5. Jump up to:a b c Ghodke-Puranik Y, Thorn CF, Lamba JK, Leeder JS, Song W, Birnbaum AK, Altman RB, Klein TE (April 2013). “Valproic acid pathway: pharmacokinetics and pharmacodynamics”Pharmacogenet. Genomics23 (4): 236–241. doi:10.1097/FPC.0b013e32835ea0b2PMC 3696515PMID 23407051.
  6. ^ “Valproic acid”DrugBank. University of Alberta. 29 July 2017. Archived from the original on 31 July 2017. Retrieved 30 July2017.
  7. ^ Scott, D.F. (1993). The history of epileptic therapy : an account of how medication was developed (1. publ. ed.). Carnforth u.a.: Parthenon Publ. Group. p. 131. ISBN 9781850703914.
  8. ^ World Health Organization (2019). World Health Organization model list of essential medicines: 21st list 2019. Geneva: World Health Organization. hdl:10665/325771. WHO/MVP/EMP/IAU/2019.06. License: CC BY-NC-SA 3.0 IGO.
  9. ^ “The Top 300 of 2021”ClinCalc. Retrieved 18 February 2021.
  10. ^ “Divalproex Sodium – Drug Usage Statistics”ClinCalc. Retrieved 18 February 2021.
  11. ^ Brayfield, Alison (ed.). Martindale: The Complete Drug Reference. London: Pharmaceutical Press. Retrieved March 3,2018.
  12. Jump up to:a b c Rossi, S, ed. (2013). Australian Medicines Handbook(2013 ed.). Adelaide: The Australian Medicines Handbook Unit Trust. ISBN 978-0-9805790-9-3.
  13. ^ Löscher W (2002). “Basic pharmacology of valproate: a review after 35 years of clinical use for the treatment of epilepsy”. CNS Drugs16 (10): 669–694. doi:10.2165/00023210-200216100-00003PMID 12269861S2CID 67999301.
  14. ^ Olsen KB, Taubøll E, Gjerstad L (2007). “Valproate is an effective, well-tolerated drug for treatment of status epilepticus/serial attacks in adults”. Acta Neurol. Scand. Suppl187: 51–4. doi:10.1111/j.1600-0404.2007.00847.xPMID 17419829S2CID 11159645.
  15. ^ Kwan SY (2010). “The role of intravenous valproate in convulsive status epilepticus in the future” (PDF). Acta Neurol Taiwan19(2): 78–81. PMID 20830628.[permanent dead link]
  16. ^ “Valproate Information”. Fda.gov. Archived from the original on 2015-05-03. Retrieved 2015-04-24.
  17. ^ Jochim, Janina; Rifkin-Zybutz, Raphael; Geddes, John; Cipriani, Andrea (7 October 2019). “Valproate for acute mania”Cochrane Database of Systematic Reviews10: CD004052. doi:10.1002/14651858.CD004052.pub2PMC 6797024PMID 31621892.
  18. Jump up to:a b Wang, Y; Xia, J; Helfer, B (2016). “Valproate for schizophrenia”Cochrane Database of Systematic Reviews11: CD004028.pub4. doi:10.1002/14651858.CD004028.pub4PMC 6734130PMID 27884042. Archived from the originalon 2017-07-29. Retrieved 2017-07-27.
  19. ^ Pirritano D, Plastino M, Bosco D, Gallelli L, Siniscalchi A, De Sarro G (2014). “Gambling disorder during dopamine replacement treatment in Parkinson’s disease: a comprehensive review”Biomed Res Int2014: 1–9. doi:10.1155/2014/728038PMC 4119624PMID 25114917.
  20. ^ Connolly B, Fox SH (2014). “Treatment of cognitive, psychiatric, and affective disorders associated with Parkinson’s disease”Neurotherapeutics11 (1): 78–91. doi:10.1007/s13311-013-0238-xPMC 3899484PMID 24288035.
  21. ^ Averbeck BB, O’Sullivan SS, Djamshidian A (2014). “Impulsive and compulsive behaviors in Parkinson’s disease”Annu Rev Clin Psychol10: 553–80. doi:10.1146/annurev-clinpsy-032813-153705PMC 4197852PMID 24313567.
  22. Jump up to:a b c d e f g h i “Depakote- divalproex sodium tablet, delayed release”Archived from the original on 5 March 2016. Retrieved 10 November 2015.
  23. ^ Činčárová L, Zdráhal Z, Fajkus J (2013). “New perspectives of valproic acid in clinical practice”. Expert Opin Investig Drugs22(12): 1535–1547. doi:10.1517/13543784.2013.853037PMID 24160174S2CID 11855893.
  24. Jump up to:a b c d e f g h i j k l m n o “Valpro sodium valproate” (PDF). TGA eBusiness Services. Alphapharm Pty Limited. 16 December 2013. Retrieved 14 February 2014.
  25. ^ “Depakote 250mg Tablets – Summary of Product Characteristics”electronic Medicines Compendium. Sanofi. 28 November 2013. Archived from the original on 1 February 2014. Retrieved 18 January 2014.
  26. ^ Wu S, Legido A, De Luca F (2004). “Effects of valproic acid on longitudinal bone growth”. J Child Neurol19 (1): 26–30. doi:10.1177/088307380401900105011PMID 15032379S2CID 19827846.
  27. ^ Robinson PB, Harvey W, Belal MS (1988). “Inhibition of cartilage growth by the anticonvulsant drugs diphenylhydantoin and sodium valproate”Br J Exp Pathol69 (1): 17–22. PMC 2013195PMID 3126792.
  28. ^ Guo CY, Ronen GM, Atkinson SA (2002). “Long-term valproate and lamotrigine treatment may be a marker for reduced growth and bone mass in children with epilepsy”. Epilepsia42 (9): 1141–7. doi:10.1046/j.1528-1157.2001.416800.xPMID 11580761S2CID 25499280.
  29. ^ Guo CY, Ronen GM, Atkinson SA (2002). “Long-term valproate and lamotrigine treatment may be a marker for reduced growth and bone mass in children with epilepsy”. Epilepsia42 (9): 1141–7. doi:10.1046/j.1528-1157.2001.416800.xPMID 11580761S2CID 25499280.
  30. ^ “Could Depakote cause Mydriasis”. eHealthMe.com. 2014-11-18. Archived from the original on 2014-12-05. Retrieved 2015-04-24.
  31. ^ Bilo, Leonilda; Meo, Roberta (October 2008). “Polycystic ovary syndrome in women using valproate: a review”. Gynecological Endocrinology24 (10): 562–70. doi:10.1080/09513590802288259PMID 19012099S2CID 36426338.
  32. ^ Chukwu, J; Delanty, N; Webb, D; Cavalleri, GL (January 2014). “Weight change, genetics and antiepileptic drugs”. Expert Review of Clinical Pharmacology7 (1): 43–51. doi:10.1586/17512433.2014.857599PMID 24308788S2CID 33444886.
  33. ^ New evidence in France of harm from epilepsy drug valproateArchived 2017-04-21 at the Wayback Machine BBC, 2017
  34. ^ Koch S, Göpfert-Geyer I, Jäger-Roman E, et al. (February 1983). “[Anti-epileptic agents during pregnancy. A prospective study on the course of pregnancy, malformations and child development]”. Dtsch. Med. Wochenschr. (in German). 108 (7): 250–7. doi:10.1055/s-2008-1069536PMID 6402356.
  35. ^ Moore SJ, Turnpenny P, Quinn A, et al. (July 2000). “A clinical study of 57 children with fetal anticonvulsant syndromes”J. Med. Genet37 (7): 489–97. doi:10.1136/jmg.37.7.489PMC 1734633PMID 10882750.
  36. ^ Ornoy A (2009). “Valproic acid in pregnancy: how much are we endangering the embryo and fetus?”. Reprod. Toxicol28 (1): 1–10. doi:10.1016/j.reprotox.2009.02.014PMID 19490988.
  37. ^ Kulkarni ML, Zaheeruddin M, Shenoy N, Vani HN (2006). “Fetal valproate syndrome”. Indian J Pediatr73 (10): 937–939. doi:10.1007/bf02859291PMID 17090909S2CID 38371596.
  38. ^ Adab N, Kini U, Vinten J, et al. (November 2004). “The longer term outcome of children born to mothers with epilepsy”J. Neurol. Neurosurg. Psychiatry75 (11): 1575–83. doi:10.1136/jnnp.2003.029132PMC 1738809PMID 15491979This argues that the fetal valproate syndrome constitutes a real clinical entity that includes developmental delay and cognitive impairments, but that some children might exhibit some developmental delay without marked dysmorphism.
  39. ^ Umur AS, Selcuki M, Bursali A, Umur N, Kara B, Vatansever HS, Duransoy YK (2012). “Simultaneous folate intake may prevent adverse effect of valproic acid on neurulating nervous system”. Childs Nerv Syst28 (5): 729–737. doi:10.1007/s00381-011-1673-9PMID 22246336S2CID 20344828.
  40. ^ Cassels, Caroline (December 8, 2006). “NEAD: In Utero Exposure To Valproate Linked to Poor Cognitive Outcomes in Kids”. Medscape. Archived from the original on July 31, 2011. Retrieved 2007-05-23.
  41. ^ Meador KJ, Baker GA, Finnell RH, Kalayjian LA, Liporace JD, Loring DW, Mawer G, Pennell PB, Smith JC, Wolff MC (2006). “In utero antiepileptic drug exposure: fetal death and malformations”Neurology67 (3): 407–412. doi:10.1212/01.wnl.0000227919.81208.b2PMC 1986655PMID 16894099.
  42. ^ Christensen J, Grønborg TK, Sørensen MJ, Schendel D, Parner ET, Pedersen LH, Vestergaard M (2013). “Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism”JAMA309 (16): 1696–1703. doi:10.1001/jama.2013.2270PMC 4511955PMID 23613074.
  43. ^ Rasalam AD, Hailey H, Williams JH, et al. (August 2005). “Characteristics of fetal anticonvulsant syndrome associated autistic disorder”. Dev Med Child Neurol47 (8): 551–5. doi:10.1017/S0012162205001076PMID 16108456.
  44. ^ Autism Society of America: About Autism Archived 2011-01-10 at the Wayback Machine
  45. ^ I.Q. Harmed by Epilepsy Drug in Utero Archived 2015-12-29 at the Wayback Machine By RONI CARYN RABIN, New York Times, April 15, 2009
  46. ^ Meador KJ, Baker GA, Browning N, Clayton-Smith J, Combs-Cantrell DT, Cohen M, Kalayjian LA, Kanner A, Liporace JD, Pennell PB, Privitera M, Loring DW (2009). “Cognitive function at 3 years of age after fetal exposure to antiepileptic drugs”N. Engl. J. Med360 (16): 1597–1605. doi:10.1056/NEJMoa0803531PMC 2737185PMID 19369666.
  47. ^ Valproate Products: Drug Safety Communication – Risk of Impaired Cognitive Development in Children Exposed In Utero (During Pregnancy) Archived 2011-09-02 at the Wayback Machine. FDA. June 2011
  48. ^ Luat, AF (20 September 2007). “Paroxysmal tonic upgaze of childhood with co-existent absence epilepsy”. Epileptic Disorders9(3): 332–6. doi:10.1684/epd.2007.0119 (inactive 31 May 2021). PMID 17884759.
  49. ^ Ouvrier, RA (July 1988). “Benign paroxysmal tonic upgaze of childhood”. Journal of Child Neurology3 (3): 177–80. doi:10.1177/088307388800300305PMID 3209843S2CID 38127378.
  50. ^ Valproate Not To Be Used for Migraine During Pregnancy, FDA Warns Archived 2013-07-09 at the Wayback Machine
  51. ^ “New measures to avoid valproate exposure in pregnancy endorsed”European Medicines Agency. 31 May 2018.
  52. Jump up to:a b c d Bentley, Suzanne (Dec 11, 2013). “Valproic Acid Level”MedscapeArchived from the original on 2015-05-04. Retrieved 2015-06-05.
  53. Jump up to:a b c d “Free Valproic Acid Assay (Reference – 2013.03.006) Notice of Assessment” (PDF). Canadian Agency for Drugs and Technologies in Health (CADTH) with INESSS’s permission. April 2014. Archived from the original (PDF) on 2016-03-03. Retrieved 2015-06-05.
  54. ^ Rissardo, Jamir Pitton; Caprara, Ana Letícia Fornari; Durante, Ícaro (2021). “Valproate-associated Movement Disorder: A Literature Review”Prague Medical Report122 (3): 140–180. doi:10.14712/23362936.2021.14ISSN 1214-6994.
  55. ^ Sztajnkrycer MD (2002). “Valproic acid toxicity: overview and management”. J. Toxicol. Clin. Toxicol40 (6): 789–801. doi:10.1081/CLT-120014645PMID 12475192S2CID 23031095.
  56. ^ Patsalos PN, Berry DJ (2013). “Therapeutic drug monitoring of antiepileptic drugs by use of saliva”. Ther Drug Monit35 (1): 4–29. doi:10.1097/FTD.0b013e31827c11e7PMID 23288091S2CID 15338188.
  57. ^ Thanacoody RH (2009). “Extracorporeal elimination in acute valproic acid poisoning”. Clin Toxicol47 (7): 609–616. doi:10.1080/15563650903167772PMID 19656009S2CID 13592043.
  58. ^ R. Baselt, Disposition of Toxic Drugs and Chemicals in Man, 8th edition, Biomedical Publications, Foster City, CA, 2008, pp. 1622–1626.
  59. Jump up to:a b Lheureux PE, Penaloza A, Zahir S, Gris M (2005). “Science review: carnitine in the treatment of valproic acid-induced toxicity – what is the evidence?”Crit Care9 (5): 431–440. doi:10.1186/cc3742PMC 1297603PMID 16277730.
  60. ^ Mock CM, Schwetschenau KH (2012). “Levocarnitine for valproic-acid-induced hyperammonemic encephalopathy”. Am J Health Syst Pharm69 (1): 35–39. doi:10.2146/ajhp110049PMID 22180549.
  61. ^ Matsuoka M, Igisu H (1993). “Comparison of the effects of L-carnitine, D-carnitine and acetyl-L-carnitine on the neurotoxicity of ammonia”. Biochem. Pharmacol46 (1): 159–164. doi:10.1016/0006-2952(93)90360-9PMID 8347126.
  62. ^ Herzog, Andrew; Farina, Erin (June 9, 2005). “Serum Valproate Levels with Oral Contraceptive Use”. Epilepsia46 (6): 970–971. doi:10.1111/j.1528-1167.2005.00605.xPMID 15946343S2CID 7696039.
  63. ^ Kay, Hee Yeon; Greene, Derek L.; Kang, Seungwoo; Kosenko, Anastasia; Hoshi, Naoto (2015-10-01). “M-current preservation contributes to anticonvulsant effects of valproic acid”The Journal of Clinical Investigation125 (10): 3904–3914. doi:10.1172/JCI79727ISSN 0021-9738PMC 4607138PMID 26348896.
  64. ^ Chang P, Walker MC, Williams RS (2014). “Seizure-induced reduction in PIP3 levels contributes to seizure-activity and is rescued by valproic acid”Neurobiol. Dis62: 296–306. doi:10.1016/j.nbd.2013.10.017PMC 3898270PMID 24148856.
  65. ^ Kostrouchová M, Kostrouch Z, Kostrouchová M (2007). “Valproic acid, a molecular lead to multiple regulatory pathways” (PDF). Folia Biol. (Praha)53 (2): 37–49. PMID 17448293. Archived from the original (PDF) on 2014-02-21. Retrieved 2014-02-13.
  66. Jump up to:a b Chiu CT, Wang Z, Hunsberger JG, Chuang DM (2013). “Therapeutic potential of mood stabilizers lithium and valproic acid: beyond bipolar disorder”Pharmacol. Rev65 (1): 105–142. doi:10.1124/pr.111.005512PMC 3565922PMID 23300133.
  67. Jump up to:a b Death AK, McGrath KC, Handelsman DJ (2005). “Valproate is an anti-androgen and anti-progestin” (PDF). Steroids70 (14): 946–53. doi:10.1016/j.steroids.2005.07.003hdl:10453/16875PMID 16165177S2CID 25958985.
  68. Jump up to:a b Wyllie E, Cascino GD, Gidal BE, Goodkin HP (17 February 2012). Wyllie’s Treatment of Epilepsy: Principles and Practice. Lippincott Williams & Wilkins. pp. 288–. ISBN 978-1-4511-5348-4Archived from the original on 6 June 2014.
  69. ^ Uchida, Hiroshi; Maruyama, Tetsuo; Arase, Toru; Ono, Masanori; Nagashima, Takashi; Masuda, Hirotaka; Asada, Hironori; Yoshimura, Yasunori (2005). “Histone acetylation in reproductive organs: Significance of histone deacetylase inhibitors in gene transcription”Reproductive Medicine and Biology4 (2): 115–122. doi:10.1111/j.1447-0578.2005.00101.xISSN 1445-5781PMC 5891791PMID 29662388.
  70. Jump up to:a b Isojärvi, Jouko I T; Taubøll, Erik; Herzog, Andrew G (2005). “Effect of Antiepileptic Drugs on Reproductive Endocrine Function in Individuals with Epilepsy”. CNS Drugs19 (3): 207–223. doi:10.2165/00023210-200519030-00003ISSN 1172-7047PMID 15740176S2CID 9893959.
  71. Jump up to:a b c d e f Haberfeld H, ed. (2021). Austria-Codex (in German). Vienna: Österreichischer Apothekerverlag. Depakine chrono retard 300 mg Filmtabletten.
  72. ^ Kumar, S.; Wong, H.; Yeung, S. A.; Riggs, K. W.; Abbott, F. S.; Rurak, D. W. (2000). “Disposition of valproic acid in maternal, fetal, and newborn sheep. II: Metabolism and renal elimination”. Drug Metabolism and Disposition: The Biological Fate of Chemicals28(7): 857–864. PMID 10859160.
  73. ^ Schneemann; Young, Lloyd; Koda-Kimble, Mary Anne, eds. (2001). Angewandte Arzneimitteltherapie (in German). Springer. pp. 28–29. ISBN 3540413561.
  74. Jump up to:a b Valproate FDA Professional Drug Information. Accessed 2021-08-06.
  75. Jump up to:a b c d e f g h “Pharmacology”Valproic AcidDrugBank. University of Alberta. 31 August 2017.
  76. ^ Burton BS (1882). “On the propyl derivatives and decomposition products of ethylacetoacetate”. Am. Chem. J3: 385–395.
  77. ^ Meunier H, Carraz G, Neunier Y, Eymard P, Aimard M (1963). “[Pharmacodynamic properties of N-dipropylacetic acid]” [Pharmacodynamic properties of N-dipropylacetic acid]. Thérapie (in French). 18: 435–438. PMID 13935231.
  78. ^ Perucca E (2002). “Pharmacological and therapeutic properties of valproate: a summary after 35 years of clinical experience”. CNS Drugs16 (10): 695–714. doi:10.2165/00023210-200216100-00004PMID 12269862S2CID 803106.
  79. ^ Henry TR (2003). “The history of valproate in clinical neuroscience”. Psychopharmacol Bull. 37 Suppl 2: 5–16. PMID 14624229.
  80. ^ Joint Formulary Committee (2013). British National Formulary (BNF) (65 ed.). London, UK: Pharmaceutical Press. ISBN 978-0-85711-084-8.
  81. ^ Rimmer EM, Richens A (May–June 1985). “An update on sodium valproate”. Pharmacotherapy5 (3): 171–84. doi:10.1002/j.1875-9114.1985.tb03413.xPMID 3927267S2CID 7700266.
  82. ^ Glauser, Tracy A; Cnaan, Avital; Shinnar, Shlomo; Hirtz, Deborah G; Dlugos, Dennis; Masur, David; Clark, Peggy O; Capparelli, Edmund V; Adamson, Peter C (2010). “Ethosuximide, valproic acid, and lamotrigine in childhood absence epilepsy”New England Journal of Medicine362 (9): 790–9. doi:10.1056/NEJMoa0902014PMC 2924476PMID 20200383.
  83. ^ Jiang, Mei (6 April 2015). “Co-Administration of Valproic Acid and Lamotrigine in the Treatment of Refractory Epilepsy (P1.238)”Neurology84 (14 Supplement): P1.238 – via http://www.neurology.org.
  84. ^ Berendsen, S.; Kroonen, J.; Seute, T.; Snijders, T.; Broekman, M. L. D.; Spliet, W. G. M.; Willems, M.; Artesi, M.; Bours, V.; Robe, P. A. (1 September 2014). “O9.06 * Prognostic Relevance and Oncogenic Correlates of Epilepsy in Glioblastoma Patients”Neuro-Oncology16 (suppl_2): ii21. doi:10.1093/neuonc/nou174.77PMC 4185847.
  85. ^ Vasudev K, Mead A, Macritchie K, Young AH (2012). “Valproate in acute mania: is our practice evidence based?”. Int J Health Care Qual Assur25 (1): 41–52. doi:10.1108/09526861211192395PMID 22455007.
  86. ^ Bond DJ, Lam RW, Yatham LN (2010). “Divalproex sodium versus placebo in the treatment of acute bipolar depression: a systematic review and meta-analysis”. J Affect Disord124 (3): 228–334. doi:10.1016/j.jad.2009.11.008PMID 20044142.
  87. ^ Haddad PM, Das A, Ashfaq M, Wieck A (2009). “A review of valproate in psychiatric practice”. Expert Opin Drug Metab Toxicol5(5): 539–51. doi:10.1517/17425250902911455PMID 19409030S2CID 74028228.
  88. ^ Frazee LA, Foraker KC (2008). “Use of intravenous valproic acid for acute migraine”. Ann Pharmacother42 (3): 403–7. doi:10.1345/aph.1K531PMID 18303140S2CID 207263036.
  89. ^ Wang, Yijun; Xia, Jun; Helfer, Bartosz; Li, Chunbo; Leucht, Stefan (2016). “Valproate for schizophrenia”The Cochrane Database of Systematic Reviews11: CD004028. doi:10.1002/14651858.CD004028.pub4ISSN 1469-493XPMC 6734130PMID 27884042.
  90. Jump up to:a b Baillon, Sarah F.; Narayana, Usha; Luxenberg, Jay S.; Clifton, Andrew V. (2018-10-05). “Valproate preparations for agitation in dementia”The Cochrane Database of Systematic Reviews2018(10): CD003945. doi:10.1002/14651858.CD003945.pub4ISSN 1469-493XPMC 6516950PMID 30293233.
  91. ^ Gill D, Derry S, Wiffen PJ, Moore RA (2011). “Valproic acid and sodium valproate for neuropathic pain and fibromyalgia in adults”Cochrane Database Syst Rev (10): CD009183. doi:10.1002/14651858.CD009183.pub2PMC 6540387PMID 21975791.
  92. ^ Aliyev ZN, Aliyev NA (July–August 2008). “Valproate treatment of acute alcohol hallucinosis: a double-blind, placebo-controlled study”. Alcohol Alcohol43 (4): 456–459. doi:10.1093/alcalc/agn043PMID 18495806.
  93. ^ Jacobson PL, Messenheimer JA, Farmer TW (1981). “Treatment of intractable hiccups with valproic acid”. Neurology31 (11): 1458–60. doi:10.1212/WNL.31.11.1458PMID 6796902S2CID 1578958.
  94. ^ Sotaniemi K (1982). “Valproic acid in the treatment of nonepileptic myoclonus”. Arch. Neurol39 (7): 448–9. doi:10.1001/archneur.1982.00510190066025PMID 6808975.
  95. ^ Wheeler SD (July–August 1998). “Significance of migrainous features in cluster headache: divalproex responsiveness”. Headache38 (7): 547–51. doi:10.1046/j.1526-4610.1998.3807547.xPMID 15613172S2CID 27948702.
  96. ^ Siemes H, Spohr HL, Michael T, Nau H (September–October 1988). “Therapy of infantile spasms with valproate: results of a prospective study”. Epilepsia29 (5): 553–60. doi:10.1111/j.1528-1157.1988.tb03760.xPMID 2842127S2CID 23789333.
  97. ^ Smith SM (2005). “Valproic acid and HIV-1 latency: beyond the sound bite” (PDF). Retrovirology2 (1): 56. doi:10.1186/1742-4690-2-56PMC 1242254PMID 16168066. Archived from the original (PDF) on 2015-09-24. Retrieved 2014-02-13.
  98. ^ Routy JP, Tremblay CL, Angel JB, Trottier B, Rouleau D, Baril JG, Harris M, Trottier S, Singer J, Chomont N, Sékaly RP, Boulassel MR (2012). “Valproic acid in association with highly active antiretroviral therapy for reducing systemic HIV-1 reservoirs: results from a multicentre randomized clinical study”. HIV Med13 (5): 291–6. doi:10.1111/j.1468-1293.2011.00975.xPMID 22276680S2CID 27571864.
  99. ^ Archin NM, Cheema M, Parker D, Wiegand A, Bosch RJ, Coffin JM, Eron J, Cohen M, Margolis DM (2010). “Antiretroviral intensification and valproic acid lack sustained effect on residual HIV-1 viremia or resting CD4+ cell infection”PLOS ONE5 (2): e9390. Bibcode:2010PLoSO…5.9390Adoi:10.1371/journal.pone.0009390PMC 2826423PMID 20186346.
  100. Jump up to:a b Hardy JR, Rees EA, Gwilliam B, Ling J, Broadley K, A’Hern R (2001). “A phase II study to establish the efficacy and toxicity of sodium valproate in patients with cancer-related neuropathic pain” (PDF). J Pain Symptom Manage21 (3): 204–9. doi:10.1016/S0885-3924(00)00266-9PMID 11239739.[permanent dead link]
  101. ^ Candelaria M, Herrera A, Labardini J, González-Fierro A, Trejo-Becerril C, Taja-Chayeb L, Pérez-Cárdenas E, de la Cruz-Hernández E, Arias-Bofill D, Vidal S, Cervera E, Dueñas-Gonzalez A (2011). “Hydralazine and magnesium valproate as epigenetic treatment for myelodysplastic syndrome. Preliminary results of a phase-II trial”. Ann. Hematol90 (4): 379–387. doi:10.1007/s00277-010-1090-2PMID 20922525S2CID 13437134.
  102. ^ Bug G, Ritter M, Wassmann B, Schoch C, Heinzel T, Schwarz K, Romanski A, Kramer OH, Kampfmann M, Hoelzer D, Neubauer A, Ruthardt M, Ottmann OG (2005). “Clinical trial of valproic acid and all-trans retinoic acid in patients with poor-risk acute myeloid leukemia”. Cancer104 (12): 2717–2725. doi:10.1002/cncr.21589PMID 16294345S2CID 1802132.
  103. ^ Kuendgen A, Schmid M, Schlenk R, Knipp S, Hildebrandt B, Steidl C, Germing U, Haas R, Dohner H, Gattermann N (2006). “The histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination with all-trans retinoic acid in patients with acute myeloid leukemia”. Cancer106 (1): 112–119. doi:10.1002/cncr.21552PMID 16323176S2CID 43747497.
  104. ^ Fredly H, Gjertsen BT, Bruserud O (2013). “Histone deacetylase inhibition in the treatment of acute myeloid leukemia: the effects of valproic acid on leukemic cells, and the clinical and experimental evidence for combining valproic acid with other antileukemic agents” (PDF). Clin Epigenetics5 (1): 12. doi:10.1186/1868-7083-5-12PMC 3733883PMID 23898968. Archived from the original (PDF) on 2014-02-21. Retrieved 2014-02-13.
  105. ^ Coronel J, Cetina L, Pacheco I, Trejo-Becerril C, González-Fierro A, de la Cruz-Hernandez E, Perez-Cardenas E, Taja-Chayeb L, Arias-Bofill D, Candelaria M, Vidal S, Dueñas-González A (2011). “A double-blind, placebo-controlled, randomized phase III trial of chemotherapy plus epigenetic therapy with hydralazine valproate for advanced cervical cancer. Preliminary results”. Med. Oncol. 28 Suppl 1: S540–6. doi:10.1007/s12032-010-9700-3PMID 20931299S2CID 207372333.
  106. ^ Rocca A, Minucci S, Tosti G, Croci D, Contegno F, Ballarini M, Nolè F, Munzone E, Salmaggi A, Goldhirsch A, Pelicci PG, Testori A (2009). “A phase I-II study of the histone deacetylase inhibitor valproic acid plus chemoimmunotherapy in patients with advanced melanoma”Br. J. Cancer100 (1): 28–36. doi:10.1038/sj.bjc.6604817PMC 2634690PMID 19127265.
  107. ^ Munster P, Marchion D, Bicaku E, Lacevic M, Kim J, Centeno B, Daud A, Neuger A, Minton S, Sullivan D (2009). “Clinical and biological effects of valproic acid as a histone deacetylase inhibitor on tumor and surrogate tissues: phase I/II trial of valproic acid and epirubicin/FEC” (PDF). Clin. Cancer Res15 (7): 2488–96. doi:10.1158/1078-0432.CCR-08-1930PMID 19318486S2CID 3230087.
  108. ^ Hicks CW, Pandya MM, Itin I, Fernandez HH (2011). “Valproate for the treatment of medication-induced impulse-control disorders in three patients with Parkinson’s disease”. Parkinsonism Relat. Disord17 (5): 379–81. doi:10.1016/j.parkreldis.2011.03.003PMID 21459656.
  109. ^ Sriram A, Ward HE, Hassan A, Iyer S, Foote KD, Rodriguez RL, McFarland NR, Okun MS (2013). “Valproate as a treatment for dopamine dysregulation syndrome (DDS) in Parkinson’s disease”. J. Neurol260 (2): 521–7. doi:10.1007/s00415-012-6669-1PMID 23007193S2CID 21544457.
  110. ^ Aizenman, N. C. (7 May 2012). “Abbott Laboratories to pay $1.6 billion over illegal marketing of Depakote”Washington Post. Retrieved 27 June 2018.
  111. ^ Schmidt, Michael; Thomas, Katie (8 May 2012). “Abbott settles marketing lawsuit”New York Times. Retrieved 27 June 2018.
  112. ^ Gervain J, Vines BW, Chen LM, Seo RJ, Hensch TK, Werker JF, Young AH (2013). “Valproate reopens critical-period learning of absolute pitch”Frontiers in Systems Neuroscience7: 102. doi:10.3389/fnsys.2013.00102PMC 3848041PMID 24348349.
  113. ^ Thomson, Helen. “Learning drugs reawaken grown-up brain’s inner child”New Scientist. New Scientist Ltd. Retrieved 8 May2021.
  114. ^ David Taylor; Carol Paton; Shitij Kapur (2009). The Maudsley Prescribing Guidelines, Tenth Edition (10, revised ed.). CRC Press. p. 124. ISBN 9780203092835.
  115. ^ “Depakene- valproic acid capsule, liquid filled”DailyMed. 19 September 2019. Retrieved 14 April 2020.
  116. Jump up to:a b c d “Australian product information epilim (sodium valproate) crushable tablets, enteric-coated tablets, syrup, liquid” (PDF). TGA eBS. 15 April 2020. Retrieved 15 April 2020.
  117. ^ “Sodium valproate — Pharmaceutical Schedule”. Pharmaceutical Management Agency. Archived from the originalon 4 March 2016. Retrieved 22 June 2014.
  118. ^ South African Electronic Package Inserts: Convulex

External links

Clinical data
Trade namesDepakote, Epilim, Convulex, others
Other namesValproic acid; Sodium valproate (sodium); Valproate semisodium (semisodium); 2-Propylvaleric acid
AHFS/Drugs.comMonograph
MedlinePlusa682412
License dataUS DailyMedValproic_acidUS FDAValproic%20acid
Pregnancy
category
AU: D
Routes of
administration
By mouthintravenous
ATC codeN03AG01 (WHO)
Legal status
Legal statusAU: S4 (Prescription only)CA℞-onlyUK: POM (Prescription only)US: ℞-only
Pharmacokinetic data
BioavailabilityRapid absorption
Protein binding80–90%[1]
MetabolismLiverglucuronide conjugation 30–50%, mitochondrial β-oxidation over 40%
Elimination half-life9–16 hours[1]
ExcretionUrine (30–50%)[1]
Identifiers
showIUPAC name
CAS Number99-66-1 
PubChem CID3121
IUPHAR/BPS7009
DrugBankDB00313 
ChemSpider3009 
UNII614OI1Z5WI
KEGGD00399 
ChEBICHEBI:39867 
ChEMBLChEMBL109 
NIAID ChemDB057177
CompTox Dashboard (EPA)DTXSID6023733 
ECHA InfoCard100.002.525 
Chemical and physical data
FormulaC8H16O2
Molar mass144.214 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI
  (verify)

Patent 

Publication numberPriority datePublication dateAssigneeTitleCA1144558A *1979-10-221983-04-12Francis E. FischerProcess for making sodium hydrogen divalproateUS4988731A *1979-08-201991-01-29Abbott LaboratoriesSodium hydrogen divalproate oligomerUS5212326A *1979-08-201993-05-18Abbott LaboratoriesSodium hydrogen divalproate oligomerWO2001032595A1 *1999-11-022001-05-10Cilag AgMethod for producing compounds of the valproinic acidUS20030018215A1 *2001-06-292003-01-23Procos S.P.A.Process for the preparation of sodium divalproatePublication numberPriority datePublication dateAssigneeTitleUS20110040122A1 *2009-08-112011-02-17Sci Pharmtech, Inc.Method for preparing metal salt of valproic acidCN102942467A *2012-10-172013-02-27山东方明药业集团股份有限公司Preparation method of divalproex sodiumCN103183600A *2011-12-302013-07-03北大方正集团有限公司Method for preparing divalproex sodium

////// divalproex, Anticonvulsant,  Antimigraine, Antimanic, valproic acid, sodium valproate

CCCC(CCC)C(O)=O

NEW DRUG APPROVALS

ONE TIME

$10.00

LORNOXICAM


Lornoxicam skeletal.svg
ChemSpider 2D Image | Lornoxicam | C13H10ClN3O4S2
Lornoxicam

LORNOXICAM

chlortenoxicam

  • Molecular FormulaC13H10ClN3O4S2
  • Average mass371.819 Da

70374-39-9[RN]Chlortenoxicam, CTX, ER09126G7A
2H-thieno[2,3-e]-1,2-thiazine-3-carboxamide, 6-chloro-4-hydroxy-2-methyl-N-2-pyridinyl-, 1,1-dioxide
6233
6-Chlor-4-hydroxy-2-methyl-N-(pyridin-2-yl)-2H-thieno[2,3-e][1,2]thiazin-3-carboxamid-1,1-dioxid
6-Chloro-4-hydroxy-2-methyl-N-(2-pyridinyl)-2H-thieno[2,3-e][1,2]thiazine-3-carboxamide 1,1-dioxide

  • Chlortenoxicam, Ro-13-9297
  • ATC:M01AC05
  • CCRIS 8589
  • Ro 13-9297

Lorcam (Taisho Pharmaceutical Co.) / Xafon (Nycomed)LornoxicamCAS Registry Number: 70374-39-9 
CAS Name: 6-Chloro-4-hydroxy-2-methyl-N-2-pyridinyl-2H-thieno[2,3-e]-1,2-thiazine-3-carboxamide 1,1-dioxide 
Additional Names: 6-chloro-4-hydroxy-2-methyl-3-(2-pyridylcarbamoyl)-2H-thieno[2,3-e]-1,2-thiazine-1,1-dioxide; chlortenoxicam 
Manufacturers’ Codes: Ro-13-9297; TS-110 
Trademarks: Xefo (Nycomed) 
Molecular Formula: C13H10ClN3O4S2 
Molecular Weight: 371.82 
Percent Composition: C 41.99%, H 2.71%, Cl 9.53%, N 11.30%, O 17.21%, S 17.25% 
Literature References: Cyclooxygenase inhibitor; structurally similar to tenoxicam, q.v.
Prepn: R. Pfister et al.,DE2838851eidem,US4180662 (both 1979 to Hoffmann-La Roche).Clinical pharmacokinetics: S. I. Ankier et al.,Postgrad. Med. J.64, 752 (1988). Symposium on pharmacology and clinical experience: ibid.66, Suppl. 4, S1-S50 (1990). Overview of pharmacology and safety assessment: T. P. Pruss et al.,ibid. S18. 
Properties: Orange to yellow crystals, mp 225-230° (dec). pKa2 4.7. uv max: 371 nm. Partition coefficient (n-octanol/pH 7.4 buffer): 1.8. LD50 orally in mice, rats, rabbits, dogs, monkeys: >10 mg/kg (Pruss). 
Melting point: mp 225-230° (dec) 
pKa: pKa2 4.7 
Log P: Partition coefficient (n-octanol/pH 7.4 buffer): 1.8 
Absorption maximum: uv max: 371 nm 
Toxicity data: LD50 orally in mice, rats, rabbits, dogs, monkeys: >10 mg/kg (Pruss) 
Therap-Cat: Anti-inflammatory; analgesic. 
Keywords: Analgesic (Non-Narcotic); Anti-inflammatory (Nonsteroidal); Thiazinecarboxamides.

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

/////////////////////////////////////////////////////////////////////////////////////////////////////
SYN

CAS-RNFormulaChemical NameCAS Index Name
504-29-0C5H6N22-aminopyridine2-Pyridinamine
7790-94-5ClHO3Schlorosulfonic acidChlorosulfuric acid
56946-84-0C5H5Cl2NO2S22,5-dichloro-N-methyl-3-thiophenesulfonamide3-Thiophenesulfonamide, 2,5-dichloro-N-methyl-
3172-52-9C4H2Cl2S2,5-dichlorothiopheneThiophene, 2,5-dichloro- 

SYN 
Synthesis of lornoxicam (DE2838851)

File:Lornoxicam synthesis.svg

The sulfonation of 2,5-dichlorothiophene (I) with ClSO3H -SOCl2 gives 2,5-dichlorothiophene-3-sulfonic acid chloride (II), which by reaction with methylamine in CHCl3 yields the corresponding methylamide (III). The carboxylation of (III) with butyllithium and CO2 in ether affords 5-chloro-3-(N-methylsulfamoyl)thiophene-2-carboxylic acid (IV), which is esterified with PCl5 and methanol to the methyl ester (V). The condensation of (V) with methyl iodoacetate (VI) by means of NaH in DMF gives 5-chloro-3-[N-(methoxycarbonylmethyl)-N-methylsulfamoyl]thiophene-2-carboxylic acid methyl ester (VII), which is cyclized with sodium methoxide in methanol yielding 6-chloro-4-hydroxy-2-methyl-2H-thieno[2,3-e]-1,2-thiazine-3-carboxylic acid methyl ester 1,1-dioxide (VIII). Finally, this compound is treated with 2-aminopyridine (IX) in refluxing xylene.

Lornoxicam is an NSAID indicated in the treatment of mild to moderate pain, as well as rheumatoid arthritis and osteoarthritis.

Lornoxicam, also known as chlortenoxicam, is a nonsteroidal anti-inflammatory drug (NSAID) of the oxicam class with analgesic (pain relieving), anti-inflammatory and antipyretic (fever reducing) properties. It is available in oral and parenteral formulations.

It was patented in 1977 and approved for medical use in 1997.[1] Brand names include Xefo and Xefocam among others.

Lornoxicam (chlortenoxicam) is a new nonsteroidal anti-inflammatory drug (NSAID) of the oxicam class with analgesic, anti-inflammatory and antipyretic properties. Lornoxicam differs from other oxicam compounds in its potent inhibition of prostaglandin biosynthesis, a property that explains the particularly pronounced efficacy of the drug. Lornoxicam is approved for use in Japan.

Medical uses

Lornoxicam is used for the treatment of various types of pain, especially resulting from inflammatory diseases of the joints, osteoarthritis, surgery, sciatica, and other inflammations.[2]

Non‐Steroidal Anti‐Inflammatory Drugs (NSAIDs) in Metal Complexes and Their Effect at the Cellular Level - Banti - 2016 - European Journal of Inorganic Chemistry - Wiley Online Library

Contraindications

The drug is contraindicated in patients who must not take other NSAIDs, possible reasons including salicylate sensitivity, gastrointestinal bleeding and bleeding disorders, and severe impairment of heart, liver or kidney function. Lornoxicam is not recommended during pregnancy and breastfeeding and is contraindicated during the last third of pregnancy.[2]

Adverse effects

Lornoxicam has side effects similar to other NSAIDs, most commonly mild ones like gastrointestinal disorders (nausea and diarrhea) and headache. Severe but seldom side effects include bleeding, bronchospasms and the extremely rare Stevens–Johnson syndrome.[2]

Interactions

Interactions with other drugs are typical of NSAIDs. Combination with vitamin K antagonists like warfarin increases the risk of bleeding. Combination with ciclosporin can lead to reduced kidney function, and to acute kidney injury in rare cases. Lornoxicam can also increase the adverse effects of lithiummethotrexate and digoxin and its derivatives. The effect of diureticsACE inhibitors and angiotensin II receptor antagonists can be reduced, but this is only relevant in patients with special risks like heart failure. As with piroxicamcimetidine can increase plasma levels but is unlikely to cause relevant interactions.[3]

PAPER

https://www.mdpi.com/2218-0532/71/4/303

str1

PATENT

CN 113480561

The present invention relates to the prepn. of high purity loroxicam.  In particular, the prepn. method comprises a step of taking 6-chloro-4-hydroxy-2-methyl-2H-thieno[2,3-e]-1,2-Me thiazinecarboxylate-1,1-dioxide and 2-amino pyridine is used as the raw material and xylene is used as the solvent undergoes distn. reaction with solid acid catalyst, mixed gas obtained by the distn. reaction is condensed to obtain a condensate and solid acid catalyst is used to adsorb methanol in the condensate and the adsorbed condensate is recycled, filtering and refining to obtain loroxicam.  The present inventive method distills out the methanol produced by the reaction to promote the pos. progress of the reaction and then catalyzes the absorption of methanol by H2SO4/MxOy solid super acid, so that the xylene returned to the reaction system does not contain methanol, which reduces the coking of the reaction, thereby improving product quality and yield.  The prepd. lornoxicam has high purity, which can reach more than 99.9%, reduces the amt. of solvent and also suitable for industrial prodn.

PATENT

CN 112592356

The present invention relates to the prepn. of lornoxicam.  In particular, the prepn. method comprises a step of taking 6-chloro-4-hydroxy-2-methyl-2-H-thieno[2,3-e]-1,2-thiazidecarboxylic acid Me ester-1,1-dioxide and 2-aminopyridine as raw materials, xylene is used as solvent, adding stabilizer, and carrying out aminolysis reaction, the solvent was removed by concn. under reduced pressure, adding org. solvent to make the slurry, filtering and refining to obtain lornoxicam.  The inventive method uses p-toluene sulfonic acid as a stabilizer, while lowering the reaction temp., it promotes the reaction to proceed forward, and improve the product quality and yield; at the same time reduce the amt. of industrial solvents, the post-treatment process is optimized and the cost of the three wastes treatment is reduced.

PATENT

IN 2014CH02116

Example: 1Preparation of 6-chloro-4-hydroxy-l,l-dioxo-l,2-dihydro-lX6-thieno [2,3-e][l,2] thiazine-3-carboxylic acid methyl ester To the mixture of methanol ( 1000 ml) and 5-chloro-3-(methoxy carbonyl methyl sulfamoyl)-thiophene-2-carboxylicacid methyl ester ( 100 g ,0.305 moles), added sodium methoxide solution (200 ml ) at 25-30°C over a period of 30-45 min. The resulting mixture was stirred for 60 min at same temperature; allowed to heat at 65-75°C and stirred for 10-12 hrs. After completion of reaction, methanol was distilled out under reduced pressure to obtained titled residual product which is directly used to next step

(Example-2). Example: – 2:Preparation of 6-chloro-4-hydroxy-2-methyl-l,l-dioxo-l,2-dihydro-U6- thieno[2,3-e][l,2] thiazine-3-carboxylic acid methyl ester 6-chloro-4-hydroxy-1,1 -dioxo-1,2-dihydro-1 X,6-thieno [2,3-e][ 1,2] thiazine-3-carboxylic acid methyl ester was suspended in DM water (500 ml) and cooled to 10-15° C, dimethyl sulphate ( 70 g) was slowly added to the mixture at 10-15°C in 30 min. The reaction mixture was raised to 25-30°C and maintained for 2-3 hours at same temperature. After completion of reaction, mixture was cooled to 10-15°C, methylene dichloride (1600 ml) was added, reaction mixture pH was adjust to 1.0 -2.0 with hydrochloric acid at 10-15° C, stir reaction mixture to separate the layers. The methylene dichloride layer was distilled out completely at below 30°C to get an residue, followed by addition of methanol (60 ml) and distilled out methanol completely under vacuum at below 50°C to get an residue; further it was crystallized by addition of methanol 190 ml and stirred for 30 min at 50-55°C; cooled the reaction mixture at 25-30°C and stirred for 60 min at same temperature. The resultant solid was filtered, washed with methanol (40 ml) and dried at 50-55°C for 4 – 6 hrs to obtain the titled product

Example: 3Preparation of 6-Chloro-4-hydroxy-2-methyl-N-2-pyridinyl-2H-thieno[2,3-e]-l,2-thiazine-3-carboxamide 1,1-dioxide (Lornoxicam) 6-chloro-4-hydroxy-2-methyl-l, 1 -dioxo-1,2-dihydro-l X.6-thieno[2,3-e][l ,2] thiazine-3-carboxylic acid methyl ester ( 50 g 0.161 moles) was suspended in O-xylene (500 ml) and allow to stirred at 70-75°C to obtained clear solution. To this clear solution slowly added the mixture of THF ( 50 ml) solution of 2-Amino pyridine ( 14 g ) and ethyl magnesium bromide 2 molar solution (100 ml) at 70-75°C and allow to stirred for 3-4 hrs at same temperature. After completion of reaction, the dilute hydrochloric acid was added to the mixture at 10-15°C and stirred for 60 min. The resultant solid was filtered, washed with water (100 ml) to obtain crude Lornoxicam.

Example: 4Preparation of 6-Chloro-4-hydroxy-2-methyl-N-2-pyridinyl-2H-thieno[2,3-e)-l,2-thiazine-3-carboxamide 1,1-dioxide (Lornoxicam) 6-chloro-4-hydroxy-2-methyl-l,l-dioxo-l,2-dihydro-R6-thieno[2,3-e][l,2] thiazine-3-carboxylic acid methyl ester ( 50 g 0.161 moles) was suspended in O-xylene (500 ml) and allow to stirred at 70-75°C to obtained clear solution. To this clear solution slowly added the mixture of THF ( 50 ml) solution of 2-Amino pyridine ( 14 g ) and isopropyl magnesium bromide 2 molar solution (100 ml) at 70-75°C and allow to stirred for 3-4 hrs at same temperature. After completion of reaction, the dilute hydrochloric acid was added to the mixture at 10-15°C and stirred for 60 min. The resultant solid was filtered, washed with water (100 ml) to obtain crude Lornoxicam.

Example: 5Purification of Lornoxicam.The crude Lornoxicam was suspended in methanol (500 ml) and cooled to 5-10°C, resulting suspension was basified to pH 11-13 by using sodium hydroxide solution to get clear solution; followed by filtration through hyflo bed; the obtain filtrate was acidified to pH 4.5 – 5.0 with dil. HC1 (1:1) at 5-10°C; stirred the slurry for 30 min. at 5-10°C. The resultant solid was filtered, washed with DM water (100 ml) and dried at 50-55°C to obtained pure Lornoxicam.

PATENT

.EXAMPLES:Preparation of Lornoxicam crudeExample ITo 1200ml o-xylene, 20gm Methyl-6-chloro-4-hydroxy-2-methyl-2//-thieno [2, 3-e] [1, 2] thiazine-3- carboxyate 1,1-dioxide and 6.44gm 2-aminopyridine was added. The reaction mass was stirred under nitrogen atmosphere. Temperature was raised to 140-145°C and maintained for 6hrs. The reaction mass was cooled to 30-35°C and nitrogen was removed. Reaction mass was further stirred for 3hrs- Filtered and washed twice with 50ml of o-xylene. 19.8gm of crude Lornoxicam was obtained. Purification of Lornoxicam crude

Example 219.8gm of crude Lornoxicam was added to the solvent mixture of water (5 vol with respect to Lornoxicam) and methanol (10 vol with respect to Lornoxicam) under stirring. Subsequently 48% sodium hydroxide was added to form a clear solution and 5% activated charcoal was further added. The reaction mass was heated to 50-55°C and stirred for around Ihr followed by filtration through Hyflo. To the filtrate, mixture of hydrochloric acid and water in the ratio of 1:1 was added at 50-55° C, til! the reaction mass reached pH of 2-3, and then stirred for around I hi*. The reaction mass was cooled to room temperature, filtered, and then washed with 1:1 mixture of methanol and water. Purified wet Lornoxicam was dried at 60-65°C for 6-8hrs. 19.1 gm of pure Lornoxicam was obtained. (HPLC purity- 99.95%)

Example 3!7.9gm of crude Lornoxicam (prepared as per example 1) was added to the solvent mixture of water (5 vol with respect to Lornoxicam) and methanol (10 vol with respect to Lornoxicam) under stirring. Subsequently 48% sodium hydroxide was added to form a clear solution, and 5% activated charcoal was further added. The reaction mass was heated to 50-55°C and stirred for around Ihr followed by filtration through Hyflo. To the filtrate, mixture of hydrochloric acid and water in the ratio of 1:1 was added at 50-55° C till the reaction mass reached pH of 2-3, and then stirred for around Ihr. The reaction mass was cooled to room temperature, filtered and then washed with 1:1 mixture of methanol and water. Purified wet Lornoxicam was dried at 60-65°C for 6-8hrs. 17.2 gm of pure Lornoxicam was obtained. (HPLC purity- 99.9%) clear solution and 5% activated charcoal was further added. The reaction mass was heated to 50-55°C and stirred for around lhr followed by filtration through Hyflo. To the filtrate, mixture of hydrochloric acid and water in the ratio of 1:1 was added at 50-55° C, till the reaction mass reached pH of 2-3, and then stirred for around lhr. The reaction mass was cooled to 30-35°C, filtered and then washed with 1:1 mixture of isopropyl alcohol and water. Purified wet Lornoxicam was dried at 60-65°C for 6-8hrs. 4.85 gm of pure Lornoxicam was obtained. (HPLC purity- 99.8%)

Example 55 gm of crude Lornoxicam (prepared as per example 1) was added to the solvent mixture of water (5 vol with respect to Lornoxicam) and ethanol (10 vol with respect to Lornoxicam) under stirring. Subsequently 48% sodium hydroxide was added to form a clear solution, and 5% activated charcoal was further added. The reaction mass was heated to 50-55°C and stirred for around lhr followed by filtration through Hyflo. To the filtrate, mixture of hydrochloric acid and water in the ratio of 1:1 was added at 50-55° C, til! the reaction mass reached pH of 2-3 and then stirred for around lhr. The reaction mass was cooled to 30-35°C and filtered, washed with 1:1 mixture of ethanol and water. Purified wet Lornoxicam was dried at 60-65°C for 6-8hrs. 4.8 gm of pure Lornoxicam was obtained. (HPLC purity- 99.8%)

Example 619.4 gm of crude Lornoxicam (prepared as per example I) was added to the solvent mixture of water (5 vol with respect to Lornoxicam) and methanol (10 vol with respect to Lornoxicam) under stirring. Subsequently 48% sodium hydroxide was added to form a clear solution, and 20% activated charcoal was further added. The reaction mass was stirred for around lhr at room temperature followed by filtration through Hyflo. To the filtrate, mixture of hydrochloric acid and water in the ratio of 1:1 was added till the reaction mass reached pH of 2-3 and then stirred for around 1 hr. The reaction mass was * filtered and washed with 1:1 mixture of methanol and water. Purified wet Lornoxicam was dried at 60-65°C for 6-8hrs. 18.9 gm of pure Lornoxicam was obtained. (HPLC purity- 99.3%).

PATENT

https://www.sciencedirect.com/science/article/abs/pii/S0968089603007624?via%

PATENT

https://patents.google.com/patent/WO2002000167A2/en

References

  1. ^ Fischer J, Ganellin CR (2006). Analogue-based Drug Discovery. John Wiley & Sons. p. 519. ISBN 9783527607495.
  2. Jump up to:a b c Haberfeld H, ed. (2009). Austria-Codex (in German) (2009/2010 ed.). Vienna: Österreichischer Apothekerverlag. Xefo Filmtabletten. ISBN 978-3-85200-196-8.
  3. ^ Klopp T, ed. (2010). Arzneimittel-Interaktionen (in German) (2010/2011 ed.). Arbeitsgemeinschaft für Pharmazeutische Information. ISBN 978-3-85200-207-1.
Clinical data
Trade namesXefo, Xefocam others
AHFS/Drugs.comInternational Drug Names
Pregnancy
category
Not recommended; contraindicated in months 7–9
Routes of
administration
By mouthparenteral
ATC codeM01AC05 (WHO)
Legal status
Legal statusIn general: ℞ (Prescription only)
Pharmacokinetic data
Bioavailability90–100%
Protein binding99%
MetabolismCYP2C9
Elimination half-life3–4 hours
Excretion2/3 liver, 1/3 kidney
Identifiers
showIUPAC name
CAS Number70374-39-9 
PubChem CID5282204
DrugBankDB06725 
ChemSpider10442760 
UNIIER09126G7A
KEGGD01866 
ChEBICHEBI:31783 
CompTox Dashboard (EPA)DTXSID6046133 
ECHA InfoCard100.158.646 
Chemical and physical data
FormulaC13H10ClN3O4S2
Molar mass371.81 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI
  (what is this?)  (verify)
hidevteNonsteroidal anti-inflammatory drugs (NSAIDs) (primarily M01A and M02A, also N02BA)
Pyrazolones /
Pyrazolidines
AminophenazoneAmpyroneAzapropazoneClofezoneDifenamizoleFamprofazoneFeprazoneKebuzoneMetamizoleMofebutazoneMorazoneNifenazoneOxyphenbutazonePhenazonePhenylbutazonePropyphenazoneSulfinpyrazoneSuxibuzone
SalicylatesAspirin (acetylsalicylic acid)#AloxiprinBenorylateCarbasalate calciumDiflunisalDipyrocetylEthenzamideGuacetisalMagnesium salicylateMethyl salicylateSalsalateSalicinSalicylamideSalicylic acid (salicylate)Sodium salicylate
Acetic acid derivatives
and related substances
AceclofenacAcemetacinAlclofenacAmfenacBendazacBromfenacBumadizoneBufexamacDiclofenacDifenpiramideEtodolacFelbinacFenclozic acidFentiazacIndometacinIndometacin farnesilIsoxepacKetorolacLonazolacMofezolacOxametacinProdolic acidProglumetacinSulindacTiopinacTolmetinZomepirac
OxicamsAmpiroxicamDroxicamIsoxicamLornoxicamMeloxicamPiroxicamTenoxicam
Propionic acid derivatives
(profens)
AlminoprofenBenoxaprofenCarprofenDexibuprofenDexketoprofenFenbufenFenoprofenFlunoxaprofenFlurbiprofenIbuprofen#IbuproxamIndoprofenKetoprofenLoxoprofenMiroprofenNaproxenOxaprozinPiketoprofenPirprofenSuprofenTarenflurbilTepoxalinTiaprofenic acidVedaprofenZaltoprofenCOX-inhibiting nitric oxide donatorNaproxcinod
N-Arylanthranilic acids
(fenamates)
AzapropazoneClonixinEtofenamateFloctafenineFlufenamic acidFlunixinGlafenineMeclofenamic acidMefenamic acidMorniflumateNiflumic acidTolfenamic acidFlutiazin
CoxibsApricoxibCelecoxib (+tramadol)CimicoxibDeracoxibEtoricoxibFirocoxibLumiracoxibMavacoxibParecoxibRobenacoxibRofecoxibValdecoxib
OtherAminopropionitrileBenzydamineChondroitin sulfateDiacereinFluproquazoneGlucosamineGlycosaminoglycanHyperforinNabumetoneNimesulideOxaceprolProquazoneSuperoxide dismutase/OrgoteinTenidap
CombinationsIbuprofen/famotidineIbuprofen/hydrocodoneIbuprofen/oxycodoneIbuprofen/paracetamolMeloxicam/bupivacaineNaproxen/diphenhydramineNaproxen/esomeprazole
Items listed in bold indicate initially developed compounds of specific groups. #WHO-EM Withdrawn drugsVeterinary use medications.

//////////LORNOXICAM, Ro-13-9297, TS-110, Anti-inflammatory, analgesic, chlortenoxicam, CCRIS 8589

CN1C(C(=O)NC2=CC=CC=N2)=C(O)C2=C(C=C(Cl)S2)S1(=O)=O

General References

  1. Balfour JA, Fitton A, Barradell LB: Lornoxicam. A review of its pharmacology and therapeutic potential in the management of painful and inflammatory conditions. Drugs. 1996 Apr;51(4):639-57. [Article]
  2. Vane JR: Introduction: mechanism of action of NSAIDs. Br J Rheumatol. 1996 Apr;35 Suppl 1:1-3. [Article]
  3. Radhofer-Welte S, Rabasseda X: Lornoxicam, a new potent NSAID with an improved tolerability profile. Drugs Today (Barc). 2000 Jan;36(1):55-76. [Article]
  4. Skjodt NM, Davies NM: Clinical pharmacokinetics of lornoxicam. A short half-life oxicam. Clin Pharmacokinet. 1998 Jun;34(6):421-8. [Article]
  5. Olkkola KT, Brunetto AV, Mattila MJ: Pharmacokinetics of oxicam nonsteroidal anti-inflammatory agents. Clin Pharmacokinet. 1994 Feb;26(2):107-20. [Article]
  6. Hitzenberger G, Radhofer-Welte S, Takacs F, Rosenow D: Pharmacokinetics of lornoxicam in man. Postgrad Med J. 1990;66 Suppl 4:S22-7. [Article]
  7. Pruss TP, Stroissnig H, Radhofer-Welte S, Wendtlandt W, Mehdi N, Takacs F, Fellier H: Overview of the pharmacological properties, pharmacokinetics and animal safety assessment of lornoxicam. Postgrad Med J. 1990;66 Suppl 4:S18-21. [Article]
  8. Bonnabry P, Leemann T, Dayer P: Role of human liver microsomal CYP2C9 in the biotransformation of lornoxicam. Eur J Clin Pharmacol. 1996;49(4):305-8. [Article]
wdt-15

NEW DRUG APPROVALS

ONE TIME

$10.00

FAROPENEM


Faropenem.svg
ChemSpider 2D Image | 7086 | C12H15NO5S

 

  • Molecular FormulaC12H15NO5S
  • Average mass285.316 Da

Faropenem

7086

(+)-(5R,6S)-6-((1R)-1-Hydroxyethyl)-7-oxo-3-((2R)-tetrahydro-2-furyl)-4-thia-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic Acid

(5R,6S)-6-[(1R)-1-Hydroxyethyl]-7-oxo-3-[(2R)-tetrahydro-2-furanyl]-4-thia-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid
(5R,6S)-6-[(1R)-1-Hydroxyethyl]-7-oxo-3-[(2R)-tetrahydrofuran-2-yl]-4-thia-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid
106560-14-9[RN]
4-Thia-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid, 6-[(1R)-1-hydroxyethyl]-7-oxo-3-[(2R)-tetrahydro-2-furanyl]-, (5R,6S)-
6α-[(R)-1-hydroxyethyl]-2-[(R)-tetrahydrofuran-2-yl]pen-2-em-3-carboxylic acid
4-Oxofenretinide 
4-Oxo-N-(4-hydroxyphenyl)retinamide 
6α-[(1R)-1-hydroxyethyl]-2-[(2R)-tetrahydrofuran-2-yl]-2,3-didehydropenam-3-carboxylic acid 
7305146 [Beilstein]
FaropenemCAS Registry Number: 106560-14-9 
CAS Name: (5R,6S)-6-[(1R)-1-Hydroxyethyl]-7-oxo-3-[(2R)-tetrahydro-2-furanyl]-4-thia-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid 
Additional Names: fropenem; (5R,6S,8R,2¢R)-2-(2¢-tetrahydrofuryl)-6-hydroxyethylpenem-3-carboxylate 
Molecular Formula: C12H15NO5S 
Molecular Weight: 285.32 
Percent Composition: C 50.51%, H 5.30%, N 4.91%, O 28.04%, S 11.24% 
Literature References: Orally active, b-lactamase stable, penem antibiotic.Prepn: M. Ishiguro et al.,EP199446eidem,US4997829 (1986, 1991 both to Suntory); eidem,J. Antibiot.41, 1685 (1988).Pharmacokinetics: A. Tsuji et al.,Drug Metab. Dispos.18, 245 (1990). In vitro antimicrobial spectrum: J. M. Woodcock et al.,J. Antimicrob. Chemother.39, 35 (1997). b-Lactamase stability: A. Dalhoff et al., Chemotherapy (Basel)49, 229 (2003).HPLC determn in plasma: R. V. S. Nirogi et al., Arzneim.-Forsch.55, 762 (2005). Clinical trial in urinary tract infections: S. Arakawa et al.,Nishinihon J. Urol.56, 300 (1994); in bacterial sinusitis: R. Siegert et al., Eur. Arch. Otorhinolaryngol.260, 186 (2003). 
Derivative Type: Sodium salt 
CAS Registry Number: 122547-49-3 
Additional Names: Furopenem 
Manufacturers’ Codes: ALP-201; SUN-5555; SY-5555; WY-49605 
Trademarks: Farom (Daiichi) 
Molecular Formula: C12H15NNaO5S 
Molecular Weight: 308.31 
Percent Composition: C 46.75%, H 4.90%, N 4.54%, Na 7.46%, O 25.95%, S 10.40% 
Properties: [a]D22 +60° (c = 0.10). 
Optical Rotation: [a]D22 +60° (c = 0.10) 
Derivative Type: Daloxate 
CAS Registry Number: 141702-36-5 
CAS Name: (5R,6S)-6-[(1R)-1-Hydroxyethyl]-7-oxo-3-[(2R)-tetrahydro-2-furanyl]-4-thia-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid (5-methyl-2-oxo-1,3-dioxol-4-yl)methyl ester 
Additional Names: faropenem medoxomil 
Manufacturers’ Codes: Bay-56-6854; SUN-208 
Trademarks: Orapem (Replidyne) 
Molecular Formula: C17H19NO8S 
Molecular Weight: 397.40 
Percent Composition: C 51.38%, H 4.82%, N 3.52%, O 32.21%, S 8.07% 
Literature References: Prepn: H. Iwata et al., WO9203442eidemUS5830889 (1992, 1998 both to Suntory). 
Properties: Pale yellow crystals. 
Therap-Cat: Antibacterial (antibiotics). 
Keywords: Antibacterial (Antibiotics); ?Lactams; Penems.

Faropenem is an orally active beta-lactam antibiotic belonging to the penem group.[1] It is resistant to some forms of extended-spectrum beta-lactamase.[2] It is available for oral use.[3]

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

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

Forms

Faropenem was developed by Daiichi Asubio Pharma, which markets it in two forms.

  • The sodium salt faropenem sodium, available under the trade name Farom, has been marketed in Japan since 1997. (CID 636379 from PubChem)
  • The prodrug form faropenem medoxomil[4] (also known as faropenem daloxate) has been licensed from Daiichi Asubio Pharma by Replidyne, which plans to market it in conjunction with Forest Pharmaceuticals. The trade name proposed for the product was Orapem, but company officials recently announced this name was rejected by the FDA.[5]

Clinical use

As of 8 September 2015, Faropenem has yet to receive marketing approval in the United States, and was submitted for consideration by the United States Food and Drug Administration (FDA) on 20 December 2005. The new drug application dossier submitted included these proposed indications:

  • acute bacterial sinusitis
  • community-acquired pneumonia
  • acute exacerbations of chronic bronchitis
  • uncomplicated skin and skin structure infections
  • urinary tract infections

History

The FDA refused to approve faropenem, an antibiotic manufactured by Louisville-based Replidyne. The FDA said the drug was “nonapprovable”, but did not refer to specific safety concerns about the product. The company will have to conduct new studies and clinical trials, lasting an estimated two more years, to prove the drug treats community-acquired pneumonia, bacterial sinusitis, chronic bronchitis, and skin infections.[citation needed]

In India it is available as Farobact 200/300ER CIPLA.

PATENT

https://patents.google.com/patent/WO2008035153A2/enFaropenem is an orally active β-lactam antibiotic belonging to the penem group. Faropenem is chemically known as 6-(l-hydroxyethyl)-7-oxo-3-(oxolan-2-yl)-4-thia-l-azabicyclo[3.2.0]hept-2-ene-2-carboxylicacid. The known forms of Faropenem are Faropenem sodium and the prodrug form, FaropenemMedoxomil (also known as Faropenem Daloxate). In view of the importance of the compound of the formula (I), several synthetic procedures to prepare the compound have been reported.US 4,997,829 provides process for the preparation of faropenem according to the following scheme. The process is exemplified with the allyl protected carboxyl group. One of the process involves the reaction of A- acetoxyazetidinone with tetrahydrothiofuroic acid, condensation with allyl glyoxalate in refluxing benzene, chlorination with thionyl chloride, reaction of triphenylphosphine with lutidine in hot THF, cyclization in refluxing toluene, deprotection of silyl protecting group with tetrabutylammonium fluoride, treating with triphenylphosphine and, treating with sodium 2-ethylhexanoate and (PP^)4Pd to result faropenem sodium. The process exemplified utilizes benzene as solvent, which is not environmentally acceptable. Tetrabutylammonium fluoride was used as desilylating agent that is expensive. Even though the description teaches that optically active compounds can be employed, the examples utilized the dl-compound of tetrahydrothiofuroic acid further requiring resolution.

Figure imgf000003_0001

Methods are provided for the synthesis of series of penem compounds in J Antibiotics 1988, 41(11), 1685-1693. The provided methods utilize sulfonylazetidinone as the starting materials. As one of the procedures gives lesser yield, another procedure was adopted which uses silver salts.Japanese patent, JP2949363 describes a process for deallylation and salt formation with an alkali metal salt of carboxylic acid in the presence of a catalytic amount of palladium complex for the preparation of faropenem.EP410727 describes a process for removing allyl group from a penem compound using cyclic 1,3-diketone such as dimedone.The yield and quality of the final product is always less in the above prior art methods. With the continued research, the present inventors have undertaken extensive studies for developing a process for the preparation of compound of formula (I), which is commercially viable, involves simple techniques such as crystallizations, with improved yields and quality of the product, and with lesser reaction time. None of the prior art suggests or teaches the techniques provided herein.The process is shown in Scheme-I as given below:

Figure imgf000005_0001

One-pot process for the preparation of Faropenem sodium:Sodium salt of R(+)-tetrahydrofuran-2-thiocarboxylic acid (67 g) in aqueous acetone was added slowly to a solution of AOSA (100 g) in acetone (200 mL) and stirred for 3 h at pH 8.0 to 8.5 using sodium bicarbonate solution.After completion of the reaction, the product was extracted with toluene. The combined toluene layer was washed with saturated sodium bicarbonate solution and brine solution. Toluene was removed under vacuum completely and the mass obtained, 3-(l’-tert-butyldimethylsilyloxyethyl)-4-(2′- tetrahydrofuranoylthio)-2-azetidinone was directly taken for next step.3-(r-tert-Butyldimethylsilyloxyethyl)-4-(2′-tetrahydrofuranoylthio)-2- azetidinone obtained was dissolved in toluene (1000 mL) and cooled to -10 to -5 °C under nitrogen. Triethylamine (124 mL) was added to it followed by allyl oxalyl chloride (82 g) at -10 to- 5 0C for 2 h. After completion of the reaction, cold water was added to the mass and washed with dilute hydrochloric acid and sodium bicarbonate solution. Toluene layer was separated and washed with purified water. The toluene layer containing compound of formula (VI) was concentrated under vacuum at 50 to 60 °C and taken for next step as such.Compound of formula (VI) (150 g) was dissolved in triethyl phosphite (150 mL), heated to 60 0C and stirred under nitrogen atmosphere. Toluene (3000 mL) was added, heated to 100 to 110 °C and stirred for 20- 24 h. Toluene was distilled under vacuum completely. Product obtained, allyl (1 ‘R,2″R,5R,6S)-6-(l 5-tert-butyldimethylsilyloxyethyl)-2-(2″-tetrahydrofuranyl) penem-3-carboxylate (VII) was directly taken for next step.Compound (VII) obtained was dissolved in DMF (700 mL) at 30 °C.Ammonium hydrogen difluoride (80 g) and NMP (210 mL) were added and stirred at room temperature for 25 to 35 h. The reaction mass was quenched into a mixture of water-ethyl acetate and stirred at room temperature. The ethyl acetate layer was separated and the aqueous layer extracted with ethyl acetate.  The combined ethyl acetate layer was washed with water followed by saturated sodium bicarbonate solution. The ethyl acetate layer was charcoal treated. The ethyl acetate layer containing allyl (l’R,2″R,5R,6S)-6-(l’-hydroxyethyl)-2-(2″- tetrahydrofuranyl)penem-3-carboxylate (XII) was partially distilled and taken for the next step.The ethyl acetate layer containing compound of formula (XII), Pd/C, sodium bicarbonate and purified water (1000 mL) were taken in an autoclave and maintained 5 to 10 kg pressure of hydrogen gas for 2-5 h. After completion of the reaction the Pd/C was filtered off and ethyl acetate layer separated. The pH of the mass was adjusted to 1.5 and extracted with ethyl acetate. The aqueous layer was extracted again with ethyl acetate twice. The combined ethyl acetate layer was carbon treated. Sodium-2-ethylhexanoate in ethyl acetate was added slowly and stirred. The precipitated title compound was filtered under vacuum, washed with acetone and dried. Dry weight of the product: 65-75 g.Example 9Purification of Faropenem sodiumCrude Faropenem sodium (50 g) was dissolved in purified water (200 mL) at 25-30 0C. The solution was charcoalised. Acetone (1500 mL) was added. The reaction mass was stirred further for 10 min. The precipitated solid was cooled to 0 —2 °C then filtered, washed with acetone and dried at room temperature. Weight of pure Faropenem sodium is 43 to 46 g (Purity 99.95%).Example 9aPurification of Faropenem sodiumCrude Faropenem sodium (50 g) was dissolved in purified water (200 mL) at 25-30 °C. Acetone (150O mL) was added. The reaction mass was stirred further for 10 min. The precipitated solid was cooled to 0-2 °C then filtered, washed with acetone and dried at room temperature. Weight of pure Faropenem sodium is 43 to 46 g (Purity 99.95%).

PATENT

https://patents.google.com/patent/CN103880864B/enFaropenem sodium is developed by Japanese Suntory companies, and first penemss antibiosis in listing in 1997 Element, it are similar to the several carbapenem antibiotics for listing, strong with has a broad antifungal spectrum, antibacterial activity, to beta-lactamase Stably, the features such as also having good action to extended spectrumβ-lactamase producing strains, citrobacter, enterococcus and anaerobe etc.. It is first orally active, penems antibiotics stable to beta-lactamase in the world so far.Its structural formula As follows: 
Report about Faropenem sodium preparation method is a lot, mainly has several as follows:1st, J. Antibiotics 1988, the method that reports in 41,1685, see below row reaction equation: 
Acyl group substitution reaction is carried out in the basic conditions with 4-AA and three beneze methane thiols and obtains thio trityl as protecting group Aza cyclo-butanone, then when 2-TETRAHYDROFUROYL chlorine is connected with lactams, using silver nitrate as condensing agent, but nitric acid Silver is expensive, and cost is too high, while the silver chloride for generating is difficult to filter, is not suitable for large-scale production.2nd, the classical preparation method of United States Patent (USP) US4997829 report:There is acyl with (R) tetrahydrofuran -2- thiocarboxylic acids Base substitution reaction generates thioesters, then through condensation, chlorine replacement, intramolecular Witting cyclization, slough hydroxyl protecting group and carboxylic Base protection group obtains product, and this synthetic route yield is very low, while side chain is thio-compoundss, abnormal smells from the patient is extremely smelly, and prepares complexity, There is-fixed harm to human body and environment.It is also required in chloro building-up process using pungent thionyl chloride, these factors are all It is unfavorable for industrialized production 
3rd, the method that reports in Chinese patent CN1314691 is as follows: 
Said method route is shorter, is produced using one kettle way, more convenient.But said method is related to some other salt such as acetate using heavy metal palladium in last operation The deprotecting regent of compound and triphenyl phosphorus together as pi-allyl, metal palladium reagent is expensive, while triphenyl phosphorus are most More difficult removing in step afterwards, increases operation difficulty, affects product quality.Allyloxy is used easily to produce as protection group simultaneously A kind of double bond olefinic polymerization species impurity of life, affects product quality, reduces yield.Embodiment one(R) tetrahydrofuran -2- thiocarboxylic acids (198g, 1.5 mol) are put in 3L reaction bulbs, plus 1 mol/L hydrogen-oxygens Change sodium body lotion (I.5 L) to be adjusted at 5 DEG C of pH 9- 10,0-, Deca 4AA(287g, 1. 0mo l) acetone (1 L) Solution, drop are finished, and are adjusted to pH 8 or so, 2 h of room temperature reaction with 1 mol/L sodium hydroxide. and add water (500 ml) dilution, second Acetoacetic ester (600 ml x3) is extracted, and merges organic layer, successively with 5 % sodium bicarbonate solutions (300 ml x 2) and water (300 m1 x 2) is washed, and anhydrous sodium sulphate is dried, and is filtered, and filtrate concentrates, and obtains pale yellow oil (about 360 g), directly Input the next step.Embodiment twoThe mixing of concentrated solution as obtained above, triethylamine (l70g, 1.7 mol) and dichloromethane (1.5 L), 0-5 DEG C Deca chlorine oxalic acid is finished to p-Nitrobenzyl (414.1 g, 1 .7 mo l), drop, and equality of temperature reacts 2 h, and add water (1 L) dilution, Extracted with dichloromethane (500 ml x 4), merge organic layer, molten with water (300m1 x 2) and 5 % sodium bicarbonate successively Liquid (300 m1 x 2) is washed, anhydrous sodium sulfate drying, is filtered, and concentration obtains pale yellow oil (about 530g), direct plunges into The next step..Embodiment threeAbove-mentioned gained grease, dimethylbenzene (4L) and NSC 5284 (500ml) are mixed, heating reflux reaction 5h , reduce pressure and boil off dimethylbenzene and NSC 5284, residue ethyl acetate-hexane (1:5,1 L) recrystallization, obtain yellowish Color solid (334.3g, 61%, in terms of 4AA).Example IVAbove-mentioned solid (0.60 mol of 330g.) is dissolved in methanol (2 L), adds 1.0M hydrochloric acid (0.4 L), adds palladium carbon (15.0 g), hydrogen is passed through, 40 DEG C of stirrings, response time are 16 h, and the pressure of system is 4atm, after reaction terminates, crosses and filters Catalyst is removed, is concentrated.Embodiment fiveThe product obtained after above-mentioned concentration is dissolved in tetrahydrofuran 600ml, the 2 ethyl hexanoic acid sodium of 100.0g is added Tetrahydrofuran(200ml)And water(200 ml)Mixed solution, 2 h are stirred at room temperature, have faint yellow solid generate, filter, be method Faropenem crude product 147.0g.Embodiment sixBy above-mentioned solid deionized water(2200ml)Acetone is slowly added under dissolved solution, stirring to start to become to solution Muddiness, when about adding acetone 750ml, solution starts to become cloudy, and stops adding, and continues stirring and allows its crystallize overnight, sucking filtration, acetone Washing, dries, and obtains the Faropenem sodium fine work 125.0g of white.

Syn

AU 8654460; EP 0199446; JP 1994128267; US 4997829

This compound is prepared by several related ways: 1) The reaction of silylated azetidinone (I) with tetrahydrofuran-2-thiocarboxylic acid (II) by means of NaOH in THF – water gives the azetidinone thioester (III), which is condensed with allyl glyoxylate in refluxing benzene yielding the hydroxyester (IV). The reaction of (IV) with SOCl2 affords the chloroester (V), which by reaction with triphenylphosphine by means of lutidine in hot THF is converted into the phosphoranylidene derivative (VI). The elimination of the silyl protecting group of (VI) with tetrabutylammonium fluoride gives the azetidinone (VII), which is cyclized in refluxing toluene yielding the (5R,6S)-6-[1(R)-hydroxyethyl]-2-[2(R)-tetrahydrofuryl]penem-3-carboxyli c acid allyl ester (VIII). Finally, this compound is hydrolyzed with triphenylphosphine, sodium 2-ethylhexanoate and Pd-tetrakis(triphenylphosphine). 2) The condensation of the silver salt of protected azetidinone (IX) with tetrahydrofuran-2(R)-carbonyl chloride (X) also yields the phosphoranylidene salt (VI). 3) Phosphoranylidene ester (VI) can also be cyclized first in refluxing benzene yielding the silylated penem ester (XI), which is deprotected with tetrabutylammonium fluoride to (VIII). 4) The hydrolysis of allyl ester (VIII) to the final product can also be performed with paladium tetrakis(triphenylphosphine) and sodium 4-(methoxycarbonyl)-5,5-dimethylcyclohexane-1,3-dione enolate in several different solvents such as methyl acetate, ethylacetate, tetrahydrofuran, dioxane, sec-butanol, acetonitrile, acetone, 2-butanone, 1,2-dichloroethane, chlorobenzene, toluene, or ethylene glycol dimethyl ether. 5) The preceding hydrolysis can also be performed with triphenylphosphine and paladium tetrakis(triphenylphosphine) with sodium propionate, sodium acetate or sodium lactate in tetrahydrofuran or acetone.

Treatment of the silylated azetidinone (I) with tritylmercaptan affords the tritylsulfanyl-azetidinone (II), which is converted into the silver salt (III) by reaction with AgNO3. Compound (III) is coupled with tetrahydrofuran-2(R)-carbonyl chloride (IV) — obtained by treatment of carboxylic acid (V) with thionyl chloride — providing the azetidinone thioester (VI). Coupling of azetidinone (VI) with allyl oxalyl chloride (VII) in CH2Cl2 by means of Et3N, followed by intramolecular Wittig cyclization by means of triethyl phosphite in refluxing xylene, affords penem (VIII). Alternatively, compound (VIII) can also be obtained as follows: Substitution of phenyl sulfonyl group of azetidinone (X) by tritylmercaptan by means of NaOH in acetone/water provides tritylsulfanyl-azetidinone (XI), which is condensed with allyl oxalyl chloride (VII) by means of DIEA in CH2Cl2 to give the oxalyl amide (XII). Compound (XII) is then treated with AgNO3 and pyridine in acetonitrile, providing the silver mercaptide (XIII), which is acylated with tetrahydrofuran-2(R)-carbonyl chloride (IV) in acetonitrile to afford the penem precursor (XIV). Penem (VIII) is obtained by intramolecular Wittig cyclization of (XIV) with P(OEt)3 in refluxing xylene. Finally, faropenem sodium can be obtained by removal of the tbdms protecting group of (VIII) by means of either Et3N tris(hydrogen fluoride) in ethyl acetate or tetrabutylammonium fluoride (TBAF) and HOAc in THF to give compound (IX). This is followed by allyl ester group removal of (IX), which can be performed under several different conditions: i) triphenylphosphine, sodium 2-ethylhexanoate and palladium tetrakis(triphenylphosphine); ii) palladium tetrakis(triphenylphosphine) and sodium 4-(methoxycarbonyl)-5,5-dimethylcyclohexane-1,3-dione enolate in several different solvents such as methyl acetate, ethyl acetate, tetrahydrofuran, dioxane, sec-butanol, acetonitrile, acetone, 2-butanone, 1,2-dichloroethane, chlorobenzene, toluene or ethylene glycol dimethyl ether; iii) triphenylphosphine and palladium tetrakis(triphenylphosphine) with sodium propionate, sodium acetate or sodium lactate in tetrahydrofuran or acetone; or iv) palladium acetate in the presence of P(OBu)3 and sodium propionate in THF.

Treatment of the silylated azetidinone (I) with tritylmercaptan affords the tritylsulfanylazetidinone (II), which by reaction with AgNO3 is converted into the silver salt (III). Compound (III) is coupled with tetrahydrofuran-2(R)-carbonyl chloride (IV) ?obtained by treatment of carboxylic acid (V) with thionyl chloride ?to provide the azetidinone thioester (VI). Alternatively, compound (VI) can be obtained by condensation of tetrahydrofuran-2(R)-thiocarboxylic S-acid (VII) ?obtained by treatment of carboxylic acid (V) with hydrogen sulfide ?with silylated azetidinones (I) or (VIII) by means of NaOH in THF/water. Condensation of azetidinone thioester (VI) with allyl glyoxylate (IX) in refluxing benzene gives the hydroxy ester (X), which is treated with SOCl2 to yield the chloro ester (XI). Reaction of compound (XI) with triphenylphosphine and lutidine in hot THF provides the phosphoranylidene derivative (XII), which is converted into (5R,6S)-6-[1(R)-hydroxyethyl]-2-[2(R)-tetrahydrofuryl]penem-3-carboxylic acid allyl ester, faropenem allyl ester (XIII) by removal of the silyl protecting group with tetrabutylammonium fluoride, followed by cyclization in refluxing toluene. Compound (XII) can also be obtained by condensation of the silver salt of protected azetidinone (XIV) with tetrahydrofuran-2(R)-carbonyl chloride (V).

Alternatively, faropenem allyl ester (XIII) can also be prepared by cyclization of compound (XII) in refluxing benzene to yield silylated penem allyl ester (XV), which is then deprotected with either tetrabutylammonium fluoride in AcOH or triethylamine tris(hydrogen fluoride) in methyl isobutyl ketone or toluene. Penem (XV) can also be synthesized by several related ways: a) By coupling of azetidinone (VI) with allyl oxalyl chloride (XVI) in CH2Cl2 by means of Et3N, followed by intramolecular Wittig cyclization by means of triethyl phosphite in refluxing xylene. b) Substitution of phenyl sulfonyl group of azetidinone (VIII) by tritylmercaptan by means of NaOH in acetone/water provides tritylsulfanyl-azetidinone (II), which is condensed with allyl oxalyl chloride (XVI) by means of DIEA in CH2Cl2 to give the oxalyl amide (XVII). Compound (XVII) is then treated with AgNO3 and pyridine in acetonitrile to provide the silver mercaptide (XVIII), which is acylated with tetrahydrofuran-2(R)-carbonyl chloride (IV) in acetonitrile to afford the penem precursor (XIX). Finally, compound (XV) is obtained by intramolecular Wittig cyclization of (XX) with P(OEt)3 in refluxing xylene.

Hydrolysis of faropenem allyl ester (XIII) to faropenem sodium (XX) can be performed under several different conditions: i) triphenylphosphine, sodium 2-ethylhexanoate and palladium tetrakis(triphenylphosphine); ii) palladium tetrakis(triphenylphosphine) and sodium 4-(methoxycarbonyl)- 5,5-dimethylcyclohexane-1,3-dione enolate in several different solvents such as methyl acetate, ethyl acetate, tetrahydrofuran, dioxane, sec-butanol, acetonitrile, acetone, 2-butanone, 1,2-dichloroethane, chlorobenzene, toluene, or ethylene glycol dimethyl ether; iii) triphenylphosphine and palladium tetrakis(triphenylphosphine) with sodium propionate, sodium acetate or sodium lactate in tetrahydrofuran or acetone; and iv) palladium acetate in the presence of P(OBu)3 and sodium propionate in THF. Finally, faropenem daloxate can be directly obtained from faropenem sodium (XX) by esterification with 4-(iodomethyl)-5-methyl-1,3-dioxol-2-one (XXI) in DMF.

PATENT

https://patents.google.com/patent/CN103059046A/enFaropenem (Faropenem), chemistry (5R, 6S)-6-[(1R)-hydroxyethyl by name]-2-[(2R)-and tetrahydrofuran (THF)] penem-3-carboxylic acid list sodium salt, by the first exploitation listing in 1997 years of Japanese Suntory company.This medicine is a kind of atypical beta-lactam penems antibiotics, has very strong anti-microbial activity, especially to the anti-microbial activities of the anerobes such as the gram positive organisms such as golden Portugal bacterium, penicillin-fast streptococcus pneumoniae, streptococcus faecium and bacteroides fragilis apparently higher than existing cynnematin, anti-gram-negative bacteria is active similar to oral cephalosporin, and is stable to various β-lactamases.Various clinical studyes show that this medical instrument has clinical effectiveness good, safe, the advantage that renal toxicity and neurotoxicity are little.Its structural formula is as follows: 
For synthesizing of Faropenem, existing many reports in the prior art, for example CN101125857A has reported following synthetic route: 
Take (3R, 4R)-3-[(R)-1-tert-butyl dimethyl silica ethyl]-4-[(R)-and acetoxyl group] nitrogen heterocyclic din-2-ketone is as starting raw material, and warp gets intermediate compound I with R-(+)-sulfo-tetrahydrofuran (THF)-2-formic acid condensation; Intermediate compound I is carried out acylation reaction with monoene propoxy-oxalyl chloride under the catalysis of alkali, get intermediate II; Intermediate II cyclization under the effect of triethyl-phosphite gets intermediate III; Intermediate III is sloughed hydroxyl protecting group through the effect of tetrabutylammonium, gets intermediate compound IV; Intermediate compound IV decarboxylize protecting group under [four (triphenylphosphine)] palladium and triphenylphosphine effect gets Faropenem.Find that after deliberation the method for the present synthetic Faropenem of reporting is all similar with the disclosed method of above-mentioned CN101125857A, all need remove in two steps the protecting group of hydroxyl and carboxyl, reaction scheme is longer.When removing above-mentioned protecting group, need to use a large amount of tetrabutylammonium and [four (triphenylphosphine)] palladium and triphenylphosphine; these reagent costs are high, toxicity is large; be unfavorable for large industrial production; and can introduce the heavy metal palladium; so that the heavy metal remnants in the Faropenem exceed standard, be not suitable for the production of bulk drug.And when adopting aforesaid method deprotection base, the yield in per step only can reach 60%-75%, has further increased production cost.Embodiment 6The preparation of FaropenemWith intermediate 3(364.5g, 0.8mol) use the 700mL acetic acid ethyl dissolution, to open and stir, 0 ℃ of lower dropping with the 36g trifluoroacetic acid after the dilution of 100mL ethyl acetate dripped off in 1 hour, 0 ℃ of lower reaction 2h that continues.Stopped reaction stirs the sodium bicarbonate aqueous solution of lower dropping 5%, until reaction solution pH is neutral.Emit water layer from the reactor lower end, discard.In reactor, add gradually the ethanolic soln of sodium bicarbonate, until till no longer including solid and separating out.Suction filtration, filter cake gets white solid powder 230g(productive rate 93.7% with acetone-water (10:3, v/v) recrystallization), M.P. 163-164 ℃, detect through HPLC, purity is 99.8%Reference examples 1(5R, 6S)-6-[(R)-1-hydroxyethyl]-2-[(R)-and the 2-TETRAHYDROFUROYL sulfenyl] preparation of penem-3-carboxylic acid propyleneWith (5R, 6S)-6-[(R)-the 1-tert-butyl dimethyl silica ethyl]-2-[(R)-and the 2-TETRAHYDROFUROYL sulfenyl] penem-3-carboxylic acid propylene (150g, 0.342mol) and ammonium bifluoride (59.5g, 1.025mmol) add successively among the 400mL DMF, 55~60 ℃ were reacted 5 hours, stopped reaction, suction filtration, filtrate adds water 800ml, uses ethyl acetate extraction, and organic phase is washed with 5% sodium hydrogen carbonate solution, anhydrous sodium sulfate drying, concentrated, gained incarnadine oily matter gets yellow solid 73g through the petrol ether/ethyl acetate recrystallization, yield 66%.Reference examples 2The preparation of Faropenem(the 5R that reference examples 1 is prepared, 6S)-6-[(R)-the 1-hydroxyethyl]-2-[(R)-and the 2-TETRAHYDROFUROYL sulfenyl] penem-3-carboxylic acid propylene (73g, 0.224mol), 6.5g triphenylphosphine, 6.5g [four (triphenylphosphine)] palladium adds among the 500mL methylene dichloride l successively, the ethyl acetate solution that adds the 2 ethyl hexanoic acid sodium preparation of 500mL 0.5M, stirring at room 1 hour, stopped reaction adds 15mL water in reaction solution, stir 30min, suction filtration, this solid is dissolved in the 100mL water again, adds decolorizing with activated carbon 30min, filter, filtrate adds in the 500mL acetone, place crystallization, get Faropenem 66g, yield 96%.Find that by contrast the total recovery that two steps of reference examples remove hydroxyl and carboxyl-protecting group only has about 63.4%, and single stage method of the present invention removes the yield of hydroxyl and carboxyl-protecting group and can reach more than 90%.Preparation method of the present invention can the one-step removal hydroxyl and carboxyl on protecting group, shortened the production cycle, the deprotecting regent cost is low, toxicity is little, can not cause heavy metal remaining, and have higher reaction yield, is fit to very much the industrial production of raw material medicine.

Patent

Publication numberPriority datePublication dateAssigneeTitleCN1939924A *2006-09-082007-04-04鲁南制药集团股份有限公司Industrial production of Fallopeinan sodiumWO2008035153A2 *2006-08-022008-03-27Orchid Chemicals & Pharmaceuticals LimitedProcess for the preparation of beta-lactam antibioticCN103059046A *2013-01-282013-04-24苏州二叶制药有限公司Preparation method of faropenemFamily To Family CitationsCN100522975C *2007-08-232009-08-05东北制药集团公司沈阳第一制药厂Method for preparing faropenemPublication numberPriority datePublication dateAssigneeTitleCN1884284A *2005-06-212006-12-27浙江金华康恩贝生物制药有限公司Process for the preparation of sodium faropenemCN1939924A *2006-09-082007-04-04鲁南制药集团股份有限公司Industrial production of Fallopeinan sodiumCN101125857A *2007-08-232008-02-20东北制药集团公司沈阳第一制药厂Method for preparing faropenemWO2008035153A2 *2006-08-022008-03-27Orchid Chemicals & Pharmaceuticals LimitedProcess for the preparation of beta-lactam antibiotic

Publication numberPriority datePublication dateAssigneeTitle

EP0410727A1 *1989-07-261991-01-30Suntory LimitedProcesses for removing allyl groupsUS4997829A *1985-03-091991-03-05Suntory LimitedPenem compounds, and use thereofEP0574940A1 *1992-06-181993-12-22Tanabe Seiyaku Co., Ltd.Method for removing the protecting group for carboxyl groupWO2007039885A1 *2005-10-052007-04-12Ranbaxy Laboratories LimitedA process for the preparation of faropenemFamily To Family Citations 
Publication numberPriority datePublication dateAssigneeTitleCN102964357A *2012-11-112013-03-13苏州二叶制药有限公司Faropenem sodium and tablet thereofCN103059046A *2013-01-282013-04-24苏州二叶制药有限公司Preparation method of faropenemCN103880864A *2014-03-252014-06-25江苏正大清江制药有限公司Method for synthesizing faropenem sodiumCN104086516A *2014-07-182014-10-08成都樵枫科技发展有限公司Synthetic method of R-(+)-sulfotetrahydrofuran-2-formic acidCN101941981B *2009-07-032015-01-21湖南华纳大药厂有限公司Catalyst composition and method for preparing faropenem sodiumCN106860405A *2015-12-142017-06-20山东新时代药业有限公司A kind of faropenem sodium granules and preparation method thereofCN108840877A *2018-06-122018-11-20赤峰迪生药业有限责任公司A kind of preparation method of oxygen cephalosporin intermediate 

References

  1. ^ Critchley IA, Brown SD, Traczewski MM, Tillotson GS, Janjic N (December 2007). “National and regional assessment of antimicrobial resistance among community-acquired respiratory tract pathogens identified in a 2005-2006 U.S. Faropenem surveillance study”Antimicrob. Agents Chemother51 (12): 4382–9. doi:10.1128/AAC.00971-07PMC 2168020PMID 17908940.
  2. ^ Mushtaq S, Hope R, Warner M, Livermore DM (May 2007). “Activity of faropenem against cephalosporin-resistant Enterobacteriaceae”J. Antimicrob. Chemother59 (5): 1025–30. doi:10.1093/jac/dkm063PMID 17353220.
  3. ^ Milazzo I, Blandino G, Caccamo F, Musumeci R, Nicoletti G, Speciale A (March 2003). “Faropenem, a new oral penem: antibacterial activity against selected anaerobic and fastidious periodontal isolates”J. Antimicrob. Chemother51 (3): 721–5. doi:10.1093/jac/dkg120PMID 12615878.
  4. ^ Gettig JP, Crank CW, Philbrick AH (January 2008). “Faropenem medoxomil”Ann Pharmacother42 (1): 80–90. doi:10.1345/aph.1G232PMID 18094341. Archived from the original on 2013-02-03.
  5. ^ (Q1 06 Investor Conf Call)(CID 6918218 from PubChem)

External links

Clinical data
AHFS/Drugs.comInternational Drug Names
Routes of
administration
Oral
ATC codeJ01DI03 (WHO)
Identifiers
CAS Number106560-14-9 
PubChem CID65894
ChemSpider59303 
UNIIF52Y83BGH3
ChEBICHEBI:51257 
ChEMBLChEMBL556262 
CompTox Dashboard (EPA)DTXSID0046430 
Chemical and physical data
FormulaC12H15NO5S
Molar mass285.31 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI
  (what is this?)  (verify)

///////////Faropenem, ALP-201, SUN-5555, SY-5555, WY-49605, ANTIBACTERIALS, DIICHI, Daiichi Asubio Pharma

wdt-14

NEW DRUG APPROVALS

ONE TIME

$10.00

AUPM 170, CA 170, PD-1-IN-1


str1
 https://www.nature.com/articles/s42003-021-02191-1
str1
str1

(2S,3R)-2-(3-((S)-3-amino-1-(3-((R)-1-amino-2-hydroxyethyl)-1,2,4-oxadiazol-5-yl)-3-oxopropyl)ureido)-3-hydroxybutanoic acid

CA-170
GLXC-15291
str1
PD-1-IN-1 Chemical Structure
Molecular Weight (MW) 360.33
Formula C12H20N6O7
CAS No. 1673534-76-3

N-[[[(1S)-3-Amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]amino]carbonyl]-L-threonine

L-Threonine, N-[[[(1S)-3-amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]amino]carbonyl]-

 AUPM 170, CA 170, AUPM-170, CA-170, PD-1-IN-1

Novel inhibitor of programmed cell dealth-1 (PD-1)

CA-170 (also known as AUPM170 or PD-1-IN-1) is a first-in-class, potent and orally available small molecule inhibitor of the immune checkpoint regulatory proteins PD-L1 (programmed cell death ligand-1), PD-L2 and VISTA (V-domain immunoglobulin (Ig) suppressor of T-cell activation (programmed death 1 homolog; PD-1H). CA-170 was discovered by Curis Inc. and has potential antineoplastic activities. CA-170 selectively targets PD-L1 and VISTA, both of which function as negative checkpoint regulators of immune activation. Curis is currently investigating CA-170 for the treatment of advanced solid tumours and lymphomas in patients in a Phase 1 trial (ClinicalTrials.gov Identifier: NCT02812875).

References: www.clinicaltrials.gov (NCT02812875); WO 2015033299 A1 20150312.

Aurigene Discovery Technologies Limited INNOVATOR

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

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

CURIS AND AURIGENE ANNOUNCE AMENDMENT OF COLLABORATION FOR THE DEVELOPMENT AND COMMERCIALIZATION OF CA-170

PRESS RELEASE

https://www.aurigene.com/curis-and-aurigene-announce-amendment-of-collaboration-for-the-development-and-commercialization-of-ca-170/

Curis and Aurigene Announce Amendment of Collaboration for the Development and Commercialization of CA-170

– Aurigene to fund and conduct a Phase 2b/3 randomized study of CA-170 in patients with non-squamous non-small cell lung cancer (nsNSCLC) –

– Aurigene to receive Asia rights for CA-170; Curis entitled to royalty payments in Asia –

LEXINGTON, Mass., February 5, 2020 /PRNewswire/ — Curis, Inc. (NASDAQ: CRIS), a biotechnology company focused on the development of innovative therapeutics for the treatment of cancer, today announced that it has entered into an amendment of its collaboration, license and option agreement with Aurigene Discovery Technologies, Ltd. (Aurigene). Under the terms of the amended agreement, Aurigene will fund and conduct a Phase 2b/3 randomized study evaluating CA-170, an orally available, dual
inhibitor of VISTA and PDL1, in combination with chemoradiation, in approximately 240 patients with nonsquamous
non-small cell lung cancer (nsNSCLC). In turn, Aurigene receives rights to develop and commercialize CA-170 in Asia, in addition to its existing rights in India and Russia, based on the terms of the original agreement. Curis retains U.S., E.U., and rest of world rights to CA-170, and is entitled to receive royalty payments on potential future sales of CA-170 in Asia.

In 2019, Aurigene presented clinical data from a Phase 2a basket study of CA-170 in patients with multiple tumor types, including those with nsNSCLC. In the study, CA-170 demonstrated promising signs of safety and efficacy in nsNSCLC patients compared to various anti-PD-1/PD-L1 antibodies.

“We are pleased to announce this amendment which leverages our partner Aurigene’s expertise and resources to support the clinical advancement of CA-170, as well as maintain our rights to CA-170 outside of Asia,” said James Dentzer, President and Chief Executive Officer of Curis. “Phase 2a data presented at the European Society for Medical Oncology (ESMO) conference last fall supported the potential for CA-170 to serve as a therapeutic option for patients with nsNSCLC. We look forward to working with our partner Aurigene to further explore this opportunity.”

“Despite recent advancements, patients with localized unresectable NSCLC struggle with high rates of recurrence and need for expensive intravenous biologics. The CA-170 data presented at ESMO 2019 from Aurigene’s Phase 2 ASIAD trial showed encouraging results in Clinical Benefit Rate and Prolonged PFS and support its potential to provide clinically meaningful benefit to Stage III and IVa nsNSCLC patients, in combination with chemoradiation and as oral maintenance” said Kumar Prabhash, MD, Professor of Medical Oncology at Tata Memorial Hospital, Mumbai, India.

Murali Ramachandra, PhD, Chief Executive Officer of Aurigene, commented, “Development of CA-170, with its unique dual inhibition of PD-L1 and VISTA, is the result of years of hard-work and commitment by many people, including the patients who participated in the trials, caregivers and physicians, along with the talented teams at Aurigene and Curis. We look forward to further developing CA-170 in nsNSCLC.”

About Curis, Inc.

Curis is a biotechnology company focused on the development of innovative therapeutics for the treatment of cancer, including fimepinostat, which is being investigated in combination with venetoclax in a Phase 1 clinical study in patients with DLBCL. In 2015, Curis entered into a collaboration with Aurigene in the areas of immuno-oncology and precision oncology. As part of this collaboration, Curis has exclusive licenses to oral small molecule antagonists of immune checkpoints including, the VISTA/PDL1 antagonist CA-170, and the TIM3/PDL1 antagonist CA-327, as well as the IRAK4 kinase inhibitor, CA- 4948. CA-4948 is currently undergoing testing in a Phase 1 trial in patients with non-Hodgkin lymphoma.
In addition, Curis is engaged in a collaboration with ImmuNext for development of CI-8993, a monoclonal anti-VISTA antibody. Curis is also party to a collaboration with Genentech, a member of the Roche Group, under which Genentech and Roche are commercializing Erivedge® for the treatment of advanced basal cell carcinoma. For more information, visit Curis’ website at http://www.curis.com.

About Aurigene

Aurigene is a development stage biotech company engaged in discovery and clinical development of novel and best-in-class therapies to treat cancer and inflammatory diseases and a wholly owned subsidiary of Dr. Reddy’s Laboratories Ltd. (BSE: 500124, NSE: DRREDDY, NYSE: RDY). Aurigene is focused on precision- oncology, oral immune checkpoint inhibitors, and the Th-17 pathway. Aurigene currently has several programs from its pipeline in clinical development. Aurigene’s ROR-gamma inverse agonist AUR-101 is currently in phase 2 clinical development under a US FDA IND. Additionally, Aurigene has multiple compounds at different stages of pre-clinical development. Aurigene has partnered with many large and mid-pharma companies in the United States and Europe and has 15 programs  currently in clinical development. For more information, please visit Aurigene’s website at https://www.aurigene.com/

Curis with the option to exclusively license Aurigene’s orally-available small molecule antagonist of programmed death ligand-1 (PD-L1) in the immuno-oncology field

Addressing immune checkpoint pathways is a well validated strategy to treat human cancers and the ability to target PD-1/PD-L1 and other immune checkpoints with orally available small molecule drugs has the potential to be a distinct and major advancement for patients.

Through its collaboration with Aurigene, Curis is now engaged in the discovery and development of the first ever orally bioavailable, small molecule antagonists that target immune checkpoint receptor-ligand interactions, including PD-1/PD-L1 interactions.  In the first half of 2016, Curis expects to file an IND application with the U.S. FDA to initiate clinical testing of CA-170, the first small molecule immune checkpoint antagonist targeting PD-L1 and VISTA.  The multi-year collaboration with Aurigene is focused on generation of small molecule antagonists targeting additional checkpoint receptor-ligand interactions and Curis expects to advance additional drug candidates for clinical testing in the coming years. The next immuno-oncology program in the collaboration is currently targeting the immune checkpoints PD-L1 and TIM3.

In November 2015, preclinical data were reported. Data demonstrated tha the drug rescued and sustained activation of T cells functions in culture. CA-170 resulted in anti-tumor activity in multiple syngeneic tumor models including melanoma and colon cancer. Similar data were presented at the 2015 AACR-NCI-EORTC Molecular Targets and Cancer Therapeutics Conference in Boston, MA

By August 2015, preclinical data had been reported. Preliminary data demonstrated that in in vitro studies, small molecule PD-L1 antagonists induced effective T cell proliferation and IFN-gamma production by T cells that were specifically suppressed by PD-L1 in culture. The compounds were found to have effects similar to anti-PD1 antibodies in in vivo tumor models

 (Oral Small Molecule PD-L1/VISTAAntagonist)

Certain human cancers express a ligand on their cell surface referred to as Programmed-death Ligand 1, or PD-L1, which binds to its cognate receptor, Programmed-death 1, or PD-1, present on the surface of the immune system’s T cells.  Cell surface interactions between tumor cells and T cells through PD-L1/PD-1 molecules result in T cell inactivation and hence the inability of the body to mount an effective immune response against the tumor.  It has been previously shown that modulation of the PD-1 mediated inhibition of T cells by either anti-PD1 antibodies or anti-PD-L1 antibodies can lead to activation of T cells that result in the observed anti-tumor effects in the tumor tissues.  Therapeutic monoclonal antibodies targeting the PD-1/PD-L1 interactions have now been approved by the U.S. FDA for the treatment of certain cancers, and multiple therapeutic monoclonal antibodies targeting PD-1 or PD-L1 are currently in development.

In addition to PD-1/PD-L1 immune regulators, there are several other checkpoint molecules that are involved in the modulation of immune responses to tumor cells1.  One such regulator is V-domain Ig suppressor of T-cell activation or VISTA that shares structural homology with PD-L1 and is also a potent suppressor of T cell functions.  However, the expression of VISTA is different from that of PD-L1, and appears to be limited to the hematopoietic compartment in tissues such as spleen, lymph nodes and blood as well as in myeloid hematopoietic cells within the tumor microenvironment.  Recent animal studies have demonstrated that combined targeting/ blockade of PD-1/PD-L1 interactions and VISTA result in improved anti-tumor responses in certain tumor models, highlighting their distinct and non-redundant functions in regulating the immune response to tumors2.

As part of the collaboration with Aurigene, in October 2015 Curis licensed a first-in-class oral, small molecule antagonist designated as CA-170 that selectively targets PD-L1 and VISTA, both of which function as negative checkpoint regulators of immune activation.  CA-170 was selected from the broad PD-1 pathway antagonist program that the companies have been engaged in since the collaboration was established in January 2015.  Preclinical data demonstrate that CA-170 can induce effective proliferation and IFN-γ (Interferon-gamma) production (a cytokine that is produced by activated T cells and is a marker of T cell activation) by T cells that are specifically suppressed by PD-L1 or VISTA in culture.  In addition, CA-170 also appears to have anti-tumor effects similar to anti-PD-1 or anti-VISTA antibodies in multiple in vivo tumor models and appears to have a good in vivo safety profile.  Curis expects to file an IND and initiate clinical testing of CA-170 in patients with advanced tumors during the first half of 2016.

Jan 21, 2015

Curis and Aurigene Announce Collaboration, License and Option Agreement to Discover, Develop and Commercialize Small Molecule Antagonists for Immuno-Oncology and Precision Oncology Targets

— Agreement Provides Curis with Option to Exclusively License Aurigene’s Antagonists for Immuno-Oncology, Including an Antagonist of PD-L1 and Selected Precision Oncology Targets, Including an IRAK4 Kinase Inhibitor —

— Investigational New Drug (IND) Application Filings for Both Initial Collaboration Programs Expected this Year —

— Curis to issue 17.1M shares of its Common Stock as Up-front Consideration —

— Management to Host Conference Call Today at 8:00 a.m. EST —

LEXINGTON, Mass. and BANGALORE, India, Jan. 21, 2015 (GLOBE NEWSWIRE) — Curis, Inc. (Nasdaq:CRIS), a biotechnology company focused on the development and commercialization of innovative drug candidates for the treatment of human cancers, and Aurigene Discovery Technologies Limited, a specialized, discovery stage biotechnology company developing novel therapies to treat cancer and inflammatory diseases, today announced that they have entered into an exclusive collaboration agreement focused on immuno-oncology and selected precision oncology targets. The collaboration provides for inclusion of multiple programs, with Curis having the option to exclusively license compounds once a development candidate is nominated within each respective program. The partnership draws from each company’s respective areas of expertise, with Aurigene having the responsibility for conducting all discovery and preclinical activities, including IND-enabling studies and providing Phase 1 clinical trial supply, and Curis having responsibility for all clinical development, regulatory and commercialization efforts worldwide, excluding India and Russia, for each program for which it exercises an option to obtain a license.

The first two programs under the collaboration are an orally-available small molecule antagonist of programmed death ligand-1 (PD-L1) in the immuno-oncology field and an orally-available small molecule inhibitor of Interleukin-1 receptor-associated kinase 4 (IRAK4) in the precision oncology field. Curis expects to exercise its option to obtain exclusive licenses to both programs and file IND applications for a development candidate from each in 2015.

“We are thrilled to partner with Aurigene in seeking to discover, develop and commercialize small molecule drug candidates generated from Aurigene’s novel technology and we believe that this collaboration represents a true transformation for Curis that positions the company for continued growth in the development and eventual commercialization of cancer drugs,” said Ali Fattaey, Ph.D., President and Chief Executive Officer of Curis. “The multi-year nature of our collaboration means that the parties have the potential to generate a steady pipeline of novel drug candidates in the coming years. Addressing immune checkpoint pathways is now a well validated strategy to treat human cancers and the ability to target PD-1/PD-L1 and other immune checkpoints with orally available small molecule drugs has the potential to be a distinct and major advancement for patients. Recent studies have also shown that alterations of the MYD88 gene lead to dysregulation of its downstream target IRAK4 in a number of hematologic malignancies, including Waldenström’s Macroglobulinemia and a subset of diffuse large B-cell lymphomas, making IRAK4 an attractive target for the treatment of these cancers. We look forward to advancing these programs into clinical development later this year.”

Dr. Fattaey continued, “Aurigene has a long and well-established track record of generating targeted small molecule drug candidates with bio-pharmaceutical collaborators and we have significantly expanded our drug development capabilities as we advance our proprietary drug candidates in currently ongoing clinical studies. We believe that we are well-positioned to advance compounds from this collaboration into clinical development.”

CSN Murthy, Chief Executive Officer of Aurigene, said, “We are excited to enter into this exclusive collaboration with Curis under which we intend to discover and develop a number of drug candidates from our chemistry innovations in the most exciting fields of cancer therapy. This unique collaboration is an opportunity for Aurigene to participate in advancing our discoveries into clinical development and beyond, and mutually align interests as provided for in our agreement.  Our scientists at Aurigene have established a novel strategy to address immune checkpoint targets using small molecule chemical approaches, and have discovered a number of candidates that modulate these checkpoint pathways, including PD-1/PD-L1. We have established a large panel of preclinical tumor models in immunocompetent mice and can show significant in vivo anti-tumor activity using our small molecule PD-L1 antagonists.  We are also in the late stages of selecting a candidate that is a potent and selective inhibitor of the IRAK4 kinase, demonstrating excellent in vivo activity in preclinical tumor models.”

In connection with the transaction, Curis has issued to Aurigene approximately 17.1 million shares of its common stock, or 19.9% of its outstanding common stock immediately prior to the transaction, in partial consideration for the rights granted to Curis under the collaboration agreement. The shares issued to Aurigene are subject to a lock-up agreement until January 18, 2017, with a portion of the shares being released from the lock-up in four equal bi-annual installments between now and that date.

The agreement provides that the parties will collaborate exclusively in immuno-oncology for an initial period of approximately two years, with the option for Curis to extend the broad immuno-oncology exclusivity.

In addition Curis has agreed to make payments to Aurigene as follows:

  • for the first two programs: up to $52.5 million per program, including $42.5 million per program for approval and commercial milestones, plus specified approval milestone payments for additional indications, if any;
  • for the third and fourth programs: up to $50 million per program, including $42.5 million per program for  approval and commercial milestones, plus specified approval milestone payments for additional indications, if any; and
  • for any program thereafter: up to $140.5 million per program, including $87.5 million per program in approval and commercial milestones, plus specified approval milestone payments for additional indications, if any.

Curis has agreed to pay Aurigene royalties on any net sales ranging from high single digits to 10% in territories where it successfully commercializes products and will also share in amounts that it receives from sublicensees depending upon the stage of development of the respective molecule.
About Immune Checkpoint  Modulation and Programmed Death 1 Pathway

Modulation of immune checkpoint pathways has emerged as a highly promising therapeutic approach in a wide range of human cancers. Immune checkpoints are critical for the maintenance of self-tolerance as well as for the protection of tissues from excessive immune response generated during infections. However, cancer cells have the ability to modulate certain immune checkpoint pathways as a mechanism to evade the immune system. Certain immune checkpoint receptors or ligands are expressed by various cancer cells, targeting of which may be an effective strategy for generating anti-tumor activity. Some immune-checkpoint modulators, such as programmed death 1 (PD-1) protein, specifically regulate immune cell effector functions within tissues. One of the mechanisms by which tumor cells block anti-tumor immune responses in the tumor microenvironment is by upregulating ligands for PD-1, such as PD-L1. Hence, targeting of PD-1 and/or PD-L1 has been shown to lead to the generation of effective anti-tumor responses.
About Curis, Inc.

Curis is a biotechnology company focused on the development and commercialization of novel drug candidates for the treatment of human cancers. Curis’ pipeline of drug candidates includes CUDC-907, a dual HDAC and PI3K inhibitor, CUDC-427, a small molecule antagonist of IAP proteins, and Debio 0932, an oral HSP90 inhibitor. Curis is also engaged in a collaboration with Genentech, a member of the Roche Group, under which Genentech and Roche are developing and commercializing Erivedge®, the first and only FDA-approved medicine for the treatment of advanced basal cell carcinoma. For more information, visit Curis’ website at www.curis.com.

About Aurigene

Aurigene is a specialized, discovery stage biotechnology company, developing novel and best-in-class therapies to treat cancer and inflammatory diseases. Aurigene’s Programmed Death pathway program is the first of several immune checkpoint programs that are at different stages of discovery and preclinical development. Aurigene has partnered with several large- and mid-pharma companies in the United States and Europe and has delivered multiple clinical compounds through these partnerships. With over 500 scientists, Aurigene has collaborated with 6 of the top 10 pharma companies. Aurigene is an independent, wholly owned subsidiary of Dr. Reddy’s Laboratories Ltd. (NYSE:RDY). For more information, please visit Aurigene’s website at http://aurigene.com/.

POSTER

STR3
STR3
STR3

WO2011161699, WO2012/168944, WO2013144704 and WO2013132317 report peptides or peptidomimetic compounds which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signaling pathway.

PATENT

WO 2015033299

Inventors

  • SASIKUMAR, Pottayil Govindan Nair
  • RAMACHANDRA, Muralidhara
  • NAREMADDEPALLI, Seetharamaiah Setty Sudarshan

Priority Data

4011/CHE/2013 06.09.2013 IN

Example 4: Synthesis of Co

str1

The compound was synthesised using similar procedure as depicted in Example 2 for synthesising compound 2 using 
instead of H-Ser(‘Bu)-0’Bu (in synthesis of compound 2b) to yield 0.35 g crude material of the title compound. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 361.2 (M+H)+, HPLC: tR = 12.19 min.

Pottayil Sasikumar

Pottayil Sasikumar

Murali Ramachandra

Murali Ramachandra

REFERENCES

US20150073024

WO2011161699A227 Jun 201129 Dec 2011Aurigene Discovery Technologies LimitedImmunosuppression modulating compounds
WO2012168944A121 Dec 201113 Dec 2012Aurigene Discovery Technologies LimitedTherapeutic compounds for immunomodulation
WO2013132317A14 Mar 201312 Sep 2013Aurigene Discovery Technologies LimitedPeptidomimetic compounds as immunomodulators
WO2013144704A128 Mar 20133 Oct 2013Aurigene Discovery Technologies LimitedImmunomodulating cyclic compounds from the bc loop of human pd1

http://www.curis.com/pipeline/immuno-oncology/pd-l1-antagonist

http://www.curis.com/images/stories/pdfs/posters/Aurigene_PD-L1_VISTA_AACR-NCI-EORTC_2015.pdf

References:

1) https://bmcimmunol.biomedcentral.com/articles/10.1186/s12865-021-00446-4

2) https://www.nature.com/articles/s42003-021-02191-1

3) https://www.esmoopen.com/article/S2059-7029(20)30108-3/fulltext

4) https://www.mdpi.com/1420-3049/24/15/2804

////////Curis, Aurigene,  AUPM 170, CA 170, AUPM-170, CA-170, PD-L1, VISTA antagonist, PD-1-IN-1, phase 2, CANCER

N[C@@H](CO)c1nc(on1)[C@@H](NC(=O)N[C@H](C(=O)O)[C@@H](C)O)CC(N)=O

NEW DRUG APPROVALS

ONE TIME

$9.00

AUR 101


AUR 101

AUR101-201

ANTIINNFLAMATORY

AUR-101, a ROR gamma inverse agonist for autoimmune disorders like psoriasis

AUR-101 is an ROR-gammaT inverse agonist in phase II clinical development at Aurigene for the treatment of patients with moderate-to-severe chronic plaque-type psoriasis.

  • DrugsAUR 101 (Primary)
  • IndicationsPlaque psoriasis
  • FocusAdverse reactions; First in man
  • AcronymsINDUS
  • SponsorsAurigene Discovery Technologies
  • OriginatorAurigene Discovery Technologies
  • ClassAntipsoriatics; Small molecules
  • Mechanism of ActionNuclear receptor subfamily 1 group F member 3 inverse agonists
  • Phase IIPsoriasis
wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

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

AURIGENE ANNOUNCES FIRST PATIENT DOSED WITH AUR101 IN PHASE II STUDY IN PATIENTS WITH MODERATE TO SEVERE PSORIASIS

https://www.aurigene.com/aurigene-announces-first-patient-dosed-with-aur101-in-phase-ii-study-in-patients-with-moderate-to-severe-psoriasis/

PRESS RELEASE

Aurigene Announces First Patient Dosed with AUR101 in Phase II Study in Patients with Moderate to Severe Psoriasis

Bangalore, February 17, 2020 — Aurigene, a development stage biotechnology company, today announced dose administration for the first patient in INDUS-2, a Phase II double blind placebo-controlled three-arm study of AUR101 in patients with moderate to severe psoriasis. AUR101 is an oral small molecule inverse agonist of RORγ and has shown desirable pharmacodynamic modulation of IL-17 and acceptable safety in a completed Phase I human study conducted in Australia.

“The initiation of this Phase II study under a US FDA IND represents a significant milestone for Aurigene, as it marks the first program which Aurigene has led from the bench side to the clinic all by itself,” said Murali Ramachandra, PhD, Chief Executive Officer of Aurigene. “We look forward to producing important clinical data by the end of 2020 to guide our future development plans and demonstrating Aurigene’s unique expertise in conducting Proof-of-Concept studies in a quality and fast-paced manner.”

About AUR101-201 and the Phase II Study of AUR101 in Patients with Moderate to Severe Psoriasis

The purpose of the Phase II multi-center, blinded, placebo-controlled, three-arm study is to evaluate the clinical activity of AUR101 in patients with moderate to severe psoriasis. In two of the arms, AUR101 will be administered twice daily, at 400 mg PO BID and 600 mg PO BID, for 12 weeks. Patients in the third arm will receive matched blinded placebo in a double dummy fashion. The trial is listed at clinicaltrials.gov with identifier NCT04207801.

About Aurigene

Aurigene is a development stage biotech company engaged in discovery and clinical development of novel and best-in-class therapies to treat cancer and inflammatory diseases and a wholly owned subsidiary of Dr. Reddy’s Laboratories Ltd. (BSE: 500124, NSE: DRREDDY,NYSE: RDY). Aurigene is focused on precision- oncology, oral immune checkpoint inhibitors, and the Th-17 pathway. Aurigene currently has several programs from its pipeline in clinical development. Aurigene has also submitted an IND to DCGI, India for a Phase IIb/III trial of CA-170, a dual inhibitor of PD-L1 and VISTA, in non-squamous NSCLC. Additionally, Aurigene has multiple compounds at different stages of pre-clinical development. Aurigene has partnered with many large and mid-pharma companies in the United States and Europe and has 15 programs currently in clinical development. For more information, please visit Aurigene’s website at https://www.aurigene.com/.

CLIP

Signalling of multiple interleukin (IL)-17 family cytokines via IL-17 receptor A drives psoriasis-related inflammatory pathways

https://onlinelibrary.wiley.com/doi/10.1111/bjd.20090

M.A.X. Tollenaere,J. Hebsgaard,D.A. Ewald,P. Lovato,S. Garcet,X. Li,S.D. Pilger,M.L. Tiirikainen,M. Bertelsen,J.G. Krueger,H. Norsgaard,First published: 01 April 2021 https://doi.org/10.1111/bjd.20090Citations: 2Funding sources LEO Pharma A/S funded this study.Conflicts of interest M.A.X.T., J.H., D.A.E., P.L., S.D.P., M.L.T., M.B. and H.N. are employees of LEO Pharma. J.G.K. received grants paid to his institution from Novartis, Pfizer, Amgen, Lilly, Boehringer, Innovaderm, BMS, Janssen, AbbVie, Paraxel, LEO Pharma, Vitae, Akros, Regeneron, Allergan, Novan, Biogen MA, Sienna, UCB, Celgene, Botanix, Incyte, Avillion and Exicure; and personal fees from Novartis, Pfizer, Amgen, Lilly, Boehringer, Biogen Idec, AbbVie, LEO Pharma, Escalier, Valeant, Aurigene, Allergan, Asana, UCB, Sienna, Celgene, Nimbus, Menlo, Aristea, Sanofi, Sun Pharma, Almirall, Arena and BMS.Data Availability Statement The gene array dataset described in this publication has been deposited in NCBI’s Gene Expression Omnibus and is accessible through GEO Series accession number GSE158448 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE158448).

CLOP

https://www.drugdiscoverychemistry.com/Anti-Inflammatories/16

10:35 Small Molecule Inhibitors of RORgamma and IRAK4 for the Treatment of Autoimmune Disorders

Susanta_SamajdarSusanta Samajdar, Ph.D., Director, Medicinal Chemistry, Aurigene Discovery Technologies Limited

Although biologics such as anti-TNFα antibody are fairly successful in the treatment of autoimmune disorders, there is significant unmet need due to heterogeneity in diseases and lack of response to established therapies in some patients. While biologics typically target one cytokine signaling pathway, small molecule therapeutics directed towards intracellular target(s) can interfere in the signaling from multiple cytokines potentially leading to improved response. Development of small molecule oral inhibitors of IRAK4 and RORgamma to target TLR/IL-R and Th17 pathway respectively will be discussed.

PATENT

2448/CHE/2015 15.05.2015 IN

PATENT

PATENT

This application claims the benefit of Indian provisional application number 5641/CHE/2013 filed on 06th December 2013 which hereby incorporated by reference.

PATENT

  • KOTRABASAIAH UJJINAMATADA, Ravi
  • PANDIT, Chetan

 2049005-13-0

2-​Quinolinecarboxamide​, 6-​(2,​6-​dimethyl-​4-​pyrimidinyl)​-​N-​[[4-​(ethylsulfonyl)​phenyl]​methyl]​-​5,​6,​7,​8-​tetrahydro-​6-​methyl-​5-​oxo-​, (6S)​-

Molecular Weight492.59, C26 H28 N4 O4 S

EXAMPLE

PATENT

CLIP

https://www.sciencedirect.com/science/article/abs/pii/S0223523419301011

2013239366 CA 170

wdt-12

NEW DRUG APPROVALS

ONE TIME

$10.00

///////////////////////AUR 101, AURIGENE, ROR, IL-17, PHASE 2, CDSCO, Ravi Ujjinamatada, KOTRABASAIAH UJJINAMATADA Ravi, PANDIT Chetan, AUR101-201, plaque-type psoriasis

IMG_3542.jpg
Ravi Ujjinamatada

Ravi Ujjinamatada

JNJ-A07


str2

JNJ-A07

S + FORM

CAS 2135640-93-4 ROT (+)S

Butanoic acid, 4-[3-[[1-(4-chlorophenyl)-2-[2,3-dihydro-6-(trifluoromethoxy)-1H-indol-1-yl]-2-oxoethyl]amino]-5-methoxyphenoxy]-, (+)-

(+)-4-[3-[[1-(4-Chlorophenyl)-2-[2,3-dihydro-6-(trifluoromethoxy)-1H-indol-1-yl]-2-oxoethyl]amino]-5-methoxyphenoxy]butanoic acid

(+)-4-[3-([(1S)-1-(4-Chlorophenyl)-2-oxo-2-[6-(trifluoromethoxy)-2,3-dihydro-1H-indol-1-yl]ethyl]amino)-5-methoxyphenoxy]butanoic acidMolecular FormulaC28 H26 Cl F3 N2 O6Molecular Weight578.964

REF

Kaptein, S.J.F., Goethals, O., Kiemel, D. et al. A pan-serotype dengue virus inhibitor targeting the NS3–NS4B interaction. Nature (2021). https://doi.org/10.1038/s41586-021-03990-6

09938-scicon2-jnj.jpg

JNJ-018

CAS 2135640-91-2 +/-, R,S

CAS 2135640-92-3 ROT (-)R

Butanoic acid, 4-[3-[[1-(4-chlorophenyl)-2-[2,3-dihydro-6-(trifluoromethoxy)-1H-indol-1-yl]-2-oxoethyl]amino]-5-methoxyphenoxy]-, (-)-

(-)-4-[3-[[1-(4-Chlorophenyl)-2-[2,3-dihydro-6-(trifluoromethoxy)-1H-indol-1-yl]-2-oxoethyl]amino]-5-methoxyphenoxy]butanoic acid

  • Janssen (Originator)
  • Katholieke Universiteit Leuven (Originator)
  • NS4B Protease (Dengue Virus) Inhibitors
  • Serine Protease NS3/Non-Structural Protein NS4B Protease (Dengue Virus) Interaction Inhibitors

A pan-serotype dengue virus inhibitor targeting the NS3–NS4B interaction

https://www.nature.com/articles/s41586-021-03990-6

https://www.nature.com/articles/s41586-021-03990-6#citeas

Abstract

Dengue virus causes approximately 96 million symptomatic infections annually, manifesting as dengue fever or occasionally as severe dengue1,2. There are no antiviral agents available to prevent or treat dengue. Here, we describe a highly potent dengue virus inhibitor (JNJ-A07) that exerts nanomolar to picomolar activity against a panel of 21 clinical isolates that represent the natural genetic diversity of known genotypes and serotypes. The molecule has a high barrier to resistance and prevents the formation of the viral replication complex by blocking the interaction between two viral proteins (NS3 and NS4B), thus revealing a previously undescribed mechanism of antiviral action. JNJ-A07 has a favourable pharmacokinetic profile that results in outstanding efficacy against dengue virus infection in mouse infection models. Delaying start of treatment until peak viraemia results in a rapid and significant reduction in viral load. An analogue is currently in further development.

2-(4-Chlorophenyl)-1-(6-(trifluoromethoxy)indolin-1-yl)-ethanone (1)

127 A mixture of 6-(trifluoromethoxy)indoline ([CAS 959235-95-1], 2 g, 9.84 mmol), 2-(4-chlorophenyl)acetic acid 128 ([CAS 1878-66-6], 1.85 g, 10.8 mmol), HATU (5.6 g, 14.8 mmol) and diisopropylethylamine (4.9 mL, 29.5

129 mmol) in DMF (40 mL) was stirred at room temperature for 12 h. Water was added and the precipitate was

130 filtered off. The residue was taken up with EtOAc. The organic solution was washed with a 10 % aqueous

131 solution of K2CO3, brine, dried over MgSO4, filtered, and the solvent was evaporated under reduced pressure. 132 The residue was purified by chromatography on silica gel (15-40 pm, 80 g, heptane/EtOAc gradient 90/10 to 133 60/40). The pure fractions were combined and the solvent was concentrated under reduced pressure to give 2-(4-

134 chlorophenyl)-1-(6-(trifluoromethoxy)indolin-1-yl)-ethanone 1 (3 g, yield: 86 %).

135 1 H NMR (400 MHz, DMSO-d6) d ppm 7.99 (s, 1 H), 7.37 – 7.41 (m, 2 H), 7.29 – 7.34 (m, 3 H), 6.97 (dd, J = 8.1, 1.3 Hz, 1 H), 4.25 (t, J = 8.6 Hz, 2 H), 3.88 (s, 2 H), 3.18 (t, J = 8.5 Hz, 2 H); 13

136 C NMR (101 MHz, 137 CHLOROFORM-d) δ ppm 168.91, 148.65, 148.63, 144.05, 133.16, 132.26, 130.63, 129.54, 128.93, 124.87, 120.50 (q, J=257.2 Hz), 116.38, 110.83, 77.26, 48.86, 42.52, 27.59; LC-MS: [M+H]+

138 728; purity 99 % (method LCMS2); Melting Point: 116-131 °C (DSC peak: 120.2 °C); HRMS (ESI+) m/z: [M]+ 139 calcd for C17H13ClF3NO2,

140 356.0660; found, 356.0657

141 2-Bromo-2-(4-chlorophenyl)-1- (6-(trifluoromethoxy)indolin-1-yl)ethanone (2)

142 At -78 °C, under nitrogen flow, LiHMDS (1.5 M in THF, 11.2 mL, 16.9 mmol) was added dropwise to a mixture 143 of 1 (3 g, 8.43 mmol) in THF (50 mL). The mixture was stirred for 15 min at -78 °C and a solution of N

144 bromosuccinimide (1.65 g, 9.3 mmol) in THF (30 mL) was added dropwise. After stirring for 2 h at -78 °C, the 145 reaction was quenched with a saturated aqueous solution of NH4Cl. The mixture was extracted with EtOAc. The 146 organic layer was separated, dried over MgSO4, filtered, and the solvent was evaporated under reduced pressure

147 to give 2-bromo-2-(4-chlorophenyl)-1- (6-(trifluoromethoxy)indolin-1-yl)ethanone 2 (3.6 g, yield: 98 %) as an 148 oil. The compound was used without further purification in the next step.

149 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 8.19 (s), 7.52 – 7.57 (m), 7.34 – 7.39 (m), 7.17 (d, J=8.2 Hz), 6.92 (dd, J=8.2, 1.1 Hz), 5.56 (s), 4.37 (td, J=10.1, 6.5 Hz), 4.09 (td, J=10.1, 6.7 Hz), 3.12 – 3.31 (m); 13

150 C NMR

151 (101 MHz, CHLOROFORM-d) δ ppm 164.90 (s), 148.68 (d, J=2.2 Hz), 143.75 (s), 135.46 (s), 133.99 (s), 152 130.52 (s), 129.79 (s), 129.10 (s), 125.01 (s), 117.20 (s), 120.47 (q, J=257.2 Hz), 111.36 (s), 48.88 (s), 46.61 (s), 27.65 (s); LC-MS: [M+H]+ 436; purity 100 % (method LCMS2); HRMS (ESI+) m/z: [M]+ 153 calcd for

154 C17H13O2NBrClF3, 433.9765; found, 433.9764

155 tert-Butyl 4-(3-amino-5-methoxyphenoxy)butanoate (3) 156 To a mechanically stirred solution of tert-butyl 4-bromobutanoate ([CAS 110661- 5 91-1], 42.3 g, 0.19 mol) in

157 DMF (600 mL) was added in portions a solid mixture of 3-amino-5-methoxyphenol ([CAS 162155-27-3], 26.4 158 g, 0.19 mol) and Cs2CO3 (123.6 g, 0.379 mol). The reaction mixture was stirred at 60 °C for 65 h, and allowed to

159 reach room temperature. The mixture was poured out into water (2.5 L). The product was extracted with Et2O (2 160 x). The combined organic layers were washed with brine, dried over MgSO4, and filtered. The solvent was

161 evaporated under reduced pressure, and then co-evaporated with toluene. The residue was purified by normal 162 phase HPLC (Stationary phase: silica gel 60A 25-40 pm (Merck), Mobile phase: gradient EtOAc/heptane 20/80 163 to 60/40), yielding tert-butyl 4-(3-amino-5-methoxyphenoxy)butanoate 3 as an oil (27 g, yield: 50 %).

164 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 5.89 – 5.92 (m), 5.86 (d, J=2.2 Hz), 3.92 (t, J=6.2 Hz), 3.73 (s), 3.66 (br s), 2.40 (t, J=7.4 Hz), 1.98 – 2.08 (m), 1.45 (s); 13

165 C NMR (101 MHz, CHLOROFORM-d) δ ppm 172.61 166 (s), 161.69 (s), 161.02 (s), 148.35 (s), 94.33 (s), 93.89 (s), 91.52 (s), 80.35 (s), 66.74 (s), 55.17 (s), 32.07 (s), 28.13 (s), 24.78 (s); LC-MS: [M+H]+ 282; purity 94 % (method LCMS2); HRMS (ESI+) m/z: [M]+

167 calcd for 168 C15H24O4N, 282.1700; found, 282.1695

169 tert-Butyl 4-(3-((1-(4-chlorophenyl)-2-oxo-2-(6-(trifluoromethoxy)indolin-1-yl)ethyl)amino)-5-

170 methoxyphenoxy)butanoate (4)

171 A mixture of 2 (3.6 g, 8.3 mmol), 3 (2.3 g, 8.3 mmol) and diisopropylethylamine (1.7 ml, 9.94 mmol) in CH3CN 172 (80 mL) was stirred at 70 °C for 4 h. The mixture was concentrated under reduced pressure, diluted with EtOAc,

173 and washed with 1 N aqueous HCl and water. The organic phase was separated, dried over MgSO4, filtered, and 174 the solvent was evaporated under reduced pressure. The compound was purified by flash chromatography on 175 silica gel (15-40 pm, 120 g, heptane/EtOAc 80/20). The pure fractions were combined and evaporated to dryness

176 to give, after crystallization from diisopropyl ether, tert-butyl 4-(3-((1-(4-chlorophenyl)-2-oxo-2-(6-

177 (trifluoromethoxy)indolin-1-yl)ethyl)amino)-5-methoxyphenoxy)butanoate 4 (2.6 g, yield: 49 %).

178 1 H NMR (400 MHz, DMSO-d6) d ppm 8.03 (s, 1 H), 7.55 (d, J = 8.6 Hz, 2 H), 7.43 (d, J = 8.6 Hz, 2 H), 7.33 (d, 179 J = 8.1 Hz, 1 H), 7.01 (dd, J = 8.1, 1.5 Hz, 1 H), 6.44 (d, J = 8.8 Hz, 1 H), 5.94 (d, J = 2.0 Hz, 2 H), 5.75 (t, J = 180 2.0 Hz, 1 H), 5.55 (d, J = 8.8 Hz, 1 H), 4.51 (td, J = 10.3, 6.5 Hz, 1 H), 4.04 (td, J = 10.3, 7.3 Hz, 1 H), 3.84 (t, J 6 181 = 6.3 Hz, 2 H), 3.62 (s, 3 H), 3.09 – 3.23 (m, 2 H), 2.31 (t, J = 7.3 Hz, 2 H), 1.86 (quin, J = 6.8 Hz, 2 H), 1.39 (s, 9 H); 13

182 C NMR (101 MHz, CHLOROFORM-d) δ ppm 172.57, 168.84, 161.66, 161.02, 148.65, 148.63, 147.68,

183 143.79, 135.66, 134.48, 129.58, 129.42, 129.38, 124.99, 116.92, 120.50 (q, J=257.2 Hz), 111.13, 93.02, 92.72, 91.06, 80.38, 77.25, 66.79, 59.74, 55.17, 48.31, 32.09, 28.15, 27.64, 24.77; LC-MS: [M+H]+

184 635; purity: 98 % (method LCMS3); Melting Point: 109-125 °C (DSC peak: 116.1 °C); HRMS (ESI+) m/z: [M]+ 185 calcd for 186 C32H34ClF3N2O6, 635.2130; found, 635.2127 187 (+)-4-(3-((1-(4-Chlorophenyl)-2-oxo2-(6-(trifluoromethoxy)indolin-1-yl)ethyl)amino)-5-

188 methoxyphenoxy)butanoic acid (JNJ-A07) 189 A solution of 4 (2.4 g, 3.8 mmol) in 4 M HCl in dioxane (24 mL) was stirred at 5 °C for 3 h and at room 190 temperature for 3 h. The precipitate was filtered off and dried to afford 4-(3-((1-(4-chlorophenyl)-2-oxo2-(6- 191 (trifluoromethoxy)indolin-1-yl)ethyl)amino)-5-methoxyphenoxy)butanoic acid as an HCl salt (racemic JNJ192 A07, 2 g, 0.8 eq. HCl, 0.07 eq. H2O). This salt was neutralized prior to chiral separation by dissolving it in 193 EtOAc and treating this solution with 1 N aqueous NaOH and evaporation of the organic layer under reduced 194 pressure.

The enantiomers were separated via preparative chiral SFC (Stationary phase: Chiralcel® OD-H 5 pm 195 250 x 30 mm, Mobile phase: 50 % CO2, 50 % iPrOH (+ 0.3 % iPrNH2)) and further purified via preparative 196 achiral SFC (Stationary phase: Cyano® 6 pm 150 x 21.2 mm, Mobile phase: 80 % CO2, 20 % MeOH (+ 0.3 % 197 iPrNH2)). The product fractions were combined and evaporated under reduced pressure. Each enantiomer was 198 taken up with EtOAc and washed with 1 N aqueous HCl. The organic layers were separated, dried over MgSO4, 199 filtered, and the solvent was evaporated under reduced pressure. The first eluted enantiomer was solidified from 200 diethyl ether/diisopropyl ether to give the epimer of JNJ-A07 (616 mg, yield: 28 %).

The second eluted

201 enantiomer was solidified from diethyl ether/diisopropyl ether to give JNJ-A07 (715 mg, yield: 32 %).

202 203 Epimer of JNJ-A07:

204 1 H NMR (500 MHz, DMSO-d6) δ ppm 12.12 (br s, 1 H), 8.04 (br s, 1 H), 7.55 (br d, J = 8.2 Hz, 2 H), 7.44 (br d, 205 J = 8.5 Hz, 2 H), 7.34 (br d, J = 7.9 Hz, 1 H), 7.01 (br d, J = 7.6 Hz, 1 H), 6.45 (br s, 1 H), 5.95 (br d, J = 10.1 206 Hz, 2 H), 5.76 (s, 1 H), 5.57 (br s, 1 H), 4.47 – 4.57 (m, 1 H), 3.99 – 4.11 (m, 1 H), 3.85 (br t, J = 6.3 Hz, 2 H), 3.62 (s, 3 H), 3.08 – 3.27 (m, 2 H), 2.34 (br t, J = 7.3 Hz, 2 H), 1.87 (quin, J = 6.7 Hz, 2 H); 13

207 C NMR (101 MHz, 208 DMSO-d6) δ ppm 174.56, 169.79, 161.46, 160.71, 149.08, 147.64, 144.48, 137.28, 132.91, 131.95, 130.56, 209 128.89, 126.34, 120.58 (d, J=256.0 Hz), 116.69, 109.52, 93.08, 92.80, 90.23, 66.65, 58.69, 55.20, 48.65, 30.57, 27.48, 24.72; LC-MS: [M+H]+ 579; purity: 100 % (method LCMS1);

Chiral SFC: [M+H]+ 210 579; chiral purity 100 % (method SFC1); [a]D20 211 : -48.5° (589 nm, c 0.27 w/v %, DMF, 20 °C); Melting Point: 62-80 °C (DSC peak: 70.6 °C); HRMS (ESI+) m/z: [M]+ 212 calcd for C28H27O6N2ClF3, 579.1504; found, 579.1501 213 214

JNJ-A07: 215 1 H NMR (500 MHz, DMSO-d6) δ ppm 12.12 (brs, 1 H), 8.04 (br s, 1 H), 7.55 (br d, J = 8.2 Hz, 2 H), 7.44 (br d, 216 J = 8.2 Hz, 2 H), 7.34 (br d, J = 7.9 Hz, 1 H), 7.01 (br d, J = 7.9 Hz, 1 H), 6.45 (br s, 1 H), 5.95 (br d, J = 10.1 217 Hz, 2 H), 5.76 (br s, 1 H), 5.57 (s, 1 H), 4.46 – 4.59 (m, 1 H), 3.99 – 4.10 (m, 1 H), 3.85 (br t, J = 6.1 Hz, 2 H), 3.62 (s, 3 H), 3.09 – 3.27 (m, 2 H), 2.34 (br t, J = 7.3 Hz, 2 H), 1.87 (br t, J = 6.8 Hz, 2 H); 13

218 C NMR (101 MHz,

219 DMSO-d6) δ ppm 174.53 (C28), 169.79 (C10), 161.47 (C20), 160.72 (C22), 149.08 (C18), 147.65 (C6), 144.48 220 (C8), 137.29 (C12), 132.92 (C15), 131.95 (C3), 130.56 (C13, C17), 128.89 (C14, C16), 126.34 (C4), 120.58 (q, 221 J = 255.1 Hz, C9), 116.67 (C5), 109.51 (C7), 93.11 (C23), 92.81 (C21), 90.26 (C19), 66.66 (C25), 58.70 (C11), 55.21 (C24), 48.67 (C1), 30.57 (C27), 27.49 (C2), 24.72 (C26); LC/MS: [M+H]+

222 579; purity 100 % (method LCMS1); Chiral SFC: [M+H]+ 579; chiral purity 100 % (method SFC1); [a]D20

223 : +42.9° (589 nm, c 0.28 w/v %, 224 DMF, 20 °C); Melting point: 62-78 °C (DSC peak: 71.3 °C) ; HRMS (ESI+) m/z calcd for C28H27O6N2ClF3 [M]+ 225 , 579.1504, found 579.1500; Elemental analysis requires C, 58.09 %; H, 4.53 %; N, 4.84 % found C, 226 58.60 %; H, 4.59 %; N, 4.80 %

CLIP

https://www.bioworld.com/articles/512333-potent-selective-pan-serotype-dengue-inhibitor-developed

Blocking the interaction between two dengue virus (DENV) nonstructural proteins, NS3 and NS4B, with a newly developed small-molecule inhibitor resulted in potent antiviral activity in mouse models, according to an international collaborative study led by scientists at the University of Leuven (KU Leuven), CD3 the Centre for Drug Design and Discovery in Leuven, and Janssen Pharmaceutica in Beerse, Belgium.

This protein interaction represents a promising new target for the development of pan-serotype DENV inhibitors with a high barrier to resistance, with the potency of the inhibition warranting further development of these compounds, the authors reported in the October 6, 2021, edition of Nature.

“This is the first study to show that blocking the NS3/NS4B interaction has potent antiviral activity in mice warranting the further development of such inhibitors,” said study co-leader Johan Neyts, professor of virology at KU Leuven.

Dengue is currently among the leading threats to global public health, with an estimated 96 million individuals developing dengue disease, which is probably an underestimation.

In addition, the incidence of dengue has increased approximately 30-fold over the past 50 years. DENV is now endemic in the subtropical regions of 128 countries, with an estimated 4 billion people at risk of infection, predicted to increase to 6 billion by 2080.

This dengue upsurge is driven by various factors, most notably rapid urbanization and the spread of the Aedes mosquito vectors due to climate change.

The DENV has four serotypes that are further classified into genotypes, which are increasingly co-circulating in endemic regions. Antibodies to infection with one serotype can lead to a more severe second infection with a different serotype increases the risk of potentially life-threatening severe dengue.

The DENV vaccine Dengvaxia (Sanofi-Pasteur), which has been approved in several countries for individuals aged at least 9 years, is only recommended for those with previous DENV exposure.

Moreover, there are currently no available antiviral agents for dengue prevention or treatment, while development of pan-serotype DENV inhibitors has proven challenging.

“The major developmental challenge has been to obtain ultrapotent antivirals that also have equipotent activity against the four DENV serotypes,” Neyts told BioWorld Science.

Such drugs should lower viral loads during an ongoing infection, thereby reducing dengue-associated morbidity and mortality, as well as transmission.

In their new Nature study, researchers co-led by Neyts, Patrick Chaltin, managing director of CD3 the Centre for Drug Design and Discovery, and Marnix Van Loock, R&D Lead Emerging Pathogens, Janssen Global Public Health at Janssen Pharmaceutica, identified potential new DENV inhibitors using large-scale cell-based anti-DENV-2 screening.

“We screened tens of thousands of molecules and interesting hits were further optimized to eventually obtain JNJ-A07 and other ultrapotent and selective analogues, with roughly 2,000 analogues being synthesized and tested,” said Neyts.

Notably, the promising small molecule JNJ-A07 was demonstrated to have nanomolar to picomolar activity against a panel of 21 clinical isolates representing the natural genetic diversity of known DENV genotypes and serotypes.

The molecule was then shown to have a high barrier to resistance “by months of culturing the dengue virus in suboptimal concentrations of the inhibitor,” Neyts said.

JNJ-A07 was then shown to prevent formation of the viral replication complex by blocking the interaction between the nonstructural proteins NS3 and NS4B, thereby revealing a previously undescribed mechanism of antiviral action.

JNJ-A07 was further demonstrated to have a favorable pharmacokinetic (PK) profile resulting in outstanding efficacy against DENV infection in mouse models.

“JNJ-A07’s favorable PK profile resulted from optimization of the ADME [absorption, distribution metabolism and excretion] properties of the analogues within this chemical series,” Janssen’s Van Loock told BioWorld Science.

“This enabled us to administer the compound [twice daily] in mice and assess its efficacy, which resulted in a significantly reduced viral load and protected against mortality in a mouse lethal challenge model.”

However, “additional research will be required in preclinical models, to understand how these findings reflect those in humans, as currently no translational models are available to assess the potential effect in humans,” noted Van Loock.

Delaying treatment commencement until peak viremia had developed was shown to result in a rapid and significant reduction in viral load in the mouse models of infection.

This is an important finding, as “one wants an antiviral effect that is independent of how much [viral] replication is ongoing,” Van Loock said.

“In these mice, the reduction in viral load was also very pronounced if the treatment was initiated on the day of peak viral load, when the effect was quantified 24 hours later.”

On safety, said Neyts, as JNJ-A07 and its analogues “target specific viral proteins that have no homologues in eukaryotic cells, we expect a considerable safety window, with these agents being very well tolerated.” The safety and potency of DENV inhibition established in this study justifies the further development of these novel antivirals, with an analogue being currently in further development.

Further development will include “using our know-how to also develop drugs against the other member of the flavivirus family to which DENV belongs, including Japanese encephalitis, Zika, yellow fever, West Nile virus, et cetera,” said Neyts.

Meanwhile, “Janssen has moved the compound into clinical development and continues to work closely in this regard with teams at KU Leuven and elsewhere,” said Van Loock.

“We will be sharing information about progress of the compound’s clinical development during the American Society of Tropical Medicine and Hygiene meeting this November.”

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

/////////////////////////////////////////////////////////////////////////////////////////////////////PatentWO2017167951https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017167951

  • IN201827040889
  • US2020299235
  • US2019112266
  • US10689340

Due to the presence of said chiral carbon atom, a “compound of formula (I)” can be the (R)-enantiomer, the (S)-enantiomer, the racemic form, or any possible combination of the two individual enantiomers in any ratio. When the absolute (R)-or (S)-configuration of an enantiomer is not known, this enantiomer can also be identified by indicating whether the enantiomer is dextrorotatory (+)- or levorotatory (-)- after measuring the specific optical rotation of said particular enantiomer.

In an aspect the present invention relates to a first group of compound of formula (I) wherein the compounds of formula (I) have the (+) specific rotation.

In a further aspect the present invention relates to a second ground of compounds of formula (I) wherein the compounds of formula (I) have the (-) specific rotation.

Example 4: synthesis of 4-(3-((1 -(4-chlorophenyl)-2-oxo-2-(6-(trifluoromethoxy)-indolin-1 -yl)ethyl)amino)-5-methoxyphenoxy)butanoic acid (Compound 4) and chiral separation into Enantiomers 4A and 4B.

Synthesis of intermediate 4a:

A mixture of 6-(trifluoromethoxy)indoline [CAS 959235-95-1] (2 g, 9.84 mmol), 2-(4-chlorophenyl)acetic acid [CAS 1878-66-6] (1 .85 g, 10.8 mmol), HATU (5.6 g, 14.8 mmol) and diisopropylethylamine (4.9 ml_, 29.5 mmol) in DMF (40 ml_) was stirred at room temperature for 12 h. Water was added and the precipitate was filtered off. The residue was taken up with EtOAc. The organic solution was washed with a 10% aqueous solution of K2CO3, brine, dried over MgSO4, filtered and the solvent was evaporated under reduced pressure. The residue was purified by chromatography on silica gel (15-40 μιτι, 80 g, heptane/EtOAc gradient 90/10 to 60/40). The pure fractions were combined and the solvent was concentrated under reduced pressure to give 2-(4-chlorophenyl)-1 -(6-(trifluoromethoxy)indolin-1 -yl)-ethanone 4a (3 g).

Synthesis of intermediate 4b:

At -78°C, under N2 flow, LiHMDS 1 .5 M in THF (1 1 .2 ml_, 16.9 mmol) was added dropwise to a mixture of 2-(4-chlorophenyl)-1 -(6-(trifluoromethoxy)indolin-1 -yl)-ethanone 4a (3 g, 8.43 mmol) in THF (50 ml_). The mixture was stirred for 15 min at -78°C and a solution of /V-bromosuccinimide (1 .65 g, 9.3 mmol) in THF (30 ml_) was added dropwise. After stirring for 2 h at -78°C, the reaction was quenched with a saturated solution of NH CI. The mixture was extracted with EtOAc. The organic layer was separated, dried over MgSO4, filtered and the solvent was evaporated under reduced pressure to give 2-bromo-2-(4-chlorophenyl)-1 -(6-(trifluoromethoxy)indolin-1 -yl)ethanone 4b (3.6 g). The compound was used as such in the next step.

Synthesis of intermediate 4c:

A mixture of 2-bromo-2-(4-chlorophenyl)-1 -(6-(trifluoromethoxy)indolin-1 -yl)-ethanone 4b (3.6 g, 8.3 mmol), terf-butyl 4-(3-amino-5-methoxyphenoxy)-butanoate 1a (2.3 g, 8.3 mmol) and diisopropylethylamine (1 .7 mL, 9.94 mmol) in CH3CN (80 mL) was stirred at 70°C for 4 h. The mixture was concentrated under reduced pressure, diluted with EtOAc, and washed with 1 N HCI and water. The organic phase was separated, dried over MgSO4, filtered and the solvent was evaporated under reduced pressure. The compound was purified by flash chromatography on silica gel (15-40 μιτι, 120 g, heptane/EtOAc 80/20). The pure fractions were combined and evaporated to dryness to give, after crystallization from diisopropyl ether, te/t-butyl 4-(3-((1 -(4-chlorophenyl)-2-oxo-2-(6-(trifluoro-methoxy)indolin-1 -yl)ethyl)amino)-5-methoxyphenoxy)butanoate 4c (2.6 g).

Synthesis of Compound 4 and chiral separation into Enantiomers 4A and 4B: A solution of terf-butyl 4-(3-((1 -(4-chlorophenyl)-2-oxo-2-(6-(trifluoromethoxy)-indolin-1 -yl)ethyl)amino)-5-methoxyphenoxy)butanoate 4c (2.4 g, 3.8 mmol) in 4M HCI in dioxane (24 mL) was stirred at 5°C for 3 h and at room temperature for 3h. The precipitate was filtered off and dried to afford 4-(3-((1 -(4-chlorophenyl)-2-oxo-2-(6-(trifluoromethoxy)indolin-1 -yl)ethyl)amino)-5-methoxyphenoxy)butanoic acid as an HCI salt (Compound 4, 2 g, 0.8 equiv. HCI, 0.07 equiv. H2O). Compound 4 (2 g, HCI salt) was neutralized prior to chiral separation by treatment of a solution of Compound 4 (HCI salt) in ethylacetate with 1 N NaOH and evaporation of the organic layer under reduced pressure. The enantiomers were separated via Preparative Chiral SFC (Stationary phase: Chiralcel® OD-H 5 μηη 250 x 30 mm, Mobile phase: 50% CO2, 50% iPrOH (+ 0.3% iPrNH2)) and further purified via Preparative achiral SFC (Stationary phase: Cyano® 6 μιτι 150×21 .2mm, Mobile phase: 80% CO2, 20% MeOH (+ 0.3% iPrNH2)). The product fractions were combined and evaporated under reduced pressure. The two enantiomers were taken up with EtOAc and washed with 1 N HCI. The organic layers were separated, dried over MgSO4, filtered and the solvent was evaporated under reduced pressure. The first eluted enantiomer was solidified from ether/diisopropyl ether to give Enantiomer 4A (616 mg). The second eluted enantiomer was solidified from ether/diisopropyl ether to give Enantiomer 4B (715 mg).

It is also possible to separate the enantiomers starting from the HCI salt of the racemate using the same conditions for chiral separation.

Compound 4:

1H NMR (500 MHz, DMSO-c/6) δ ppm 1 .87 (quin, J=6.9 Hz, 2 H) 2.34 (t, J=7.3 Hz, 2 H) 3.07 – 3.28 (m, 2 H) 3.62 (s, 3 H) 3.85 (t, J=6.5 Hz, 2 H) 4.04 (td, J=10.5, 7.1 Hz, 1 H) 4.52 (td, J=10.3, 6.5 Hz, 1 H) 5.57 (s, 1 H) 5.76 (t, J=2.2 Hz, 1 H) 5.90 – 6.00 (m, 2 H) 7.01 (dd, J=8.2, 1 .6 Hz, 1 H) 7.33 (d, J=8.2 Hz, 1 H) 7.41 – 7.48 (m, 2 H) 7.55 (d, J=8.5 Hz, 2 H) 8.03 (s, 1 H)

LC/MS (method LC-B): Rt 2.70 min, MH+ 579

Melting point: 150°C

Enantiomer 4A:

1H NMR (500 MHz, DMSO-c/6) δ ppm 1 .87 (quin, J=6.7 Hz, 2 H) 2.34 (br t, J=7.3 Hz, 2 H) 3.08 – 3.27 (m, 2 H) 3.62 (s, 3 H) 3.85 (br t, J=6.3 Hz, 2 H) 3.99 -4.1 1 (m, 1 H) 4.47 – 4.57 (m, 1 H) 5.57 (br s, 1 H) 5.76 (s, 1 H) 5.95 (br d, J=10.1 Hz, 2 H) 6.45 (br s, 1 H) 7.01 (br d, J=7.6 Hz, 1 H) 7.34 (br d, J=7.9 Hz, 1 H) 7.44 (br d, J=8.5 Hz, 2 H) 7.55 (br d, J=8.2 Hz, 2 H) 8.04 (br s, 1 H) 12.12 (br s, 1 H) LC/MS (method LC-A): Rt 2.95 min, MH+ 579

[a]D20: -48.5° (c 0.27, DMF)

Chiral SFC (method SFC-A): Rt 1 .13 min, MH+ 579, chiral purity 100%.

Enantiomer 4B:

1H NMR (500 MHz, DMSO-c/6) δ ppm 1 .87 (br t, J=6.8 Hz, 2 H) 2.34 (br t, J=7.3 Hz, 2 H) 3.09 – 3.27 (m, 2 H) 3.62 (s, 3 H) 3.85 (br t, J=6.1 Hz, 2 H) 3.99 -4.10 (m, 1 H) 4.46 – 4.59 (m, 1 H) 5.57 (s, 1 H) 5.76 (br s, 1 H) 5.95 (br d, J=10.1 Hz, 2 H) 6.45 (br s, 1 H) 7.01 (br d, J=7.9 Hz, 1 H) 7.34 (br d, J=7.9 Hz, 1 H) 7.44 (br d, J=8.2 Hz, 2 H) 7.55 (br d, J=8.2 Hz, 2 H) 8.04 (br s, 1 H) 12.12 (br s, 1 H) LC/MS (method LC-A): Rt 2.94 min, MH+ 579

[a]D20: +42.9° (c 0.28, DMF)

Chiral SFC (method SFC-A): Rt 2.13 min, MH+ 579, chiral purity 100%.

Patent

WO2021094563

The compounds of formula I according to the present invention may be synthesized according to methods described in the art, as disclosed in WO 2016/180696. The compounds of formula II according to the present invention may be prepared according to methods described in the art, as disclosed in WO2017/167951.

Compound (b) of the present invention was tested in AG129 mouse viremia model. The synthesis of compound (b) is described in WO 2017/167951, under Example 4.


compound (b)

PATENT

WO 2018215316

The compounds of formula (I) of the present invention all have at least one asymmetric carbon atom as indicated in the figure below by the carbon atom labelled with * :

Ref

https://doi.org/10.1038/s41586-021-03990-6

https://medicaldialogues.in/medicine/news/researchers-identify-first-drug-that-is-effective-against-dengue-infection-83187

////////////////////JNJ-A07, DENGUE, VIRUS, PRECLINICAL

NEW DRUG APPROVALS

ONE TIME

$10.00

XL 114, AUR 104 and XL 102, AUR 102 (NO CONCLUSIONS, ONLY PREDICTIONS)


File:Animated-Flag-India.gif - Wikimedia Commons
XL 102

XL 114

FOR BOTH, JUST PREDICTION

PREDICTIONS

or

front page image
Figure imgf000002_0001
Figure imgf000024_0001

N[C@@H](CO)c1nc(on1)[C@@H](NC(=O)N[C@H](C(=O)O)C(C)O)CC(N)=O

(2S)-2-[[(1S)-3-Amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]carbamoylamino]-3-hydroxybutanoic acid.png
SVG Image

(2S)-2-[[(1S)-3-amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]carbamoylamino]-3-hydroxybutanoic acid

CAS 2305027-62-5

C12 H20 N6 O7, 360.32Threonine, N-[[[(1S)-3-amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]amino]carbonyl]-, (2S,3ξ)-N[C@@H](CO)c1nc(on1)[C@@H](NC(=O)N[C@H](C(=O)O)C(C)O)CC(N)=O

ALSO SEE

Figure imgf000003_0002
str1
(2S,3R)-2-[[(1S)-3-Amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]carbamoylamino]-3-hydroxybutanoic acid.png

1673534-76-3C12 H20 N6 O7, 360.32
L-Threonine, N-[[[(1S)-3-amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]amino]
(2S,3R)-2-[[(1S)-3-amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]carbamoylamino]-3-hydroxybutanoic acidN-[[[(1S)-3-Amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]amino]carbonyl]-L-threonine

CAS 1673534-76-3

PD-1-IN-1 free base, EX-A1918, CS-6240NSC-799645CA-170 (AUPM-170)|PDL1 inhibitorHY-101093, PD-1-IN-1

N[C@@H](CO)c1nc(on1)[C@@H](NC(=O)N[C@H](C(=O)O)[C@@H](C)O)CC(N)=O

XL 114, AUR 104

A novel covalent inhibitor of FABP5 for cancer therapy

XL 102,  AUR 102

A potent, selective and orally bioavailable inhibitor of cyclin-dependent kinase 7 (CDK7)

NO CONCLUSIONS, ONLY PREDICTIONS

PREDICTIONS MORE

(2R,3R)-2-[[(1S)-3-Amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]carbamoylamino]-3-hydroxybutanoic acid.png
SVG Image

(2R,3R)-2-[[(1S)-3-amino-1-[3-[(1R)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]carbamoylamino]-3-hydroxybutanoic acid

C12H20N6O7, 360.32

(2S,3S)-2-[[(1S)-3-Amino-1-[3-[(1S)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]carbamoylamino]-3-hydroxybutanoic acid.png
SVG Image

(2S,3S)-2-[[(1S)-3-amino-1-[3-[(1S)-1-amino-2-hydroxyethyl]-1,2,4-oxadiazol-5-yl]-3-oxopropyl]carbamoylamino]-3-hydroxybutanoic acid

XL102, AUR 102

XL102 is a potent, selective and orally bioavailable covalent inhibitor of CDK7, which is an important regulator of the cellular transcriptional and cell cycle machinery. CDK7 helps regulate cell cycle progression, with overexpression observed in multiple cancers, such as breast, prostate and ovarian cancers. In preclinical studies, XL102 revealed potent anti-proliferative activity, induced cell death in a large panel of cancer cell lines and caused tumor growth inhibition and regression in xenograft models, demonstrating its potential as a targeted antitumor agent.

In late 2020, Exelixis exercised its option to in-license XL102 (formerly AUR102) from Aurigene per the companies’ July 2019 collaboration, option and license agreement. Exelixis has assumed responsibility for the future clinical development, manufacturing and commercialization of XL102. Aurigene retains limited development and commercial rights for India and Russia.

SYN

ABOUT Fatty acid-binding proteins (FABPs)

Fatty acid-binding proteins (FABPs) are involved in binding and storing hydrophobic ligands such as long-chain fatty acids, as well as transporting them to the appropriate compartments in the cell. Epidermal fatty acid-binding protein (FABP5) is an intracellular lipid-binding protein that is abundantly expressed in adipocytes and macrophages. Previous studies have revealed that the FABP5 expression level is closely related to malignancy in various types of cancer. However, its precise functions in the metabolisms of cancer cells remain unclear. Here, we revealed that FABP5 knockdown significantly induced downregulation of the genes expression, such as hormone-sensitive lipase (HSL), monoacylglycerol lipase (MAGL), elongation of long-chain fatty acid member 6 (Elovl6), and acyl-CoA synthetase long-chain family member 1 (ACSL1), which are involved in altered lipid metabolism, lipolysis, and de novo FA synthesis in highly aggressive prostate and breast cancer cells. Moreover, we demonstrated that FABP5 induced inflammation and cytokine production through the nuclear factor-kappa B signaling pathway activated by reactive oxygen species and protein kinase C in PC-3 and MDA-MB-231 cells. Thus, FABP5 might regulate lipid quality and/or quantity to promote aggressiveness such as cell growth, invasiveness, survival, and inflammation in prostate and breast cancer cells. In the present study, we have revealed for the first time that high expression of FABP5 plays a critical role in alterations of lipid metabolism, leading to cancer development and metastasis in highly aggressive prostate and breast cancer cells.

Fatty acid-binding protein, epidermal is a protein that in humans is encoded by the FABP5 gene

Function

This gene encodes the fatty acid binding protein found in epidermal cells, and was first identified as being upregulated in psoriasis tissue. Fatty acid binding proteins are a family of small, highly conserved, cytoplasmic proteins that bind long-chain fatty acids and other hydrophobic ligands. It is thought that FABPs roles include fatty acid uptake, transport, and metabolism.[6]

The phytocannabinoids (THC and CBD) inhibit endocannabinoid anandamide (AEA) uptake by targeting FABP5, and competition for FABPs may in part or wholly explain the increased circulating levels of endocannabinoids reported after consumption of cannabinoids.[7] Results show that cannabinoids inhibit keratinocyte proliferation, and therefore support a potential role for cannabinoids in the treatment of psoriasis.[8]

Interactions

FABP5 has been shown to interact with S100A7.[

ABOUT CD47/SIRPa axis

CD47/SIRPa axis is established as a critical regulator of myeloid cell activation and serves as an immune checkpoint for macrophage mediated phagocytosis. Because of its frequent upregulation in several cancers, CD47 contributes to immune evasion and cancer progression. CD47 regulates phagocytosis primarily through interactions with SIRPla expressed on macrophages. Blockade of SIRPla/CD47 has been shown to dramatically enhance tumor cell phagocytosis and dendritic cells maturation for better antigen presentation leading to substantially improved antitumor responses in preclinical models of cancer (M. P. Chao et al. Curr Opin Immunol. 2012 (2): 225-232). Disruption of CD47-SIRPa interaction is now being evaluated as a therapeutic strategy for cancer with the use of monoclonal antibodies targeting CD47 or SIRPa and engineered receptor decoys.

CD47 is expressed on virtually all non-malignant cells, and blocking the CD47 or the loss of CD47 expression or changes in membrane distribution can serve as markers of aged or damaged cells, particularly on red blood cells (RBC). Alternatively, blocking SIRPa also allows engulfment of targets that are not normally phagocytosed, for those cells where pre-phagocytic signals are also present. CD47 is a broadly expressed transmembrane glycoprotein with a single Ig-like domain and five membrane- spanning regions, which functions as a cellular ligand for SIRPa with binding mediated through the NH2-terminal V-like domain of SIRPa. SIRPa is expressed primarily on myeloid cells, including macrophages, granulocytes, myeloid dendritic cells (DCs), mast cells, and their precursors, including hematopoietic stem cells.

CD47 is also constitutively upregulated on a number of cancers such as Non-Hodgkin Lymphoma (NHL), Acute myeloid leukemia (AML), breast, colon, glioblastoma, glioma, ovarian, bladder and prostate cancers, etc. Overexpression of CD47 by tumor cells, which efficiently helps them to escape immune surveillance and killing by innate immune cells. However, in most of the tumor types, blockade of the CD47-SIRPa interaction as a single agent may not be capable of inducing significant phagocytosis and antitumor immunity, necessitating the need to combine with other therapeutic agents. The concomitant engagement of activating receptors such as Fc-receptors (FcRs) or other prophagocytic receptors (collectively known as “eat-me” signals) may be necessary for exploiting the maximum potential of the CD-47-SIPRa pathway blockade.

The role of engagement of prophagocytic receptors is proved by inefficiency to trigger phagocytosis either by anti-CD47 F(ab) fragments, single chain variable fragments of CD-47 or non-Fc portion- containing SIRPa proteins in blocking of the CD47-SIRPa interaction. When activating prophagocytic receptors are engaged, as evident in the case of using Fc portion-containing blocking anti-CD47 antibodies, CD47- SIRPa blockade is able to trigger more efficient phagocytosis. Combining CD47-SIRPa blocking agents with therapeutic antibodies (Fc-containing) targeting tumor antigens stimulate activating Fc receptors (FcRs) leading to efficient phagocytosis. The Fc portion of therapeutic antibody targeting tumor antigen also induces antibody-dependent cellular cytotoxicity (ADCC), which also adds to the therapeutic efficacy. Hence antibodies selected from the group consisting of rituximab, herceptin, trastuzumab, alemtuzumab, bevacizumab, cetuximab and panitumumab, daratumumab due to its tumor targeting nature and ADCC, can trigger more efficient phagocytosis.

Earlier approaches to disrupt CD47- SIRPa interaction utilized monoclonal antibodies targeting CD47 or SIRPa and engineered receptor decoys fused to Fc fragment. However, a concern with this approach is that CD47 is highly expressed on both hematopoietic and non-hematopoietic normal cells. Hence along with tumor cells CD47-SIRPa blocking agents containing Fc-portion may also target many normal cells potentially leading to their elimination by macrophages. The interaction of blocking antibodies with normal cells is considered as a major safety issue resulting in anemia, thrombocytopenia, and leukopenia. These agents may also affect solid tissues rich in macrophages such as liver, lung, and brain. Hence it may be ideal to block the CD47- SIRPa interaction by agents devoid of Fc portion, such as small

molecules, peptides, Fab fragments etc. while activating prophagocytic receptors in tumor cells by appropriate combinations to induce efficient phagocytosis of tumor cells.

Apart from Fc Receptors, a number of other prophagocytic receptors are also reported to promote engulfment of tumor cells in response to CD47-SIRPa blockade by triggering the phagocytosis. These include receptors for SLAMF7, Mac-l, calreticulin and possibly yet to identified receptors. B cell tumor lines such as Raji and other diffuse large B cell lymphoma express SLAMF7 and are implicated in triggering prophagocytic signals during CD47-SIRPa blockade.

Therapeutic agents known to activate prophagocytic receptors are also therefore ideal partners for use in combination with CD47-SIRPa blocking agents to achieve efficient phagocytosis. These agents include proteasome inhibitors (bortezomib, ixazomib and carfilzomib), Anthracyclines (Doxorubicin, Epirubicin, Daunorubicin, Idarubicin, Mitoxantrone) Oxaliplatin, Cyclophosphamide, Bleomycin, Vorinostat, Paclitaxel, 5-Fluorouracil, Cytarabine, BRAF inhibitory drugs (Dabrafenib, Vemurafenib), PI3K inhibitor, Docetaxel, Mitomycin C, Sorafenib, Tamoxifen and oncolytic viruses.

Apart from the specific agents known to have effect on‘eat me’ signals other agents including Abiraterone acetate, Afatinib, Aldesleukin, Aldesleukin, Alemtuzumab, Anastrozole, Axitinib, Belinostat, Bendamustine, Bicalutamide, Blinatumomab, Bosutinib, Brentuximab, Busulfan, Cabazitaxel, Capecitabine, Carboplatin, Carfilzomib, Carmustine, Ceritinib, Clofarabine, Crizotinib, Dacarbazine, Dactinomycin, Dasatinib, Degarelix, Denileukin, Denosumab, Enzalutamide, Eribulin, Erlotinib, Everolimus, Exemestane, Exemestane, Fludarabine, Fulvestrant, Gefitinib, Goserelin, Ibritumomab, Imatinib, Ipilimumab, Irinotecan, Ixabepilone, Lapatinib, Lenalidomide, Letrozole, Leucovorin, Leuprolide, Lomustine, Mechlorethamine, Megestrol, Nelarabine, Nilotinib, Nivolumab, Olaparib, Omacetaxine, Palbociclib, Pamidronate, Panitumumab, Panobinostat, Pazopanib, Pegaspargase, Pembrolizumab, Pemetrexed Disodium, Pertuzumab, Plerixafor, Pomalidomide, Ponatinib, Pralatrexate, Procarbazine, Radium 223, Ramucirumab, Regorafenib, rIFNa-2b, Romidepsin, Sunitinib, Temozolomide, Temsirolimus, Thiotepa, Tositumomab, Trametinib, Vinorelbine, Methotrexate, Ibrutinib, Aflibercept, Toremifene, Vinblastine, Vincristine, Idelalisib, Mercaptopurine and Thalidomide could potentially have effect on‘eat me’ signal pathway on combining with CD-47-SIRPa blocking agents.

In addition to the therapeutic agents mentioned above, other treatment modalities that are in use in cancer therapy also activate prophagocytic receptors, and thus can be combined with CD47-SIRPa blocking agents to achieve efficient phagocytosis. These include Hypericin-based photodynamic therapy (Hyp-PDT), radiotherapy, High-hydrostatic pressure, Photofrin-based PDT and Rose Bengal acetate -based PDT.

However, there is an unmet need for combining small molecule CD-47-SIRPa pathway inhibitors with agents capable of stimulating activating receptors such as Fc-receptors (FcRs) or other prophagocytic receptors, or combining with other treatment modalities that are in use in cancer therapy to activate prophagocytic receptors for exploiting the maximum potential of the CD-47- SIRPa pathway blockade.

CLIP

Exelixis In-Licenses Second Anti-Cancer Compound from Aurigene Following FDA Acceptance of Investigational New Drug Application for Phase 1 Clinical Trial in Non-Hodgkin’s Lymphoma

– Robust preclinical data support Exelixis’ clinical development of XL114, with phase 1 trial in Non-Hodgkin’s lymphoma expected to begin in the coming months –

– Exelixis will make an option exercise payment of $10 million to Aurigene –

https://www.businesswire.com/news/home/20211014005549/en/Exelixis-In-Licenses-Second-Anti-Cancer-Compound-from-Aurigene-Following-FDA-Acceptance-of-Investigational-New-Drug-Application-for-Phase-1-Clinical-Trial-in-Non-Hodgkin%E2%80%99s-LymphomaOctober 14, 2021 08:00 AM Eastern Daylight Time

ALAMEDA, Calif.–(BUSINESS WIRE)–Exelixis, Inc. (Nasdaq: EXEL) and Aurigene Discovery Technologies Limited (Aurigene) today announced that Exelixis has exercised its exclusive option under the companies’ July 2019 agreement to in-license XL114 (formerly AUR104), a novel anti-cancer compound that inhibits the CARD11-BCL10-MALT1 (CBM) signaling pathway, which promotes lymphocyte survival and proliferation. Exelixis has now assumed responsibility for the future clinical development, commercialization and global manufacturing of XL114. Following the U.S. Food and Drug Administration’s (FDA) recent acceptance of its Investigational New Drug (IND) application, Exelixis will soon initiate a phase 1 clinical trial evaluating XL114 monotherapy in patients with Non-Hodgkin’s lymphoma (NHL). At the American Association of Cancer Research Annual Meeting in April of this year, Aurigene presented preclinical data (Abstract 1266) demonstrating that XL114 exhibited potent anti-proliferative activity in a large panel of cancer cell lines ranging from hematological cancers to solid tumors with excellent selectivity over normal cells. In addition, oral dosing of XL114 resulted in significant dose-dependent tumor growth inhibition in diffuse large B-cell lymphoma (DLBCL) and colon carcinoma models.

“We are pleased that our agreement with Aurigene has generated a second promising compound that warrants advancement into clinical development and believe the collaboration will continue to play an important role in expanding our pipeline”

XL114 is the second molecule that Exelixis in-licensed from Aurigene under the companies’ July 2019 collaboration, option and license agreement. Exelixis previously exercised its option to in-license XL102, a potent, selective and orally bioavailable inhibitor of cyclin-dependent kinase 7 (CDK7), from Aurigene in December 2020 and initiated a phase 1 trial of XL102 as a single agent and in combination with other anti-cancer agents in patients with advanced or metastatic solid tumors in January 2021.

“We are pleased that our agreement with Aurigene has generated a second promising compound that warrants advancement into clinical development and believe the collaboration will continue to play an important role in expanding our pipeline,” said Peter Lamb, Ph.D., Executive Vice President, Scientific Strategy and Chief Scientific Officer, Exelixis. “XL114 has shown potent anti-proliferative activity in lymphoma cell lines that have aberrant activation of the CBM signaling pathway and may have a differentiated profile and potential as a best-in-class molecule that could improve outcomes for patients with Non-Hodgkin’s lymphoma and other hematologic cancers.”

XL114 was identified to have anti-proliferative activity in cell lines with constitutive activation of CBM signaling, including activated B-cell-like DLBCL (ABC-DLBCL), mantle cell lymphoma and follicular lymphoma cell lines. Further characterization of XL114 in cell-based assays demonstrated a functional role in B-cell (BCR) signaling pathways. Additionally, XL114 showed dose-dependent tumor growth inhibition in an ABC-DLBCL mouse xenograft tumor model. In preclinical development, XL114 also demonstrated a high degree of selectivity against a broad safety pharmacology panel of enzymes and receptors. While the precise molecular mechanism underlying XL114’s function in repressing BCR signaling and MALT1 activation has yet to be characterized, the fatty acid-binding protein 5 (FABP5) has been identified as a prominent XL114-binding target.

“XL114 is the second molecule that Exelixis has opted to in-license under our July 2019 agreement, underscoring the significant potential of our approach to the discovery and preclinical development of innovative cancer therapies that target novel mechanisms of action,” said Murali Ramachandra, Ph.D., Chief Executive Officer, Aurigene. “Exelixis has a track record of success in the clinical development and commercialization of anti-cancer therapies that provide patients with important new treatment options, and we are pleased that the continued advancement of XL114 will be supported by the company’s extensive clinical, regulatory and commercialization infrastructure.”

Under the terms of the July 2019 agreement, Exelixis made an upfront payment of $10 million for exclusive options to obtain an exclusive license from Aurigene to three preexisting programs, including the compounds now known as XL102 and XL114. In addition, Exelixis and Aurigene initiated three Aurigene-led drug discovery programs on mutually agreed upon targets, in exchange for an additional upfront payment of $2.5 million per program. The collaboration was expanded in 2021 to include three additional early discovery programs. Exelixis is also contributing research funding to Aurigene to facilitate discovery and preclinical development work on all nine programs. Exelixis may exercise its option for a program at any time up until the first IND for the program becomes effective. Having exercised options on two programs thus far (XL102 and XL114), if and when Exelixis exercises a future option, it will make an option exercise payment to Aurigene and assume responsibility for that program’s future clinical development and commercialization including global manufacturing. To exercise its option for XL114, Exelixis will make an option exercise payment to Aurigene of $10 million. Once Exelixis exercises its option for a program, Aurigene will be eligible for clinical development, regulatory and sales milestones, as well as royalties on future potential sales of the compound. Under the terms of the agreement, Aurigene retains limited development and commercial rights for India and Russia.

About Aurigene

Aurigene Discovery Technologies Limited is a development stage biotech company engaged in discovery and clinical development of novel and best-in-class therapies to treat cancer and inflammatory diseases and a wholly owned subsidiary of Dr. Reddy’s Laboratories Ltd. (BSE: 500124, NSE: DRREDDY, NYSE: RDY, NSEIFSC: DRREDDY). Aurigene is focused on precision-oncology, oral immune checkpoint inhibitors, and the Th-17 pathway. Aurigene’s programs currently in clinical development include an oral ROR-gamma inhibitor AUR101 for moderate to severe psoriasis in phase 2 under a U.S. FDA IND and a PD-L1/VISTA antagonist CA-170 for non-squamous non-small cell lung cancer in phase 2b/3 in India. Additionally, Aurigene has multiple compounds at different stages of pre-clinical development. Aurigene has also partnered with several large and mid-pharma companies in the U.S. and Europe and has multiple programs in clinical development. For more information, please visit Aurigene’s website at www.aurigene.com.

About Exelixis

Founded in 1994, Exelixis, Inc. (Nasdaq: EXEL) is a commercially successful, oncology-focused biotechnology company that strives to accelerate the discovery, development and commercialization of new medicines for difficult-to-treat cancers. Following early work in model system genetics, we established a broad drug discovery and development platform that has served as the foundation for our continued efforts to bring new cancer therapies to patients in need. Our discovery efforts have resulted in four commercially available products, CABOMETYX® (cabozantinib), COMETRIQ® (cabozantinib), COTELLIC® (cobimetinib) and MINNEBRO® (esaxerenone), and we have entered into partnerships with leading pharmaceutical companies to bring these important medicines to patients worldwide. Supported by revenues from our marketed products and collaborations, we are committed to prudently reinvesting in our business to maximize the potential of our pipeline. We are supplementing our existing therapeutic assets with targeted business development activities and internal drug discovery – all to deliver the next generation of Exelixis medicines and help patients recover stronger and live longer. Exelixis is a member of the Standard & Poor’s (S&P) MidCap 400 index, which measures the performance of profitable mid-sized companies. In November 2020, the company was named to Fortune’s 100 Fastest-Growing Companies list for the first time, ranking 17th overall and the third-highest biopharmaceutical company. For more information about Exelixis, please visit www.exelixis.com, follow @ExelixisInc on Twitter or like Exelixis, Inc. on Facebook.

Dinesh Chikkanna

Dinesh Chikkanna

Director, Medicinal Chemistry Aurigene Discovery Technologies

Murali Ramachandra

Murali Ramachandra

CEO at Aurigene Discovery Technologies

str1

CLIP

https://cancerres.aacrjournals.org/content/81/13_Supplement/1266

Abstract 1266: Discovery and preclinical evaluation of a novel covalent inhibitor of FABP5 for cancer therapyDinesh Chikkanna, Leena Khare Satyam, Sunil Kumar Pnaigrahi, Vinayak Khairnar, Manoj Pothuganti, Lakshmi Narayan Kaza, Narasimha Raju Kalidindi, Vijaya Shankar Nataraj, Aditya Kiran Gatta, Narasimha Rao Krishnamurthy, Sandeep Patil, DS Samiulla, Kiran Aithal, Vijay Kamal Ahuja, Nirbhay Kumar Tiwari, KB Charamannna, Pravin Pise, Thomas Anthony, Kavitha Nellore, Sanjeev Giri, Shekar Chelur, Susanta Samajdar and Murali Ramachandra 
DOI: 10.1158/1538-7445.AM2021-1266 Published July 2021 
Proceedings: AACR Annual Meeting 2021; April 10-15, 2021 and May 17-21, 2021; Philadelphia, PA

Abstract

Dysregulated fatty acid metabolism is thought to be a hallmark of cancer, wherein fatty acids function both as an energy source and as signals for enzymatic and transcriptional networks contributing to malignancy. Fatty acid-binding protein 5 (FABP5) is an intracellular protein that facilitates transport of fatty acids and plays a role in regulating the expression of genes associated with cancer progression such as cell growth, survival, and metastasis. Overexpression of FABP5 has been reported to contribute to an aggressive phenotype and a poor survival correlation in several cancers. Therefore, inhibition of FABP5 is considered as a therapeutic approach for cancers. Phenotypic screening of a library of covalent compounds for selective sensitivity of cancer cells followed by medicinal chemistry optimization resulted in the identification of AUR104 with desirable properties. Chemoproteomic-based target deconvolution revealed FABP5 as the cellular target of AUR104. Covalent adduct formation with Cys43 of FABP5 by AUR104 was confirmed by mass spectrometry. Target occupancy studies using a biotin-tagged AUR104 demonstrated potent covalent binding to FABP5 in both cell-free and cellular conditions. Ligand displacement assay with a fluorescent fatty acid probe confirmed the competitive binding mode of AUR104 with fatty acids. Binding at the fatty acid site and covalent bond formation with Cys43 were also demonstrated by crystallography. Furthermore, AUR104 showed a high degree of selectivity against a broad safety pharmacology panel of enzymes and receptors. AUR104 exhibited potent anti-proliferative activity in a large panel of cell lines derived from both hematological and solid cancers with a high degree of selectivity over normal cells. Anti-proliferative activity in lymphoma cell lines correlated with inhibition of MALT1 pathway activity, cleavage of RelB/Bcl10 and secretion of cytokines, IL-10 and IL-6. AUR104 displayed desirable drug-like properties and dose-dependent oral exposure in pharmacokinetic studies. Oral dosing with AUR104 resulted in dose-dependent anti-tumor activity in DLBCL (OCI-LY10) and NSCLC (NCI-H1975) xenograft models. In a repeated dose MTD studies in rodents and non-rodents, AUR104 showed good tolerability with an exposure multiple of >500 over cellular EC50 for up to 8 hours. In summary, we have identified a novel covalent FABP5 inhibitor with optimized properties that showed anti-tumor activity in in vitro and in vivo models with acceptable safety profile. The data presented here strongly support clinical development of AUR104.

Citation Format: Dinesh Chikkanna, Leena Khare Satyam, Sunil Kumar Pnaigrahi, Vinayak Khairnar, Manoj Pothuganti, Lakshmi Narayan Kaza, Narasimha Raju Kalidindi, Vijaya Shankar Nataraj, Aditya Kiran Gatta, Narasimha Rao Krishnamurthy, Sandeep Patil, DS Samiulla, Kiran Aithal, Vijay Kamal Ahuja, Nirbhay Kumar Tiwari, KB Charamannna, Pravin Pise, Thomas Anthony, Kavitha Nellore, Sanjeev Giri, Shekar Chelur, Susanta Samajdar, Murali Ramachandra. Discovery and preclinical evaluation of a novel covalent inhibitor of FABP5 for cancer therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1266.

Patent

US20200147054 – COMBINATION OF SMALL MOLECULE CD-47 INHIBITORS WITH OTHER ANTI-CANCER AGENTS

Muralidhara Ramachandra
Pottayil Govindan Nair Sasikumar
Girish Chandrappa Daginakatte
Kiran Aithal Balkudru

PATENT

WO 2020095256

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2020095256

Example- 1: The synthetic procedures for the preparation of compounds described in the present invention were described in co-pending Indian provisional patent application 201841001438 dated 12* Jan 2018, which is converted as PCT application

PCT/IB2019/050219, the contents of which are hereby incorporated by reference in their entirety.

str1

PATENT

WO 2018178947https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018178947&tab=PCTDESCRIPTION

PATENT

WO 2019138367

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019138367

PATENT

WO 2019073399

https://patents.google.com/patent/WO2019073399A1/en

Priority to IN201741036169

Example 4 of WO 2015/033299

Figure imgf000002_0001
Figure imgf000003_0002

PATENT

https://patents.google.com/patent/BR112020014202A2/en

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

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

PATENT

The present invention relates to substituted alkynylene compounds represented by compound of formula (I) pharmaceutically acceptable salts and stereoisomers thereof. The present invention further provides the methods of preparation of compound of formula (I) and therapeutic uses thereof as anti-cancer agents.

Patent

Example 1

(((S)-4-amino-1-(3-((S)-1,5-diaminopentyl)-1,2,4-oxadiazol-5-yl)-4-oxobutyl)carbamoyl)-L-proline (Compound 1)


 (MOL) (CDX)

Synthesis of Compound 1 b


 (MOL) (CDX)
      Ethylchloroformate (2.47 mL, 25.9 mmol) and NMM (2.9 mL, 25.9 mmol) were added to a solution of compound 1a (6.0 g, 17.3 mmol) in THF (60 mL) and stirred at −20° C. for 20 min. After 20 minutes 25% of aq.ammonia (24 mL) was added to the active mixed anhydride resulting from the reaction and the reaction mass was stirred at 0-5° C. for 30 min. The completeness of the reaction was confirmed by TLC analysis. The volatiles were evaporated under reduced pressure and partitioned between water and ethyl acetate. The organic layer was washed with NaHCO solution followed by citric acid solution and brine solution. The separated organic layer was dried over Na 2SO 4, filtered and evaporated under reduced pressure to yield 5.6 g of compound 1 b. LCMS: 346.4 [M+H] +.

Synthesis of Compound 1C


 (MOL) (CDX)
      Trifluroacetic anhydride (6.85 mL, 48.6 mmol) was added to a solution of compound 1b (5.6 g, 16.2 mmol), pyridine (7.84 mL, 97.2 mmol) in DCM (60 mL) at 0° C. and stirred at room temperature for an hour. The completion of the reaction was confirmed by TLC analysis. The volatiles were evaporated under reduced pressure and partitioned between water and CH 2Cl 2. The organic layer was washed with NaHCO solution followed by citric acid and brine solution. The separated organic layer was dried over Na 2SO 4, filtered and evaporated under reduced pressure to yield 5.42 g of compound 1c, which was used for next step directly.

Synthesis of Compound 1d


 (MOL) (CDX)
      Hydroxylamine hydrochloride (3.43 g, 49.5 mmol), water (10 mL) and K 2CO (4.54 g, 32.9 mmol) were added to a solution of compound 1c (5.4 g, 16.5 mmol) in EtOH (60 mL) and stirred at room temperature for overnight. The completion of the reaction was confirmed by TLC analysis. After the completion of reaction, the compound from the water was extracted by using the CH 2Cl followed washing the organic layer with water, brine and concentrated under reduced pressure to yield 5.8 g of compound 1d. LCMS: 361.3 [M+H] +.

Synthesis of Compound 1f


 (MOL) (CDX)
      HOBt (3.24 g, 24.0 mmol) and DIC (3.36 mL, 24.0 mmol) were added to a solution of Fmoc-Gln(Trt)-OH (compound 1e) (9.83 g, 16.1 mmol) in DMF (100 mL) at 0° C. and stirred for 15 min. Compound 1d (5.8 g, 16.1 mmol) was added to the reaction mass at the same temperature and the resulting mixture was stirred for an hour at the same temperature, followed by stirring at room temperature for an additional 2 h. The completion of the reaction was confirmed by TLC analysis. The reaction mixture was quenched with ice water; precipitated white solid was filtered; washed with water (150 mL) and dried under high under reduced pressure to yield 8.62 g of compound 1f. LCMS: 953.7 [M+H] +.

Synthesis of Compound 1g


 (MOL) (CDX)
      Acetic acid (5 mL) was added to a solution of compound 1f (5.0 g, 5.0 mmol) in acetonitrile (50 ml) at room temperature and the reaction mass was refluxed at 85° C. for 12 h. The completion of the reaction was confirmed by TLC analysis. The volatiles were evaporated under reduced pressure to obtain crude semi solid which was diluted with water and ethyl acetate. The organic layer was washed with NaHCO solution followed by citric acid and brine solution. The organic layer was dried over Na 2SO 4; filtered and evaporated under reduced pressure to obtain crude solid. Compound was purified using column chromatography to yield 4.3 g of title compound. LCMS: 935.6 [M+H] +.

Synthesis of Compound 1h


 (MOL) (CDX)
      Compound 1g (4.3 g, 4.5 mmol) was added to a solution of 20% piperidine in DMF (20 mL) at 0° C. and the reaction mass was stirred at same temperature for an hour. The completion of the reaction was confirmed by TLC analysis. After completion, the reaction mixture was quenched with ice-cold water and the resulting white precipitate was filtered and dried under vacuum. The crude product obtained was diluted with hexane, stirred and filtered to yield 3.0 g of compound 1h. LCMS: 713.4 [M+H] +.

Synthesis of Compound 1i


 (MOL) (CDX)
      Pyridine (0.33 mL, 4.2 mmol) was added to a solution of compound 1h (1.5 g, 2.1 mmol) in CH 2Cl (15 mL) and the resulting solution was stirred at room temperature for 10 min. 4-nitrophenyl chloroformate (0.84 g, 4.2 mmol) in CH 2Cl (15 mL) was added to the above mixture and the resultant mixture was stirred at room temperature for an hour. After completion of reaction (confirmed by TLC), it was diluted with CH 2Cl (50 mL) and washed with water (100 mL×2), 1N HCl (100 mL×2), water followed by brine solution (100 mL×2). The organic layer was dried over Na 2SO 4; filtered and evaporated under reduced pressure to yield 0.72 g compound 1i, which was taken to the next step without any further purification. LCMS: 878.9 [M-100].

Synthesis of compound 1j


 (MOL) (CDX)
      TEA (0.34 mL, 2.46 mm) was added to a solution of H-Pro-O tBu.HCl (0.21 g, 1.23 mmol) and compound 1i (0.72 g, 0.82 mmol) in THF (10 mL) at room temperature and stirred for 12 h. The volatiles were evaporated and portioned between ethyl acetate and water. The reaction mixture was diluted with ice cold water and extracted with EtOAc. The Organic layer was separated and dried over Na 2SO and concentrated under reduced pressure. The crude compound obtained was purified by column chromatography and compound elutes in 50% of ethyl acetate in hexane. Yield: 0.5 g of compound 1j. LCMS: 910.6 [M+H] +.

Synthesis of Compound 1


 (MOL) (CDX)
      Compound 1j (0.5 g, 0.55 mmol) was added to a cocktail mixture (10 m L) of TFA:TIPS:H 2O (95:2.5:2.5) and was stirred at room temperature for 3 h. The resulting reaction mixture was evaporated under reduced pressure, diluted with diethyl ether and filtered to yield 0.2 g of crude compound 1. The crude solid material was purified by preparative HPLC method described under experimental conditions. LCMS: 412.2 [M+H] +. HPLC t (min): 9.6.

Example 2

(S)-4-(3-((S)-1-amino-4-guanidinobutyl)-1,2,4-oxadiazol-5-yl)-4-(3-((S)-1-carboxy-2-phenylethyl) ureido)butanoic acid (Compound 7)


 (MOL) (CDX)

Synthesis of Compound 2b


 (MOL) (CDX)
      Ethylchloroformate (1.75 mL, 18.23 mmol) and NMM (2.0 mL, 18.23 mmol) were added into a solution of compound 2a (8.0 g, 15.18 mmol) in THF (45 mL) and the resulting mixture was stirred at −20° C. for 20 min. After 20 minutes 25% of aqueous ammonia (25 mL) was added to the active mixed anhydride generated and stirred at 0-5° C. for 30 min. The completeness of the reaction was confirmed by TLC analysis. The volatiles were evaporated under reduced pressure and partitioned between water and ethyl acetate. The organic layer was washed with NaHCO solution followed by citric acid solution and brine solution. The separated organic layer was dried over Na 2SO 4, filtered and evaporated under reduced pressure to yield 7.1 g of compound 2b. LCMS: 526.3 [M+H] +.

Synthesis of Compound 2c


 (MOL) (CDX)
      Trifluroacetic anhydride (TFAA) (2.83 mL, 20.26 mmol) was added to a solution of compound 2b (7.1 g, 13.51 mmol) in pyridine (7.08 g, 87.80 mmol) and the resulting mixture was stirred at room temperature for 2 h. The completion of the reaction was confirmed by TLC analysis. The volatiles were evaporated under reduced pressure and partitioned between water and ethyl acetate. The organic layer was washed with citric acid and brine solution. The separated organic layer was dried over Na 2SO 4, filtered and evaporated under reduced pressure. The crude solid was purified via column chromatography (60-120 silicagel) to yield 5.8 g of compound 2c. LCMS: 508.3 [M+H] +.

Synthesis of Compound 2d


 (MOL) (CDX)
      Hydroxylamine hydrochloride (1.56 g, 22.50 mmol), water (30 mL) and potassium carbonate (3.11 g, 11.25 mmol) were added to a solution of compound 2c (5.8 g, 11.25 mmol) in EtOH (60 mL) and stirred at 90° C. for 3 h. The completion of the reaction was confirmed by TLC analysis. The volatiles were evaporated under reduced pressure and partitioned between water and ethyl acetate. The organic layer was washed with brine solution, dried over Na 2SO then filtered and evaporated under reduced pressure, the solid obtained was washed with 20% ethyl acetate to yield 6.1 g of compound 2d. LCMS: 541.3 [M+H] +.

Synthesis of Compound 2f


 (MOL) (CDX)
      HOBt (2.28 g, 16.9 mmol) and DIC (2.62 mL, 16.9 mmol) were added to a solution of Fmoc-Glu(O tBu)-OH (compound 2e) (4.0 g, 9.02 mmol) in DMF (60 mL) at 0° C. and the resulting mixture was stirred for 15 min. Then compound 2d (6.1 g, 11.28 mmol) was added to the above mixture at the same temperature and the reaction mixture was continued stirring for an hour and then at room temperature for 2 h. The completion of the reaction was confirmed by TLC analysis. The reaction mixture was quenched with ice cold water, the precipitated white solid was filtered, washed with water (150 mL) and dried under high under reduced pressure. The solid was taken into 10% MeOH in DCM and washed the organic layer with 10% NaHCO 3, water and brine solution. The organic layer was dried over Na 2SO and concentrated under reduced pressure to yield 8.0 g of compound 2f. LCMS: 948.7 [M+H] +.

Synthesis of Compound 2g


 (MOL) (CDX)
      Acetic acid (7 mL) was added to a solution of compound 2f (7.0 g, 7.38 mmol) in THF (70 ml) at room temperature and the resulting mixture was refluxed at 70° C. for 12 h. The completion of the reaction was confirmed by TLC analysis. The volatiles were evaporated under reduced pressure to obtain crude semi solid which was diluted with water and ethyl acetate. The organic layer was washed with NaHCO solution followed by brine solution. The organic layer was dried over Na 2SO 4, filtered and evaporated under reduced pressure to get crude solid. The compound was purified by column chromatography (60-120 silicagel) to yield 5.4 g of compound 2g. LCMS: 930.5 [M+H] +.

Synthesis of Compound 2h


 (MOL) (CDX)
      Compound 2g (5.4 g, 5.80 mmol) was added to a solution of 50% piperidine in DMF (20 mL) at 0° C. and stirred at same temperature for 2 h. The completion of the reaction was confirmed by TLC analysis. The reaction mass was quenched with water (100 mL), the resulted precipitate was filtered. The solid obtained was dissolved in ethyl acetate and washed the organic layer with 10% NaHCO 3, water and brine. The organic layer was dried over Na 2SO and concentrated under reduced pressure. The crude product obtained was diluted with hexane and the resulted precipitate was filtered followed by washing with hexane to obtain 3.0 g of compound 2h. LCMS 708.6 [M+H] +.

Synthesis of Compound 2i


 (MOL) (CDX)
      Pyridine (0.75 mL, 9.3 mmol) was added to a solution of H-Phe-O tBu.HCl (2.0 g, 7.75 mmol) in CH 2Cl (20 mL) was added pyridine and the resulting solution was stirred at room temperature for 10 min. To this reaction mixture a solution of 4-nitrophenyl chloroformate (1.87 g, 9.30 mmol) in CH 2Cl (20 mL) was added and the resultant mixture was stirred at room temperature for 3 h. After completion of reaction (confirmed by TLC) it was diluted with CH 2Cl (50 mL) and washed with water (100 mL×2), 10% citric acid (100 mL×2), water (100 mL), followed by brine solution (100 mL). The organic layer was dried over Na 2SO 4, filtered and evaporated under reduced pressure to yield 1.7 g compound 2i, which was taken to the next step without any further purification.

Synthesis of Compound 2j


 (MOL) (CDX)
      TEA (0.29 mL, 2.1 mmol) was added to a solution of compound 2h (1.0 g, 1.41 mmol) and compound 2i (0.54 g, 1.41 mmol) in THF (10 mL) at room temperature and stirred for 3 h. The volatiles were evaporated and portioned between EtOAc and water. The reaction mixture was diluted with ice cold water and extracted with EtOAc followed by washing with 10% K 2CO (100 mL×4), water and brine solution. Organic layer separated and dried over Na 2SO and concentrated under reduced pressure. The crude product obtained was diluted with hexane and the resulted precipitate was filtered followed by washing with hexane yielded 0.98 g of compound 2j. LCMS: 955.6 [M+H] +.

Synthesis of Compound 7


 (MOL) (CDX)
      Compound 2j (0.5 g, 5.2 mmol) was added to cocktail mixture (5 m L) of trifluoroacetic: TIPS: water (95:2.5:2.5). The cleavage solution was stirred at room temperature for 3 h. The resulting reaction mixture was evaporated under reduced pressure, diluted with diethyl ether and filtered to yield 0.34 g of crude compound 2. The crude solid material was purified by preparative HPLC method as described under experimental conditions. LCMS: 491.1 [M+H] +. HPLC t R: (min): 11.1

PATENT

WO 2015/033299

https://patents.google.com/patent/WO2015033299A1/en?oq=WO+2015%2f033299

Pottayil Govindan Nair SasikumarMuralidhara RamachandraSeetharamaiah Setty Sudarshan Naremaddepalli

Figure imgf000024_0001

Example 1: Synthesis of Compound 1

Figure imgf000019_0001

Step la:

Figure imgf000019_0002

Ethylchloroformate (1.5 g, 13.78 mniol) and N-Methylmorpholine ( 1.4 g, 13.78 mmol) were added to a solution of compound la (3 g, 11.48 mmol) in THE (30 mL) arid stirred at -20 °C. After 20 min. Liquid ammonia (0.77 g, 45.92 mmol) was added to the active mixed anhydride formed in- situ and stirred at 0-5 °C for 20 min. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and partitioned between water and ethyl acetate. Organic layer was washed with NaHCOs, citric acid, brine solution, dried over Na2S04 and evaporated under reduced pressure to get 2.9 g of compound lb (Yield: 96.3%). LCMS: 261.0 ( Vi+H ; .

Step lb:

Figure imgf000020_0001

1 b 1cTrifluroacetic anhydride (9.7 g, 46.0 mmol) was added to a solution of compound lb (8 g, 30.7 mmol) in pyridine (24.3 g, 307.0 mmol) and stirred at room temperature for 3 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and partitioned between water and ethyl acetate. Organic layer was washed with NaHCO?,, citric acid, brine solution, dried over Na2-S04 and evaporated under reduced pressure to afford 7 g of compound lc (Yield: 94.0%). LCMS: 187.2 (M-¾u )+.

Step lc:

Figure imgf000020_0002

1 c 1dHydroxylamine hydrochloride (3 g, 43.37 mmol) and potassium carbonate (6 g, 43.37 mmol) were added to a solution of compound lc (7 g, 28.91 mmol) in EtOH (70 m L) and stirred at 90 °C for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and partitioned between water and ethyl acetate. Organic layer was washed with brine solution, dried over Na2S04 and evaporated under reduced pressure. The crude compound was purified by silica gel column chromatography (Eluent: 0-5% ethyl acetate in hexane) to get 4.2 g of compound Id (Yield: 52.8%). LCMS: 276.4 (M+H)+.Step Id:

Figure imgf000021_0001

Deoxo-Fluor® (1.83 g, 8.3 mmol) was added to a solution of Fmoc-Asn(Trt)-OH (4.5 g, 7.5 mmol) in CH2Q2 (50 mL) and stirred at 0 °C for 3 h. Then CH2CI2 was evaporated and triturated with hexane, decanted and evaporated under vacuum to get the corresponding acid fluoride. NMM (1.17 g, 1 1.6 mmol) and compound Id (1.6 g, 5.8 mmol) in THF were added to the acid fluoride and stirred at room temperature for 12 h. Then THF was evaporated and sodium acetate (0.72 g, 8.7 mmol) was added followed by EtOH (50 mL). The reaction mixture was stirred at 90 °C for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and partitioned between water and ethyl acetate. Organic layer was washed with NaHCOa, citric acid, brine solution, dried over Na2S04 and evaporated under reduced pressure, which was further purified by silica gel column chromatography (Eluent: 0-5% ethyl acetate in hexane) to afford 2.8 g of compound le (Yield: 44.4%). LCMS: 836.4 (M+Hf .Step le:

Ph3

Figure imgf000021_0002

To compound le (2.3 g, 2.7 mmol) in CH2CI2 (10 mL) diethyiarnine (10 mL) was added and the reaction mixture was stirred at room temperature for 30 min. The resulting solution was concentrated in vacuum to get gummy residue. The crude compound was purified by neutral alumina column chromatography (Eluent: 0-50% ethyl acetate in hexane then 0-5% methanol in chloroform) to get 1.4 g of If (Yield: 90 %). LCMS: 636.5 (M+Na)+.

Figure imgf000022_0001

1f 1To a solution of compound If (0.45 g) in CH2CI2 (5 mL), trifluoroacetic acid (5 mL) and catalytic amount of triisopropylsilane were added and stirred for 3 h at room temperature to remove the acid sensitive protecting groups. The resulting solution was concentrated in vacuum to afford 0.29 g of crude compound 1 which was purified using prep-HPLC method described under experimental conditions. \H NMR (DMSQ-d6, 400 MHz): δ 2.58 (m, 2H), 3.53 (m, 3H), 3.91 (t, 1H), 4.36 (t, 1H), 6.91 (s, 1H), 7.45 (s, 1H); 1 C NMR (DMSO-de, 400 MHz): δ 20.85, 45.71 , 50.23, 65.55, 171.03, 171 .41, 181.66. LCMS: 216.2 (Μ+ΗΓ; HPLC: tR = 13.1 min.Example 2: Synthesis of Co

Figure imgf000022_0002

Step 2a:

Figure imgf000022_0003

1f2a

The urea linkage was carried out by the coupling compound If (2.7 g, 4.39 mmoi) in THF (30 mL) at room temperature with compound 2b (1.67 g, 4.39 mmoi). The coupling was initiated by the addition of TEA (0.9 g, 8.78 mmoi) in THF (10 m L) and the resultant mixture was stirred at room temperature. After completion of 20 h, THF was evaporated from the reaction mass, and partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na2S04 and evaporated under reduced pressure to get compound 2a, which was further purified by silica gel column chromatography (Fluent: 0-50% ethyl acetate in hexane) to afford 3.46 g of compound 2a (Yield: 92.10%). LCMS 857.4 (M+H)+.

Figure imgf000023_0001

2aTo a solution of compound 2a (0.22 g, 0.25 mmol) in 0¾ί¾ (5 m L), trifluoroaeetic acid (5 mL) and catalytic amount of triisopropyisilane were added and stirred for 3h at room, temperature. The resulting solution was concentrated under reduced pressure to obtain 0.35 g of crude compound. The crude solid material was purified using preparative- HPLC method described under experimental conditions. LCMS: 347.1 (M+H)+; HPLC: tR = 12.9 min.

Synthesis of

Figure imgf000023_0002

2bTo the compound H-Ser(tBu)-OiBu (2 g, 9.2 mmol) in C I I■(.{■ (20 mL), triethylamine (1.39 g, 13.8 mmol) was added and the solution was stirred at room temperature for 5-10 min. To this mixture, solution of 4-Nitrophenyl chioro formate (2.22 g, 11.04 mmol) in CH2CI2 was added and the resultant mixture was stirred at room temperature for 30 min. The completion of the reaction was confirmed by TLC analysis. After completion of reaction, reaction mixture was diluted with CH2CI2 and washed with water and 5.0 M citric acid solution, dried over Na2SC>4 and evaporated under reduced pressure to get crude compound 2b, which was further purified by silica gel column chromatography (Eiuent: 0-20% ethyl acetate in hexane) to yield 2.1 g (58.9%) of 2b.Example 3: Synthesis of Compound 3

Figure imgf000023_0003

The compound was synthesised using similar procedure as depicted in Example 1 (compound 1) and D-amino acids are linked up in reverse order. Boc-D-Thr(‘Bu)-OH was used in place of Boc-Ser(‘Bu)-OH (compound la, Example 1) and Fmoc-D- Asn(trt)-OH in place of Fmoc-Asn(trt)-OH to yield 0.15 g crude material of the title compound 3. LCMS: 230.1 (M+H)+.Example 4: Synthesis of Co

Figure imgf000024_0001

The compound was synthesised using similar procedure as depicted in Example 2 for synthesising compound 2 using

Figure imgf000024_0002

instead of H-Ser(‘Bu)-0’Bu (in synthesis of compound 2b) to yield 0.35 g crude material of the title compound. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 361.2 (M+H)+, HPLC: tR = 12.19 min.Example 5: Synthesis of

Figure imgf000024_0003

The compound was synthesised using similar procedure as depicted in Example 4 (compound 4) using D-amino acids are linked up in reverse order. Boc-D-Thr(‘Bu)-OH was used in place of Boc-Ser(‘Bu)-OH, Fmoc-D-Asn(trt)-OH in place of Fmoc-Asn(trt)- OH and H-D-Ser(‘Bu)-0’Bu was used in place of H-Thr^Bu^O’Bu to yield 0.3 g crude material of the title compound. The cmde solid material was purified using preparative HPLC described under experimental conditions. LCMS: 361.3 (M+H)+. HPLC: tR = 13.58 min.Example 6: Synthesis of Compound 6

Figure imgf000024_0004

The compound was synthesised using similar procedure as depicted in Example 2 by using H-Thr(‘Bu)-OMe instead of H-Ser(‘Bu)-0’Bu (in synthesis of compound 2b) to yield 0.2 g crude material of the title compound. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 375.1 (M+H)+, HPLC: tR = 1.84 min.Example 7: Synthesis of Compound 7

Figure imgf000025_0001

Step 7a:

Figure imgf000025_0002

1f7aThe compound 7a was synthesised using similar procedure as for compound 2a (Example 2, step 2a) using H-Thr(‘Bu)-OMe instead of H-Ser(‘Bu)-OtBu to get crude material which was further purified by silica gel column chromatography (Eluent: 0-50% ethyl acetate in he ane) to get 2.0 g of compound 7a (Yield: 74 %). LCMS: 829.2 (M+H)+.Step 7b:

Figure imgf000025_0003

7a 7bTo a solution of compound 7a (0.35 g, 4.0 mmol) in THF (5 mL) was added lithium hydroxide (0.026 g, 0.63 mmol) at 0 °C and the mixture was stirred for 2 h at room temperature. The completion of the reaction was confirmed by TLC analysis. THF was evaporated from the reaction mass, and partitioned between water and ethyl acetate. Organic layer was washed with citric acid, brine solution, dried over Na2S04 and evaporated under reduced pressure to afford 7b, which was further purified by silica gel column chromatography (Eluent: 0-5% methanol in DCM) to get 0.3 g of product 7b (Yield: 86.7%). LCMS 815.2 (M+H)+.

Step 7c:

Figure imgf000026_0001

7b 7Compound 7b (0.295 g, 0.39 mmol) was anchored to Rink amide resin (0.7 g, 0.55 mmol/g) using HOBT (0.072 g, 0.54 mmol) and DIC (0.068 g, 0.54 mmol) method in DMF (10 mL). The resin was stirred for 12 h at room temperature. The resin was washed with DCM, DMF and DCM and dried. The target compound was cleaved from the rink amide resin using TFA (5 mL) and catalytic amount of TIPS. The resin was allowed to remain at room temperature for 2 h with occasional stirring. After 2 h, TFA and TIPS were evaporated under nitrogen atmosphere and the resulting residue was washed with diethyl ether to yield 0.1 g crude material of the title compound 7. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 360.0 (M+H)+, HPLC: tR = 13.88 min.Example 8: Synthesis of

Figure imgf000026_0002

The compound was synthesised using similar procedure as depicted in Example 2 (compound 2) using Fmoc-Glu(0’Bu)-OH instead of Fmoc-Asn(Trt)-OH to get 0.4 g crude material of the title compound. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 362.1 (M+H)+. HPLC: tR = 13.27 min.

PATENThttps://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019061324&tab=FULLTEXT

Patenthttps://patents.google.com/patent/WO2019067678A1/enPATENThttps://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019061324

PATENThttps://patents.google.com/patent/WO2018073754A1/en
PATENThttps://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019087087
PAPERSScientific Reports (2019), 9(1), 1-19. https://www.nature.com/articles/s41598-019-48826-6

figure1

Chemical structures of PD-L1 inhibitors developed by Aurigene (Aurigene-1) and Bristol-Meyers Squibb (BMSpep-57, BMS-103, and BMS-142). Chemical structures were generated using ChemDraw Professional 15. PATENT
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019087087

L-threonine’ mentioned in compound of formula (I) thereof can be represented by any one of the following formulae:

Publication NumberTitlePriority DateGrant Date
US-2020289477-A1Conjoint therapies for immunomodulation2017-11-06 
WO-2019073399-A1CRYSTALLINE FORMS OF 1,2,4-OXADIAZOLE SUBSTITUTED IN POSITION 32017-10-11 
AU-2018341583-A1Crystal forms of immunomodulators2017-09-29 
WO-2019061324-A1CRYSTALLINE FORMS OF IMMUNOMODULATORS2017-09-29 
WO-2019067678-A1CRYSTALLINE FORMS OF IMMUNOMODULATORS2017-09-29
Publication NumberTitlePriority DateGrant Date
US-2020247766-A1Crystal forms of immunomodulators2017-09-29 
US-2020061030-A1Dual inhibitors of vista and pd-1 pathways2016-10-20 
WO-2018073754-A1Dual inhibitors of vista and pd-1 pathways2016-10-20 
US-2020361880-A11,2,4-Oxadiazole and Thiadiazole Compounds as Immunomodulators2015-03-10 
EP-3041827-B11,2,4-oxadiazole derivatives as immunomodulators2013-09-062018-04-18
Publication NumberTitlePriority DateGrant Date
EP-3363790-B11,2,4-oxadiazole derivatives as immunomodulators2013-09-062020-02-19
US-10173989-B21,2,4-oxadiazole derivatives as immunomodulators2013-09-062019-01-08
US-10590093-B21,2,4-oxadiazole derivatives as immunomodulators2013-09-062020-03-17
US-2015073024-A11,2,4-Oxadiazole Derivatives as Immunomodulators2013-09-06 
US-2017101386-A11,2,4-Oxadiazole Derivatives as Immunomodulators2013-09-06
Publication NumberTitlePriority DateGrant Date
US-2018072689-A11,2,4-Oxadiazole Derivatives as Immunomodulators2013-09-06 
US-2019144402-A11,2,4-Oxadiazole Derivatives as Immunomodulators2013-09-06 
US-2020199086-A11,2,4-Oxadiazole Derivatives as Immunomodulators2013-09-06 
US-9771338-B21,2,4-oxadiazole derivatives as immunomodulators2013-09-062017-09-26
WO-2015033299-A11,2,4-oxadiazole derivatives as immunomodulators2013-09-06

////////////Investigational New Drug Application,  Phase 1,  Clinical Trial, Non-Hodgkin’s Lymphoma, XL 114, AUR 104, aurigene, Exelixis 

N[C@@H](CO)c1nc(on1)[C@@H](NC(=O)N[C@H](C(=O)O)C(C)O)CC(N)=O

https://patentscope.wipo.int/search/en/result.jsf?inchikey=HFOBENSCBRZVSP-WHFCDURNSA-N

NEW DRUG APPROVALS

ONE TIME

$10.00

Click here to purchase.

PATENT

The present invention relates to substituted alkynylene compounds represented by compound of formula (I) pharmaceutically acceptable salts and stereoisomers thereof. The present invention further provides the methods of preparation of compound of formula (I) and therapeutic uses thereof as anti-cancer agents.

XL 102

EXELIXIS AND AURIGENE ANNOUNCE THAT PROMISING PRECLINICAL DATA TO BE PRESENTED AT THE ENA SYMPOSIUM SUPPORT THE CLINICAL DEVELOPMENT OF A NOVEL CDK7 INHIBITOR

https://www.aurigene.com/exelixis-and-aurigene-announce-that-promising-preclinical-data-to-be-presented-at-the-ena-symposium-support-the-clinical-development-of-a-novel-cdk7-inhibitor/

Exelixis and Aurigene Announce That Promising Preclinical Data to Be Presented at the ENA Symposium Support the Clinical Development of a Novel CDK7 Inhibitor

– Detailed characterization of an oral inhibitor of CDK7 demonstrates potent activity against multiple hematologic and solid tumor cell lines, as monotherapy and in combination with chemotherapies –

October 09, 2020 03:02 AM Eastern Daylight Time

ALAMEDA, Calif.–(BUSINESS WIRE)–Exelixis, Inc. (Nasdaq: EXEL) and Aurigene Discovery Technologies Limited (Aurigene) today disclosed new preclinical data showing that AUR102 has potent anti-tumor activity in a large panel of cancer cell lines. AUR102 is a potent, selective, and orally bioavailable covalent inhibitor of cyclin-dependent kinase 7 (CDK7), which is an important regulator of the cellular transcriptional and cell cycle machinery. Exelixis has an exclusive option for AUR102 under its July 2019 exclusive collaboration, option and license agreement with Aurigene. The new data will be presented in a poster (Abstract 170) at the 32nd EORTC-NCI-AACR (ENA) Symposium, which is being held virtually on October 24-25, 2020.

“CDK7 plays a critical role in regulating cellular transcription and cell cycle machinery, making it an exciting target for cancer therapy”

“CDK7 plays a critical role in regulating cellular transcription and cell cycle machinery, making it an exciting target for cancer therapy,” said Murali Ramachandra, Ph.D., Chief Executive Officer of Aurigene. “The data to be presented at ENA 2020 demonstrate that AUR102 effectively engages CDK7 and inhibits a key mediator of the cell cycle and transcription. The ability to inhibit CDK7 activity with an orally available therapeutic such as AUR102 holds great potential to improve care and outcomes for patients with diverse cancer indications, including breast cancer, prostate cancer, leukemia and lymphoma.”

The abstract provides a summary of results from a detailed characterization of AUR102 in cancer cell lines and animal tumor models. Additional data will be presented in the poster. Key findings included in the abstract are:
• AUR102 exhibited potent anti-proliferative activity in a large panel of cell lines with induction of cell death in cell lines derived from multiple cancer types.
• The observed anti-proliferative activity correlated with cellular CDK7 target engagement and decreased levels of P-Ser5 RNAPII, a key mediator of transcription.
• AUR102 studies showed synergy when used in combination with multiple chemotherapies.
• Oral dosing with AUR102 resulted in dose-dependent anti-tumor activity, including complete tumor regression in diffuse large B-cell lymphoma, acute myeloid leukemia, and triple-negative breast cancer xenograft models.
• Inhibition of tumor growth was accompanied by complete target engagement as demonstrated in a parallel PK-PD study.
• AUR102 significantly impacts several pathways and key cancer driver and immune-response genes.

The study authors conclude that the data support clinical evaluation of AUR102 as a single agent and in combination with chemotherapies for the treatment of cancer.

“The exciting AUR102 data to be presented at ENA 2020 provide further validation of our partnering strategy, which gives us multiple opportunities to build a pipeline of best-in-class cancer therapies,” said Peter Lamb, Ph.D., Executive Vice President of Scientific Strategy and Chief Scientific Officer of Exelixis. “AUR102 could be the subject of an Investigational New Drug filing later this year, which would be an important value driver for the program itself and for our collaboration with Aurigene. We commend the Aurigene team on their ongoing success in building a robust body of data supporting the broad clinical potential of AUR102.”

Under the terms of the July 2019 agreement, Exelixis made an upfront payment of $10 million for exclusive options to license three preexisting programs from Aurigene. In addition, Exelixis and Aurigene initiated three Aurigene-led drug discovery programs on mutually agreed upon targets, in exchange for additional upfront option payments of $2.5 million per program. Exelixis is also contributing research funding to Aurigene to facilitate discovery and preclinical development work on all six programs. As the programs mature, Exelixis will have the opportunity to exercise an exclusive option for each program up until the time of Investigational New Drug (IND) filing acceptance. If Exelixis decides to exercise an option, it will make an option exercise payment to Aurigene and assume responsibility for that program’s future clinical development and commercialization including global manufacturing. Aurigene will be eligible for clinical development, regulatory, and sales milestones, as well as royalties on sales. Under the terms of the agreement, Aurigene retains limited development and commercial rights for India and Russia.

About Aurigene

Aurigene is a development stage biotech company engaged in discovery and clinical development of novel and best-in-class therapies to treat cancer and inflammatory diseases and a wholly owned subsidiary of Dr. Reddy’s Laboratories Ltd. (BSE: 500124, NSE: DRREDDY, NYSE: RDY). Aurigene is focused on precision-oncology, oral immune checkpoint inhibitors, and the Th-17 pathway. Aurigene’s programs currently in clinical development include an oral ROR-gamma inhibitor AUR101 for moderate to severe psoriasis in phase 2 under a U.S. FDA IND and a PD-L1/ VISTA antagonist CA-170 for non-squamous non-small cell lung cancer in phase 2b/3 in India. Additionally, Aurigene has multiple compounds at different stages of pre-clinical development. Aurigene has also partnered with several large and mid-pharma companies in the United States and Europe and has multiple programs in clinical development. For more information, please visit Aurigene’s website at http://www.aurigene.com.

About Exelixis

Founded in 1994, Exelixis, Inc. (Nasdaq: EXEL) is a commercially successful, oncology-focused biotechnology company that strives to accelerate the discovery, development and commercialization of new medicines for difficult-to-treat cancers. Following early work in model system genetics, we established a broad drug discovery and development platform that has served as the foundation for our continued efforts to bring new cancer therapies to patients in need. Our discovery efforts have resulted in four commercially available products, CABOMETYX® (cabozantinib), COMETRIQ® (cabozantinib), COTELLIC® (cobimetinib) and MINNEBRO® (esaxerenone), and we have entered into partnerships with leading pharmaceutical companies to bring these important medicines to patients worldwide. Supported by revenues from our marketed products and collaborations, we are committed to prudently reinvesting in our business to maximize the potential of our pipeline. We are supplementing our existing therapeutic assets with targeted business development activities and internal drug discovery – all to deliver the next generation of Exelixis medicines and help patients recover stronger and live longer. Exelixis is a member of Standard & Poor’s (S&P) MidCap 400 index, which measures the performance of profitable mid-sized companies. For more information about Exelixis, please visit http://www.exelixis.com, follow @ExelixisInc on Twitter or like Exelixis, Inc. on Facebook.

EXELIXIS AND AURIGENE ANNOUNCE THAT PROMISING PRECLINICAL DATA TO BE PRESENTED AT THE ENA SYMPOSIUM SUPPORT THE CLINICAL DEVELOPMENT OF A NOVEL CDK7 INHIBITOR

https://www.aurigene.com/exelixis-and-aurigene-announce-that-promising-preclinical-data-to-be-presented-at-the-ena-symposium-support-the-clinical-development-of-a-novel-cdk7-inhibitor/

Exelixis and Aurigene Announce That Promising Preclinical Data to Be Presented at the ENA Symposium Support the Clinical Development of a Novel CDK7 Inhibitor

– Detailed characterization of an oral inhibitor of CDK7 demonstrates potent activity against multiple hematologic and solid tumor cell lines, as monotherapy and in combination with chemotherapies –

October 09, 2020 03:02 AM Eastern Daylight Time

ALAMEDA, Calif.–(BUSINESS WIRE)–Exelixis, Inc. (Nasdaq: EXEL) and Aurigene Discovery Technologies Limited (Aurigene) today disclosed new preclinical data showing that AUR102 has potent anti-tumor activity in a large panel of cancer cell lines. AUR102 is a potent, selective, and orally bioavailable covalent inhibitor of cyclin-dependent kinase 7 (CDK7), which is an important regulator of the cellular transcriptional and cell cycle machinery. Exelixis has an exclusive option for AUR102 under its July 2019 exclusive collaboration, option and license agreement with Aurigene. The new data will be presented in a poster (Abstract 170) at the 32nd EORTC-NCI-AACR (ENA) Symposium, which is being held virtually on October 24-25, 2020.

“CDK7 plays a critical role in regulating cellular transcription and cell cycle machinery, making it an exciting target for cancer therapy”

“CDK7 plays a critical role in regulating cellular transcription and cell cycle machinery, making it an exciting target for cancer therapy,” said Murali Ramachandra, Ph.D., Chief Executive Officer of Aurigene. “The data to be presented at ENA 2020 demonstrate that AUR102 effectively engages CDK7 and inhibits a key mediator of the cell cycle and transcription. The ability to inhibit CDK7 activity with an orally available therapeutic such as AUR102 holds great potential to improve care and outcomes for patients with diverse cancer indications, including breast cancer, prostate cancer, leukemia and lymphoma.”

The abstract provides a summary of results from a detailed characterization of AUR102 in cancer cell lines and animal tumor models. Additional data will be presented in the poster. Key findings included in the abstract are:
• AUR102 exhibited potent anti-proliferative activity in a large panel of cell lines with induction of cell death in cell lines derived from multiple cancer types.
• The observed anti-proliferative activity correlated with cellular CDK7 target engagement and decreased levels of P-Ser5 RNAPII, a key mediator of transcription.
• AUR102 studies showed synergy when used in combination with multiple chemotherapies.
• Oral dosing with AUR102 resulted in dose-dependent anti-tumor activity, including complete tumor regression in diffuse large B-cell lymphoma, acute myeloid leukemia, and triple-negative breast cancer xenograft models.
• Inhibition of tumor growth was accompanied by complete target engagement as demonstrated in a parallel PK-PD study.
• AUR102 significantly impacts several pathways and key cancer driver and immune-response genes.

The study authors conclude that the data support clinical evaluation of AUR102 as a single agent and in combination with chemotherapies for the treatment of cancer.

“The exciting AUR102 data to be presented at ENA 2020 provide further validation of our partnering strategy, which gives us multiple opportunities to build a pipeline of best-in-class cancer therapies,” said Peter Lamb, Ph.D., Executive Vice President of Scientific Strategy and Chief Scientific Officer of Exelixis. “AUR102 could be the subject of an Investigational New Drug filing later this year, which would be an important value driver for the program itself and for our collaboration with Aurigene. We commend the Aurigene team on their ongoing success in building a robust body of data supporting the broad clinical potential of AUR102.”

Under the terms of the July 2019 agreement, Exelixis made an upfront payment of $10 million for exclusive options to license three preexisting programs from Aurigene. In addition, Exelixis and Aurigene initiated three Aurigene-led drug discovery programs on mutually agreed upon targets, in exchange for additional upfront option payments of $2.5 million per program. Exelixis is also contributing research funding to Aurigene to facilitate discovery and preclinical development work on all six programs. As the programs mature, Exelixis will have the opportunity to exercise an exclusive option for each program up until the time of Investigational New Drug (IND) filing acceptance. If Exelixis decides to exercise an option, it will make an option exercise payment to Aurigene and assume responsibility for that program’s future clinical development and commercialization including global manufacturing. Aurigene will be eligible for clinical development, regulatory, and sales milestones, as well as royalties on sales. Under the terms of the agreement, Aurigene retains limited development and commercial rights for India and Russia.

About Aurigene

Aurigene is a development stage biotech company engaged in discovery and clinical development of novel and best-in-class therapies to treat cancer and inflammatory diseases and a wholly owned subsidiary of Dr. Reddy’s Laboratories Ltd. (BSE: 500124, NSE: DRREDDY, NYSE: RDY). Aurigene is focused on precision-oncology, oral immune checkpoint inhibitors, and the Th-17 pathway. Aurigene’s programs currently in clinical development include an oral ROR-gamma inhibitor AUR101 for moderate to severe psoriasis in phase 2 under a U.S. FDA IND and a PD-L1/ VISTA antagonist CA-170 for non-squamous non-small cell lung cancer in phase 2b/3 in India. Additionally, Aurigene has multiple compounds at different stages of pre-clinical development. Aurigene has also partnered with several large and mid-pharma companies in the United States and Europe and has multiple programs in clinical development. For more information, please visit Aurigene’s website at http://www.aurigene.com.

About Exelixis

Founded in 1994, Exelixis, Inc. (Nasdaq: EXEL) is a commercially successful, oncology-focused biotechnology company that strives to accelerate the discovery, development and commercialization of new medicines for difficult-to-treat cancers. Following early work in model system genetics, we established a broad drug discovery and development platform that has served as the foundation for our continued efforts to bring new cancer therapies to patients in need. Our discovery efforts have resulted in four commercially available products, CABOMETYX® (cabozantinib), COMETRIQ® (cabozantinib), COTELLIC® (cobimetinib) and MINNEBRO® (esaxerenone), and we have entered into partnerships with leading pharmaceutical companies to bring these important medicines to patients worldwide. Supported by revenues from our marketed products and collaborations, we are committed to prudently reinvesting in our business to maximize the potential of our pipeline. We are supplementing our existing therapeutic assets with targeted business development activities and internal drug discovery – all to deliver the next generation of Exelixis medicines and help patients recover stronger and live longer. Exelixis is a member of Standard & Poor’s (S&P) MidCap 400 index, which measures the performance of profitable mid-sized companies. For more information about Exelixis, please visit http://www.exelixis.com, follow @ExelixisInc on Twitter or like Exelixis, Inc. on Facebook.

Exelixis Forward-Looking Statements

This press release contains forward-looking statements, including, without limitation, statements related to: Exelixis’ and Aurigene’s plans to present preclinical data in support of the continued development of AUR102 in a poster as part of the 32nd ENA Symposium; the potential for AUR102 to improve care and outcomes for patients with diverse cancer indications, including breast cancer, prostate cancer, leukemia and lymphoma; the potential for AUR102 to be the subject of an Investigational New Drug filing later in 2020; Exelixis’ potential future financial and other obligations under the exclusive collaboration, option and license agreement with Aurigene; and Exelixis’ plans to reinvest in its business to maximize the potential of the company’s pipeline, including through targeted business development activities and internal drug discovery. Any statements that refer to expectations, projections or other characterizations of future events or circumstances are forward-looking statements and are based upon Exelixis’ current plans, assumptions, beliefs, expectations, estimates and projections. Forward-looking statements involve risks and uncertainties. Actual results and the timing of events could differ materially from those anticipated in the forward-looking statements as a result of these risks and uncertainties, which include, without limitation: the availability of data at the referenced times; the level of costs associated with Exelixis’ commercialization, research and development, in-licensing or acquisition of product candidates, and other activities; uncertainties inherent in the drug discovery and product development process; Exelixis’ dependence on its relationship with Aurigene, including Aurigene’s adherence to its obligations under the exclusive collaboration, option and license agreement and the level of Aurigene’s assistance to Exelixis in completing clinical trials, pursuing regulatory approvals or successfully commercializing partnered compounds in the territories where they may be approved; the continuing COVID-19 pandemic and its impact on Exelixis’ research and development operations; complexities and the unpredictability of the regulatory review and approval processes in the U.S. and elsewhere; Exelixis’ and Aurigene’s continuing compliance with applicable legal and regulatory requirements; Exelixis’ and Aurigene’s ability to protect their respective intellectual property rights; market competition; changes in economic and business conditions; and other factors affecting Exelixis and its product pipeline discussed under the caption “Risk Factors” in Exelixis’ Quarterly Report on Form 10-Q filed with the Securities and Exchange Commission (SEC) on August 6, 2020, and in Exelixis’ future filings with the SEC. All forward-looking statements in this press release are based on information available to Exelixis as of the date of this press release, and Exelixis undertakes no obligation to update or revise any forward-looking statements contained herein, except as required by law.

Exelixis, the Exelixis logo, CABOMETYX, COMETRIQ and COTELLIC are registered U.S. trademarks. MINNEBRO is a registered Japanese trademark.

PIROXICAM


Skeletal formula of piroxicam
ChemSpider 2D Image | Piroxicam | C15H13N3O4S

PIROXICAM

  • Molecular FormulaC15H13N3O4S
  • Average mass331.346 Da

1,1-Dioxyde de 4-hydroxy-2-méthyl-N-(2-pyridinyl)-2H-1,2-benzothiazine-3-carboxamide

13T4O6VMAM

252-974-3[EINECS]

2H-1,2-Benzothiazine-3-carboxamide, 4-hydroxy-2-methyl-N-2-pyridinyl-, 1,1-dioxide

36322-90-4[RN]37134

-Hydroxy-2-methyl-3-(pyrid-2-yl-carbamoyl)-2H-1,2-benzothiazine 1,1-dioxide

Piroxicam 
CAS Registry Number: 36322-90-4 
CAS Name: 4-Hydroxy-2-methyl-N-2-pyridinyl-2H-1,2-benzothiazine-3-carboxamide 1,1-dioxide 
Additional Names: 3,4-dihydro-2-methyl-4-oxo-N-2-pyridyl-2H-1,2-benzothiazine-3-carboxamide 1,1-dioxide 
Manufacturers’ Codes: CP-16171 
Trademarks: Artroxicam (Coli); Baxo (Toyama); Bruxicam (Bruschettini); Caliment (Apotex); Erazon (Krka); Feldene (Pfizer); Flogobene (Farge); Geldene (Pfizer); Improntal (Kabi); Larapam (Lagap); Pirkam (DAK); Piroflex (Lagap); Reudene (ABC); Riacen (Chiesi); Roxicam (Gramon); Roxiden (Pulitzer); Sasulen (Andreu); Solocalm (Microsules); Zunden (Luitpold)Molecular Formula: C15H13N3O4S 
Molecular Weight: 331.35 
Percent Composition: C 54.37%, H 3.95%, N 12.68%, O 19.31%, S 9.68% 
Literature References: Non-steroidal anti-inflammatory with long half-life. Prepn (keto form): J. Lombardino, DE1943265idem,US3591584 (1970, 1971 to Pfizer).Synthesis and biological properties: J. Lombardino, E. Wiseman, J. Med. Chem.15, 848 (1972); J. Lombardino et al.,ibid.16, 493 (1973). Pharmacology: E. Wiseman et al.,Arzneim.-Forsch.26, 1300 (1976). Evaluation of ulcerogenic effects: G. Palacios et al.,Methods Find. Exp. Clin. Pharmacol.9, 353 (1987). Clinical pharmacology: L. Martinez et al.,ibid.10, 729 (1988). Review:eidem, in Pharmacological and Biochemical Properties of Drug Substancesvol. 3, M. E. Goldberg, Ed. (Am. Pharm. Assoc., Washington, DC, 1981) pp 324-346. Review of pharmacology and therapeutic efficacy: R. N. Brogden et al.,Drugs22, 165-187 (1981); eidem,ibid.28, 292-323 (1984). Symposium on clinical efficacy and safety: Am. J. Med.81, Suppl. 5B, 1-55 (1986). Comprehensive description: M. Mihalic et al.,Anal. Profiles Drug Subs.15, 509-531 (1986). 
Properties: Crystals from methanol, mp 198-200°. pKa 6.3 (2:1 dioxane-water). LD50 orally in mice: 360 mg/kg (Wiseman). 
Melting point: mp 198-200° 
pKa: pKa 6.3 (2:1 dioxane-water) 
Toxicity data: LD50 orally in mice: 360 mg/kg (Wiseman) 
Derivative Type: Cinnamic acid ester 
CAS Registry Number: 87234-24-0 
Additional Names: Piroxicam cinnamate; cinnoxicam 
Manufacturers’ Codes: SPA-S-510 
Trademarks: Sinartrol (SPA); Zelis (Proter); Zen (Prophin) 
Molecular Formula: C24H19N3O5S 
Molecular Weight: 461.49 
Percent Composition: C 62.46%, H 4.15%, N 9.11%, O 17.33%, S 6.95% 
Derivative Type: Compd with b-cyclodextrinCAS Registry Number: 121696-62-6 
Trademarks: Brexin (Chiesi); Cicladol (Master); Cycladol (Promedica) 
Molecular Formula: C57H83N3O39S 
Molecular Weight: 1466.33 
Percent Composition: C 46.69%, H 5.71%, N 2.87%, O 42.55%, S 2.19% 
Therap-Cat: Anti-inflammatory. 
Keywords: Anti-inflammatory (Nonsteroidal); Thiazinecarboxamides.

  • EINECS:252-974-3
  • LD50:250 mg/kg (M, p.o.);
    216 mg/kg (R, p.o.);
    108 mg/kg (dog, p.o.)

Piroxicam is a nonsteroidal anti-inflammatory drug (NSAID) of the oxicam class used to relieve the symptoms of painful inflammatory conditions like arthritis.[3][4] Piroxicam works by preventing the production of endogenous prostaglandins] which are involved in the mediation of pain, stiffness, tenderness and swelling.[3] The medicine is available as capsulestablets and (not in all countries) as a prescription-free gel 0.5%.[5] It is also available in a betadex formulation, which allows a more rapid absorption of piroxicam from the digestive tract.[3] Piroxicam is one of the few NSAIDs that can be given parenteral routes.

It was patented in 1968 by Pfizer and approved for medical use in 1979.[6] It became generic in 1992,[7] and is marketed worldwide under many brandnames.[1]

Medical uses

It is used in the treatment of certain inflammatory conditions like rheumatoid and osteoarthritis, primary dysmenorrhoea, postoperative pain; and act as an analgesic, especially where there is an inflammatory component.[3] The European Medicines Agency issued a review of its use in 2007 and recommended that its use be limited to the treatment of chronic inflammatory conditions, as it is only in these circumstances that its risk-benefit ratio proves to be favourable.[5][8]

Adverse effects

See also: Nonsteroidal anti-inflammatory drug

As with other NSAIDs the principal side effects include: digestive complaints like nausea, discomfort, diarrhoea and bleeds or ulceration of the stomach, as well as headache, dizziness, nervousness, depression, drowsiness, insomnia, vertigo, hearing disturbances (such as tinnitus), high blood pressureoedema, light sensitivity, skin reactions (including, albeit rarely, Stevens–Johnson syndrome and toxic epidermal necrolysis) and rarely, kidney failurepancreatitisliver damage, visual disturbances, pulmonary eosinophilia and alveolitis.[5] Compared to other NSAIDs it is more prone to causing gastrointestinal disturbances and serious skin reactions.[5]

In October 2020, the U.S. Food and Drug Administration (FDA) required the drug label to be updated for all nonsteroidal anti-inflammatory medications to describe the risk of kidney problems in unborn babies that result in low amniotic fluid.[9][10] They recommend avoiding NSAIDs in pregnant women at 20 weeks or later in pregnancy.[9][10]

Mechanism of action

See also: Nonsteroidal anti-inflammatory drug

Piroxicam is an NSAID and, as such, is a non-selective COX inhibitor possessing both analgesic and antipyretic properties.[5]

Chemical properties

Piroxicam exists as alkenol tautomer in organic solvents and as zwitterionic form in water.[11]

History

The project that produced piroxicam began in 1962 at Pfizer; the first clinical trial results were reported in 1977, and the product launched in 1980 under the brand name “Feldene”.[7][12] Major patents expired in 1992[7] and the drug is marketed worldwide under many brandnames.[1]

NMR

piroxicam usp 36322-90-4 wiki
piroxicam usp 36322-90-4 wiki

SYN

https://pubs.acs.org/doi/10.1021/jp1084444

Influence of Structure on the Spectroscopic Properties of the Polymorphs of Piroxicam

SYN

https://www.sciencedirect.com/science/article/abs/pii/S092420310400058X?via%3

PATENT

CN 101210013

https://patents.google.com/patent/CN101210013A/enIn the glassed steel reaction vessels of 2000L, add first ethyl ester thing 140Kg, dimethylbenzene 1500L, silica gel 10Kg.Be warming up to 100 ℃ of amino pyrrole 52Kg of adding 2-, continue to be warming up to the solvent refluxing temperature, keep refluxing slowly, steam the ethanol of reaction generation and the mixture of dimethylbenzene simultaneously, TLC follows the tracks of reaction, and reaction in 4.5-5 hour finishes.Underpressure distillation, the control temperature in the kettle is no more than 70 ℃, when the system volume be about cumulative volume 1/3 the time stop distillation, be cooled to normal temperature, stir 6-8h and filter, be i.e. crude product.Crude product adds methyl alcohol 1500L and adds the 15Kg gac, refluxes 30 minutes, filters, and is cooled to normal temperature, stirs 6-8h, methyl alcohol drip washing, 60-70 ℃ is dried by the fire 3-5h, measure product 140.5Kg, yield 85%.Press Cp2005 version standard detection, outward appearance; Off-white color, content 〉=99%.Methanol mother liquor reclaims methyl alcohol to overall 1/3 o’clock, and cooling stirring at normal temperature 6-8h filters and collects product, oven dry measure product 10Kg, yield 5.7%, this product meet the Cp2005 version and require to add up to yield.Add up to yield 90.7%.PAPER Bulletin of the Korean Chemical Society, 26(11), 1771-1775; 2005 

SYN

File:Piroxicam synthesis.svg - Wikimedia Commons
CAS-RNFormulaChemical NameCAS Index Name
504-29-0C5H6N22-aminopyridine2-Pyridinamine
79-04-9C2H2Cl2Ochloroacetyl chlorideAcetyl chloride, chloro-
29209-30-1C11H11NO5S3,4-dihydro-2-methyl-4-oxo-2H-1,2-benzothiazine-3-carboxylic acid methyl ester 1,1-dioxide2H-1,2-Benzothiazine-3-carboxylic acid, 3,4-dihydro-2-methyl-4-oxo-, methyl ester, 1,1-dioxide
29209-29-8C10H9NO5S3-methoxycarbonyl-4-oxo-3,4-dihydro-2H-1,2-benzothiazine 1,1-dioxide2H-1,2-Benzothiazine-3-carboxylic acid, 3,4-dihydro-4-oxo-, methyl ester, 1,1-dioxide
  1. Drebushchak, V. A.; Journal of Thermal Analysis and Calorimetry 2006, V84(3), P643-649 
  2.  Gehad, G. Mohamed; Vibrational Spectroscopy 2004, V36(1), P97-104 
  3.  Pajula, Katja; Molecular Pharmaceutics 2010, V7(3), P795-804 
  4.  Wassvik, Carola M.; European Journal of Pharmaceutical Sciences 2006, V29(3-4), P294-305
  5.  Wassvik, Carola M.; Journal of Medicinal Chemistry 2008, V51(10), P3035-3039
  6.  Zayed, M. A.; Spectrochimica Acta, Part A: Molecular and Biomolecular Spectroscopy 2004, V60A(12), P2843-2852 
  7.  Zia-ur-Rehman, Muhammad; Bulletin of the Korean Chemical Society 2005, V26(11), P1771-1775 
  8.  “Drugs – Synonyms and Properties” data were obtained from Ashgate Publishing Co. (US) 
  9.  Stulzer, H. K.; Pharmaceutical Chemistry Journal 2008, V42(4), P215-219 CAPLUS
  10.  Drebushchak, V. A.; Journal of Thermal Analysis and Calorimetry 2006, V86(2), P303-309 
  11.  Hughes, Laura D.; Journal of Chemical Information and Modeling 2008, V48(1), P220-232 
  12.  Laban, Gunter; DD 260398 A3 1988 
  13.  Svoboda, Jiri; Collection of Czechoslovak Chemical Communications 1986, V51(5), P1133-9 
  14. (26) Perillo, Isabel A.; Journal of Heterocyclic Chemistry 1983, V20(1), P155-60 
  15.  Zak, Bohumil; CS 276217 B6 1992 CAPLUS
  16.  Dalla Croce, Piero; Journal of Chemical Research, Synopses 1986, (4), P150-1
  17.  Vemavarapu, Chandra; Powder Technology 2009, V189(3), P444-453 
  18.  Sanghavi, N. M.; Indian Journal of Technology 1989, V27(2), P93-5 
  19.  “PhysProp” data were obtained from Syracuse Research Corporation of Syracuse, New York (US)
  20. Mohamed, Gehad G.; Spectrochimica Acta, Part A: Molecular and Biomolecular Spectroscopy 2004, V60A(13), P3141-3154 
  21.  Zayed, M. A.; Spectrochimica Acta, Part A: Molecular and Biomolecular Spectroscopy 2006, V64A(1), P216-232 
  22.  Habibi-Yangjeh, Aziz; Bulletin of the Korean Chemical Society 2008, V29(4), P833-841 
  23. Mahlin, Denny; Molecular Pharmaceutics 2011, V8(2), P498-506 
  24.  Kozjek, Franc; Acta Pharmaceutica Jugoslavica 1985, V35(4), P275-81 
  25.  Laban, Gunter; DD 258532 A3 1988 
  26.  Caira, Mino R.; Journal of Pharmaceutical Sciences 1998, V87(12), P1608-1614 
  27.  Mohamed, Gehad G.; Spectrochimica Acta, Part A: Molecular and Biomolecular Spectroscopy 2005, V62A(4-5), P1165-1171 
  28.  Lin, Yannan; Journal of Pharmaceutical and Biomedical Analysis 2010, V51(4), P979-984 

References

  1. Jump up to:a b c Drugs.com Drugs.com international listings for piroxicamPage accessed July 3, 2015
  2. ^ https://www.ema.europa.eu/documents/psusa/piroxicam-list-nationally-authorised-medicinal-products-psusa/00002438/202004_en.pdf
  3. Jump up to:a b c d e f g Brayfield, A, ed. (14 January 2014). “Piroxicam”Martindale: The Complete Drug Reference. London, UK: Pharmaceutical Press. Retrieved 24 June 2014.
  4. ^ “TGA Approved Terminology for Medicines, Section 1 – Chemical Substances” (PDF). Therapeutic Goods Administration, Department of Health and Ageing, Australian Government. July 1999: 97.
  5. Jump up to:a b c d e Joint Formulary Committee (2013). British National Formulary (BNF) (65 ed.). London, UK: Pharmaceutical Press. pp. 665, 673–674ISBN 978-0-85711-084-8.
  6. ^ Fischer, Jnos; Ganellin, C. Robin (2006). Analogue-based Drug Discovery. John Wiley & Sons. p. 519. ISBN 9783527607495.
  7. Jump up to:a b c Lombardino, JG; Lowe, JA 3rd (2004). “The role of the medicinal chemist in drug discovery–then and now”. Nat Rev Drug Discov3 (10): 853–62. doi:10.1038/nrd1523PMID 15459676S2CID 11225541.. See: [1] Box 1: Discovery of piroxicam (1962–1980)
  8. ^ “COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) OPINION FOLLOWING AN ARTICLE 31(2) REFERRAL PIROXICAM CONTAINING MEDICINAL PRODUCTS” (PDF). European Medicines Agency. London, UK: European Medicines Agency. 20 September 2007. Retrieved 24 June 2014.
  9. Jump up to:a b “FDA Warns that Using a Type of Pain and Fever Medication in Second Half of Pregnancy Could Lead to Complications”U.S. Food and Drug Administration (FDA) (Press release). 15 October 2020. Retrieved 15 October 2020. Public Domain This article incorporates text from this source, which is in the public domain.
  10. Jump up to:a b “NSAIDs may cause rare kidney problems in unborn babies”U.S. Food and Drug Administration. 21 July 2017. Retrieved 15 October 2020. Public Domain This article incorporates text from this source, which is in the public domain.
  11. ^ Ivanova D, Deneva V, Nedeltcheva D, Kamounah FS, Gergov G, Hansen PE, Kawauchi S, Antonov L (2015). “Tautomeric transformations of piroxicam in solution: a combined experimental and theoretical study”RSC Advances5 (40): 31852–31860. doi:10.1039/c5ra03653d.
  12. ^ Weintraub M, Jacox RF, Angevine CD, Atwater EC (1977). “Piroxicam (CP 16171) in rheumatoid arthritis: a controlled clinical trial with novel assessment techniques”. Journal of Rheumatology4 (4): 393–404. PMID 342691.

Further reading

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

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

Clinical data
Pronunciation/paɪˈrɒksɪˌkæm/
Trade namesFeldene, others[1]
Other namesPiroksikam, piroxikam
AHFS/Drugs.comMonograph
MedlinePlusa684045
Pregnancy
category
AU: C
Routes of
administration
By mouth
ATC codeM01AC01 (WHOM02AA07 (WHO), S01BC06 (WHO)
Legal status
Legal statusAU: S4 (Prescription only)CA℞-onlyUK: POM (Prescription only)US: ℞-onlyEU: Rx-only [2]
Pharmacokinetic data
Protein binding99%[3]
MetabolismLiver-mediated hydroxylation and glucuronidation[3]
Elimination half-life50 hours[3]
ExcretionUrine, faeces
Identifiers
showIUPAC name
CAS Number36322-90-4 
PubChem CID54676228
IUPHAR/BPS7273
DrugBankDB00554 
ChemSpider10442653 
UNII13T4O6VMAM
KEGGD00127 
ChEBICHEBI:8249 
ChEMBLChEMBL527 
CompTox Dashboard (EPA)DTXSID5021170 
ECHA InfoCard100.048.144 
Chemical and physical data
FormulaC15H13N3O4S
Molar mass331.35 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI
  (verify)

///////////PIROXICAM

NEW DRUG APPROVALS

ONE TIME

$10.00

PIRACETAM


Piracetam.svg

Piracetam

  • ATC:N06BX03
  • MW:142.16 g/mol
  • CAS-RN:7491-74-9
  • InChI Key:GMZVRMREEHBGGF-UHFFFAOYSA-N
  • InChI:InChI=1S/C6H10N2O2/c7-5(9)4-8-3-1-2-6(8)10/h1-4H2,(H2,7,9)
  • EINECS:231-312-7
  • LD50:9200 mg/kg (M, i.v.); 2 g/kg (M, p.o.)

CAS Registry Number: 7491-74-9 
CAS Name: 2-Oxo-1-pyrrolidineacetamide 
Additional Names: 2-pyrrolidoneacetamide; 2-pyrrolidinoneacetamide; 2-ketopyrrolidine-1-ylacetamide; 1-acetamido-2-pyrrolidinone 
Manufacturers’ Codes: UCB-6215 
Trademarks: Avigilen (Riemser); Axonyl (Pfizer); Cerebroforte (Azupharma); Encetrop (Alpharma); Gabacet (Sanofi-Synthelabo); Geram (UCB); Nootrop (UCB); Nootropil (UCB); Nootropyl (UCB); Norzetam (UCB); Normabraïn (UCB); Piracebral (Hexal); Piracetrop (Holsten); Sinapsan (Rodleben)Molecular Formula: C6H10N2O2 
Molecular Weight: 142.16 
Percent Composition: C 50.69%, H 7.09%, N 19.71%, O 22.51% 
Literature References: Prepn: H. Morren, NL6509994eidem,US3459738 (1966, 1969 both to U.C.B.). Pharmacology: Giurgea et al.,Arch. Int. Pharmacodyn. Ther.166, 238 (1967); Giurgea, Moyersoons, ibid.188, 401 (1970); Giurgea et al.,Psychopharmacologia20, 160 (1971). Metabolism and biochemical studies: Gobert, J. Pharm. Belg.27, 281 (1972). Clinical studies: W. J. Oosterveld, Arzneim.-Forsch.30, 1947 (1980); G. Chouinard et al.,Psychopharmacol. Bull.17, 129 (1981); in dyslexia: M. Di Ianni et al.,J. Clin. Psychopharmacol.5, 272 (1985).Properties: Crystals from isopropanol, mp 151.5-152.5°. 
Melting point: mp 151.5-152.5° 
Therap-Cat: Nootropic. 
Keywords: Nootropic.

Piracetam is in the racetams group, with chemical name 2-oxo-1-pyrrolidine acetamide. It is a derivative of the neurotransmitter GABA[5] and shares the same 2-oxo-pyrrolidone base structure with pyroglutamic acid. Piracetam is a cyclic derivative of GABA (gamma-aminobutyric acid). Related drugs include the anticonvulsants levetiracetam and brivaracetam, and the putative nootropics aniracetam and phenylpiracetam.Piracetam is a drug marketed as a treatment for myoclonus[3] and a cognitive enhancer.[4] Evidence to support its use is unclear, with some studies showing modest benefits in specific populations and others showing minimal or no benefit.[5][6] Piracetam is sold as a medication in many European countries. Sale of piracetam is not illegal in the United States, although it is not regulated nor approved by the FDA so it must be marketed as a dietary supplement.[4]

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

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

Efficacy

Dementia

A 2001 Cochrane review concluded that there was not enough evidence to support piracetam for dementia or cognitive problems.[6] A 2005 review found some evidence of benefit in older subjects with cognitive impairment.[5] In 2008, a working group of the British Academy of Medical Sciences noted that many of the trials of piracetam for dementia were flawed.[7]

There is no good evidence that piracetam is of benefit in treating vascular dementia.[8]

Depression and anxiety

Some sources suggest that piracetam’s overall effect on lowering depression and anxiety is higher than on improving memory.[9] However, depression is reported to be an occasional adverse effect of piracetam.[10]

Other

Piracetam may facilitate the deformability of erythrocytes in capillary which is useful for cardiovascular disease.[5][3]

Peripheral vascular effects of piracetam have suggested its use potential for vertigodyslexiaRaynaud’s phenomenon and sickle cell anemia.[5][3] There is no evidence to support piracetam’s use in sickle cell crisis prevention[11] or for fetal distress during childbirth.[12] There is no evidence for benefit of piracetam with acute ischemic stroke,[13] though there is debate as to its utility during stroke rehabilitation.[14][15]

Anti-vasospasm

Piracetam has been found to diminish erythrocyte adhesion to vascular wall endothelium, making any vasospasm in the capillary less severe. This contributes to its efficacy in promoting microcirculation, including to the brain and kidneys.[5][3]

Side effects

Symptoms of general excitability, including anxietyinsomniairritabilityheadacheagitationnervousnesstremor, and hyperkinesia, are occasionally reported.[10][16][17] Other reported side effects include somnolenceweight gainclinical depressionweakness, increased libido, and hypersexuality.[10]

According to a 2005 review, piracetam has been observed to have the following side effects: hyperkinesia, weight gain, nervousness, somnolence, depression and asthenia.[5]

Piracetam reduces platelet aggregation as well as fibrinogen concentration, and thus is contraindicated to patients suffering from cerebral hemorrhage.[5][3]

Toxicity

Piracetam does not appear to be acutely toxic at the doses used in human studies.[6][18][19]

The LD50 for oral consumption in humans has not been determined.[20] The LD50 is 5.6 g/kg for rats and 20 g/kg for mice, indicating extremely low acute toxicity.[21] For comparison, in rats the LD50 of vitamin C is 12 g/kg and the LD50 of table salt is 3 g/kg.

Mechanisms of action

Piracetam’s mechanism of action, as with racetams in general, is not fully understood. The drug influences neuronal and vascular functions and influences cognitive function without acting as a sedative or stimulant.[5] Piracetam is a positive allosteric modulator of the AMPA receptor, although this action is very weak and its clinical effects may not necessarily be mediated by this action.[22] It is hypothesized to act on ion channels or ion carriers, thus leading to increased neuron excitability.[20] GABA brain metabolism and GABA receptors are not affected by piracetam[23]

Piracetam improves the function of the neurotransmitter acetylcholine via muscarinic cholinergic (ACh) receptors[citation needed], which are implicated in memory processes.[24] Furthermore, piracetam may have an effect on NMDA glutamate receptors, which are involved with learning and memory processes. Piracetam is thought to increase cell membrane permeability.[24][25] Piracetam may exert its global effect on brain neurotransmission via modulation of ion channels (i.e., Na+, K+).[20] It has been found to increase oxygen consumption in the brain, apparently in connection to ATP metabolism, and increases the activity of adenylate kinase in rat brains.[26][27] Piracetam, while in the brain, appears to increase the synthesis of cytochrome b5,[28] which is a part of the electron transport mechanism in mitochondria. But in the brain, it also increases the permeability of some intermediates of the Krebs cycle through the mitochondrial outer membrane.[26]

Piracetam inhibits N-type calcium channels. The concentration of piracetam achieved in central nervous system after a typical dose of 1200 mg (about 100 μM)[29] is much higher than the concentration necessary to inhibit N-type calcium channels (IC50 of piracetam in rat neurons was 3 μM).[30]

History

Piracetam was first made some time between the 1950s and 1964 by Corneliu E. Giurgea.[31] There are reports of it being used for epilepsy in the 1950s.[32]

Society and culture

In 2009 piracetam was reportedly popular as a cognitive enhancement drug among students.[33]

Legal status

Piracetam is an uncontrolled substance in the United States meaning it is legal to possess without a license or prescription.[34]

Regulatory status

In the United States, piracetam is not approved by the Food and Drug Administration.[1] Piracetam is not permitted in compounded drugs or dietary supplements in the United States.[35] Nevertheless, it is available in a number of dietary supplements.[4]

In the United Kingdom, piracetam is approved as a prescription drug Prescription Only Medicine (POM) number is PL 20636/2524[36] for adult with myoclonus of cortical origin, irrespective of cause, and should be used in combination with other anti-myoclonic therapies.[37]

In Japan piracetam is approved as a prescription drug.[38]

Piracetam has no DIN in Canada, and thus cannot be sold but can be imported for personal use in Canada.[39]

In Hungary, piracetam was a prescription-only medication, but as of 2020, no prescription is required and piracetam is available as an over-the-counter drug under the name Memoril Mite, and is available in 600 mg pills.

According to the literature reports, the synthetic route of piracetam can be divided into four synthetic methods: α-pyrrolidone method, glycine method, succinic anhydride method and one-step synthesis method:[0009] I. α-pyrrolidone method, 2-pyrrolidone is a lactam, which can react with a strong base (sodium hydride or potassium hydride, sodium methoxide) to generate pyrrolidone metal salt, which can be further combined with halogenated ester or halogen Substitute amide reaction to generate N-alkylated product.[0010] In 1966, a method for preparing piracetam by reacting pyrrolidone and chloroacetamide in 1,4-dioxane with sodium hydrogen as a strong base was reported. The specific synthetic route is shown in Scheme 1:[0011]

Figure CN104478779AD00032

[0012] In this process, due to the high price of dioxane, industrial production is still difficult. On the basis of the above process, Xu Yungen used dimethyl sulfoxide as the solvent and sodium methoxide as the acid binding agent to synthesize piracetam in the presence of the phase transfer catalyst benzyltriethylammonium chloride. Due to the difficulty of solvent recovery, the cost of this route is relatively high.[0013] In 1981, Zhou Renxing et al. used sodium methoxide as a strong base to extract methanol in toluene by fractional distillation to convert pyrrolidone into the corresponding sodium salt, and then react with ethyl chloroacetate. The resulting ethyl pyrrolidone ethyl acetate was subjected to ammonolysis. Piracetam can be produced. The specific synthetic route is shown in Scheme 2.[00141

Figure CN104478779AD00041

[0015] Because the ammonolysis is carried out in a methanol solution of ammonia, the calculated amount of ethanol generated during the ammonolysis contaminates the methanol solution of ammonia used, which affects the recycling of the methanol solution of ammonia, and is therefore not conducive to process production.[0016] 2. Glycine method, glycine and its derivatives can be used as starting materials for the synthesis of pyroacetamide. Glycine can be prepared by γ-chlorination butylation, amination and cyclization.[0017] According to a British patent report in 1979, glycine trimethylsilyl ester was first condensed with γ-chlorobutyryl chloride, and the corresponding acid chloride was subjected to ammonolysis, and finally cyclized to produce piracetam. The specific synthesis method is as Scheme 3 Shown[0018]

Figure CN104478779AD00042

[0019] In this type of synthesis route, some raw materials are not easily available, which restricts industrial production.[0020] 3. Succinic acid method, succinic acid is heated and dehydrated to generate succinic anhydride, succinic anhydride then reacts with glycine to generate an aminolysis product, and the aminolysis product is reduced by sodium tetrafluoroborate, and piracetam can be synthesized by aminolysis , The specific synthetic route is shown in SCheme4. [0021]

Figure CN104478779AD00043

[0022] Because sodium tetrafluoroborate is used as a reducing agent, it is expensive, and it is difficult to expand the scale of industrial production. Succinimide generates sodium salt under the action of metal sodium, and its sodium salt reacts with chloroacetamide to generate N-alkylated product. The alkylated product can be electrolytically reduced to obtain piracetam. Since electrolytic reduction is still in the research stage in our country, the production cost of this method is relatively high.[0023] 4. One-step synthesis method, using ethyl 4-chloro-n-butyrate in the presence of sodium bicarbonate, using anhydrous ethanol as a solvent, and glycinamide hydrochloride under heating and refluxing to obtain piracetam in one step, The specific synthetic route is shown in S Cheme5.[0024]

Figure CN104478779AD00044

[0025] In this route, glycinamide hydrochloride is very easy to absorb moisture and agglomerate to affect the reaction rate, and the reaction is not easy to control, so it is difficult to achieve industrial production.

SYN

File:Piracetam synthesis02.svg - Wikimedia Commons
File:Piracetam synthesis01.svg

SYN

http://www.cjph.com.cn/EN/abstract/abstract373.shtml

With absolute ethanol as the solvent, ethyl 4-chloro-n-butanoate and glycinamide hydrochloride were refluxed for 20 h in the presence of sodium bicarbonate to obtain central stimulant piracetam. After recrystallization from isopropanol, the yield was about 58% with a purity of 99.6%.

CN104478779A - 促智药吡拉西坦的合成新方法 - Google Patents

SYN

CAS-RNFormulaChemical NameCAS Index Name
79-07-2C2H4ClNO2-chloroacetamideAcetamide, 2-chloro-
105-39-5C4H7ClO2ethyl chloroacetateAcetic acid, chloro-, ethyl ester
61516-73-2C8H13NO3ethyl 2-oxo-1-pyrrolidineacetate1-Pyrrolidineacetic acid, 2-oxo-, ethyl ester
616-45-5C4H7NO2-pyrrolidone2-Pyrrolidinone

PATENT

https://patents.google.com/patent/CN104478779A/zh

Figure CN104478779AD00051

Example 1[0036] A method for synthesizing piracetam, which includes the following steps:[0037] Preparation of α-pyrrolidone sodium salt: A 1000 mL three-necked flask was equipped with mechanical stirring, a constant pressure dropping funnel and a thorn-shaped fractionating column. The upper end of the fractionation column is connected with a thermometer, a condenser and a 500mL receiving flask. Under mechanical stirring, 46 mL (0.60 mol) of α-pyrrolidone and 250 mL of toluene were sequentially added to the three-necked flask. When the temperature of the reaction system reached 70°C, a methanol solution of sodium methoxide (28.4% (w/w); 114.0 g; 0.60 mol) was added dropwise under reduced pressure, and the distillate was collected. After the dropwise addition is completed, the temperature is increased, and the normal pressure is distilled until the distillate is completely distilled out, and the reaction is completed.[0038] Preparation of α-pyrrolidone methyl acetate: remove the fractionation device, connect a thermometer and a condenser, and connect a dropping funnel above the condenser. When the temperature of the reaction system drops to 60°C, a toluene solution of 58 mL (0.66 mol) of methyl chloroacetate is slowly added dropwise, and the reaction temperature is controlled to 80-100°C. Oh。 After the addition is complete, the insulation reaction is 5. Oh. Cool to room temperature, filter with suction, and distill the filtrate under reduced pressure. Collect the fraction (18mmHg) at 100~105°C to obtain α-pyrrolidone methyl acetate, and measure its content by HPLC (area normalization method). [C18 column (4.6mmX 200mm, 5 μm) was used for purity determination; acetonitrile-dipotassium hydrogen phosphate/phosphate buffer solution (10:90) was used as the mobile phase (the pH value of phosphoric acid was adjusted to 6.0); the flow rate was 1 . OmL/min; detection wavelength is 205nm; injection volume is 20yL][0039] Preparation of Piracetam: Put about 130 mL of methanol in a 500 mL three-necked flask, and vent ammonia to saturation. The obtained ammonia/methanol solution was mixed with 100. Og α-pyrrolidone methyl acetate and placed in a reaction kettle, reacted at 50~65°C for 10 h, allowed to cool, filtered with suction, and the filter cake was dried.[0040] The purification of piracetam: 25.50g crude piracetam and 100mL isopropanol were sequentially added in a 500mL three-necked flask, heated to reflux for 40min, activated carbon was added, reflux stirring, hot filtration, and the resulting properties were all white As a powdery solid, the filter cake was dried overnight at 50°C in a vacuum drying oven to obtain 20.85 g of a white solid with a yield of 81.76% (calculated as α-pyrrolidone, the same below).Example 2[0042] Preparation of α-pyrrolidone sodium salt: A 1000 mL three-necked flask was equipped with mechanical stirring, a constant pressure dropping funnel and a thorn-shaped fractionating column. The upper end of the fractionation column is connected with a thermometer, a condenser and a 500mL receiving flask. Under mechanical stirring, 46 mL (0.60 mol) of α-pyrrolidone and 250 mL of toluene were sequentially added to the three-necked flask. When the temperature of the reaction system reached 100°C, a methanol solution of sodium methoxide (28.4% (w/w)); 114. Og; 0.60 mol) was added dropwise under reduced pressure, and the distillate was collected. After the addition is complete, add toluene, increase the temperature, and distill at normal pressure until the distillate is completely distilled out, and the reaction is complete.[0043] Preparation of α-pyrrolidone methyl acetate: remove the fractionation device, connect a thermometer and a condenser, and connect a dropping funnel above the condenser. When the temperature of the reaction system drops to 60°C, a mixed solution of 63 mL (0.72 mol) of methyl chloroacetate and 30 mL of toluene is slowly added dropwise, and the reaction temperature is controlled to 80-100°C. Oh。 After the addition is complete, the insulation reaction is 5. Oh. Cool to room temperature, filter with suction, and distill the filtrate under reduced pressure. Collect the fraction (18mmHg) at 100~105°C to obtain methyl α-pyrrolidone acetate, and measure its content by HPLC (area normalization method). [C18 column (4.6mmX 200mm, 5 μm) was used for purity determination; acetonitrile-dipotassium hydrogen phosphate/phosphate buffer solution (10:90) was used as the mobile phase (the pH value of phosphoric acid was adjusted to 6.0); the flow rate was 1 .OmL/ min; detection wavelength is 205nm; injection volume is 20 μL][0044] Preparation of Piracetam: Put about 130 mL of methanol in a 250 mL three-necked flask, and ventilate ammonia to saturation. The obtained ammonia/methanol solution was mixed with 50.0 g of α-pyrrolidone methyl acetate and placed in a reaction kettle, reacted at 50~65°C for 12 hours, allowed to cool, filtered with suction, and the filter cake was dried.[0045] Purification of piracetam: 25.50g crude piracetam and 75mL methanol were sequentially added to a 500mL three-necked flask, heated to reflux for 40min, added activated carbon 0.5g, refluxed for 1h, hot filtered, magnetically stirred Under the conditions, the activated carbon was filtered out, and the properties were all white powdery solids, and the filter cake was dried overnight at 50°C in a vacuum drying oven to obtain 21.02g of white solids with a yield of 82.42%.Embodiment 3[0047] Preparation of α-pyrrolidone sodium salt: A 1000 mL three-necked flask was equipped with mechanical stirring, a constant pressure dropping funnel and a thorn-shaped fractionating column. The upper end of the fractionating column is connected with a thermometer, a condenser and a 1000 mL receiving bottle. Under mechanical stirring, 46 mL (0.60 mol) of α-pyrrolidone and 250 mL of toluene were sequentially added to the three-necked flask. When the temperature of the reaction system reached 70°C, a methanol solution of sodium methoxide (28.4% (w/w)); 114. Og; 0.60 mol) was added dropwise under reduced pressure, and the distillate was collected. After the dropwise addition is completed, the temperature is increased, and the normal pressure is distilled until the distillate is completely distilled out, and the reaction is completed.[0048] Preparation of α-pyrrolidone methyl acetate: remove the fractionation device, connect a thermometer and a condenser, and connect a dropping funnel above the condenser. A mixed solution of 79 mL (0.90 mol) of methyl chloroacetate and 50 mL of toluene was slowly added dropwise, and the reaction temperature was controlled to 70-90°C. Oh。 After the addition is complete, the insulation reaction is 5. Oh. Cool to room temperature, filter with suction, and distill the filtrate under reduced pressure. Collect the fraction (18mmHg) at 100~105°C to obtain methyl α-pyrrolidone acetate, and measure its content by HPLC (area normalization method). [C 18 column (4.6mmX 200mm, 5 μm) was used for purity determination; acetonitrile-dipotassium hydrogen phosphate/phosphate buffer solution (10:90) was used as the mobile phase (the pH value of phosphoric acid was adjusted to 6.0); the flow rate was 1.0mL/min; The detection wavelength is 205nm; The injection volume is 20 μL)[0049] Preparation of Piracetam: Put about 130 mL of methanol in a 250 mL three-necked flask, and vent ammonia to saturation. The obtained ammonia/methanol solution was mixed with 100. Og α-pyrrolidone methyl acetate and placed in a reaction kettle, reacted at 50~65°C for 14h, allowed to cool, filtered with suction, and the filter cake was dried.[0050] Purification of piracetam: 25.50g crude piracetam and 125mL ethanol were sequentially added in a 500mL three-necked flask, heated to reflux for 40min, added activated carbon 0.5g, refluxed for 1h, hot filtered, magnetically stirred Activated carbon was filtered off under conditions to obtain white powdery solids in all properties, and the filter cake was dried overnight at 50°C in a vacuum drying oven to obtain 20.24 g of white solids with a yield of 79.37%.Example 4[0052] Preparation of α-pyrrolidone sodium salt: A 1000 mL three-necked flask was equipped with mechanical stirring, a constant pressure dropping funnel and a thorn-shaped fractionating column. The upper end of the fractionation column is connected with a thermometer, a condenser and a 500mL receiving flask. Under mechanical stirring, 46 mL (0.60 mol) of α-pyrrolidone and 250 mL of toluene were sequentially added to the three-necked flask. When the temperature of the reaction system reached 60°C, a methanol solution of sodium methoxide (28.4% (w/w); 114.0 g; 0.60 mol) was added dropwise under reduced pressure, and the distillate was collected. After the dropwise addition is completed, the temperature is increased, and the normal pressure is distilled until the distillate is completely distilled out, and the reaction is completed.[0053] Preparation of α-pyrrolidone methyl acetate: remove the fractionation device, connect a thermometer and a condenser, and connect a dropping funnel above the condenser. A mixed solution of 105 mL (1.20 mol) of methyl chloroacetate and 70 mL of toluene was slowly added dropwise, and the reaction temperature was controlled to be 60~70°C. Oh。 After the addition is complete, the insulation reaction is 5. Oh. Cool to room temperature, filter with suction, and distill the filtrate under reduced pressure. Collect the fraction (18mmHg) at 100~105°C to obtain methyl α-pyrrolidone acetate, and measure its content by HPLC (area normalization method). [C 18 column (4.6mmX 200mm, 5 μm) was used for purity determination; acetonitrile-dipotassium hydrogen phosphate/phosphate buffer solution (10:90) was used as the mobile phase (the pH value of phosphoric acid was adjusted to 6.0); the flow rate was 1.0mL/min; The detection wavelength is 205nm; The injection volume is 20 μL)[0054] Preparation of Piracetam: Put about 130 mL of methanol in a 500 mL three-necked flask, and ventilate ammonia to saturation. The obtained ammonia/methanol solution was mixed with 100. Og α-pyrrolidone methyl acetate and placed in a reaction kettle, reacted at 50~65°C for 16h, allowed to cool, filtered with suction, and the filter cake was dried.[0055] The purification of piracetam: 25.50g crude piracetam and 100mL methanol were sequentially added into a 500mL three-necked flask, heated to reflux for 40min, added activated carbon, refluxed for dissolution, hot filtered, and the properties were all white powders The solid, the filter cake was dried overnight at 50°C in a vacuum drying oven to obtain 20.69 g of a white solid, with a yield of 81. 13%.[0056] Chemical analysis of the white crystals synthesized in each of the foregoing examples, and the obtained physical property values are as follows, thereby confirming that the synthesized product is piracetam.[0057] Melting point: 151.6-152. (TC[0058] ESI-MS m / z: 165. 06 [M + Na] +[0059] 1H-NMR (400MHz, DMS〇-d6, ppm) δ : 7. 38 (s, 1H), 7. 09 (s, 1H), 3. 74 (s, 2H), 3. 36 (t, J =7. 08Hz, 2H), 2. 23 (t, J = 7. 84Hz, 2H), I. 93 (m, 2H).[0060] 13C-NMR(100MHz, DMS0-d6, ppm) δ : 17. 80, 30. 42, 45. 28, 47. 74, 170. 21,174. 90. 
PATENTCN110903230A *2019-12-042020-03-24Beijing Yuekang Kechuang Pharmaceutical Technology Co., Ltd.An industrialized preparation method of Pramiracetam sulfate 
PATENTCN104478779A2015-04-01New synthetic method of nootropic drug Piracetam

References

  1. Jump up to:a b “Piracetam”DrugBank database.
  2. ^ Leaflet of Piracetam.
  3. Jump up to:a b c d e “Nootropil Tablets 1200 mg”(emc). 15 February 2017. Retrieved 14 April 2019.
  4. Jump up to:a b c Cohen, Pieter A.; Zakharevich, Igor; Gerona, Roy (25 November 2019). “Presence of Piracetam in Cognitive Enhancement Dietary Supplements”JAMA Internal Medicine180 (3): 458–459. doi:10.1001/jamainternmed.2019.5507PMC 6902196PMID 31764936.
  5. Jump up to:a b c d e f g h i Winblad B (2005). “Piracetam: a review of pharmacological properties and clinical uses”CNS Drug Reviews11 (2): 169–82. doi:10.1111/j.1527-3458.2005.tb00268.xPMC 6741724PMID 16007238.
  6. Jump up to:a b c Flicker, L; Grimley Evans, G (2001). “Piracetam for dementia or cognitive impairment”. The Cochrane Database of Systematic Reviews (2): CD001011. doi:10.1002/14651858.CD001011PMID 11405971.
  7. ^ Horne G, et al. (May 2008). Brain science, addiction and drugs(PDF) (Report). Academy of Medical Sciences. p. 145. ISBN 978-1-903401-18-7.
  8. ^ Farooq MU, Min J, Goshgarian C, Gorelick PB (September 2017). “Pharmacotherapy for Vascular Cognitive Impairment”. CNS Drugs(Review). 31 (9): 759–776. doi:10.1007/s40263-017-0459-3PMID 28786085S2CID 23271739Other medications have been considered or tried for the treatment of VCI or VaD. These include […] piracetam. There is no convincing evidence about the efficacy of these medications in the treatment of VCI.
  9. ^ Malykh AG, Sadaie MR (February 2010). “Piracetam and piracetam-like drugs: from basic science to novel clinical applications to CNS disorders”. Drugs70 (3): 287–312. doi:10.2165/11319230-000000000-00000PMID 20166767S2CID 12176745.
  10. Jump up to:a b c Nootropil®. Arzneimittel-Kompendium der Schweiz. 2013-09-12. Retrieved 2013-10-27.
  11. ^ Al Hajeri A, Fedorowicz Z (February 2016). “Piracetam for reducing the incidence of painful sickle cell disease crises”The Cochrane Database of Systematic Reviews2: CD006111. doi:10.1002/14651858.CD006111.pub3PMC 7390168PMID 26869149.
  12. ^ Hofmeyr, GJ; Kulier, R (13 June 2012). “Piracetam for fetal distress in labour”The Cochrane Database of Systematic Reviews (6): CD001064. doi:10.1002/14651858.CD001064.pub2PMC 7048034PMID 22696322.
  13. ^ Ricci S, Celani MG, Cantisani TA, Righetti E (September 2012). “Piracetam for acute ischaemic stroke”The Cochrane Database of Systematic Reviews (9): CD000419. doi:10.1002/14651858.CD000419.pub3PMC 7034527PMID 22972044.
  14. ^ Zhang J, Wei R, Chen Z, Luo B (July 2016). “Piracetam for Aphasia in Post-stroke Patients: A Systematic Review and Meta-analysis of Randomized Controlled Trials”. CNS Drugs30 (7): 575–87. doi:10.1007/s40263-016-0348-1PMID 27236454S2CID 22955205.
  15. ^ Yeo SH, Lim ZI, Mao J, Yau WP (October 2017). “Effects of Central Nervous System Drugs on Recovery After Stroke: A Systematic Review and Meta-Analysis of Randomized Controlled Trials”. Clinical Drug Investigation37 (10): 901–928. doi:10.1007/s40261-017-0558-4PMID 28756557S2CID 6520934.
  16. ^ Chouinard G, Annable L, Ross-Chouinard A, Olivier M, Fontaine F (1983). “Piracetam in elderly psychiatric patients with mild diffuse cerebral impairment”. Psychopharmacology81 (2): 100–106. doi:10.1007/BF00429000PMID 6415738S2CID 32702769.
  17. ^ Hakkarainen H, Hakamies L (1978). “Piracetam in the treatment of post-concussional syndrome. A double-blind study”. European Neurology17 (1): 50–55. doi:10.1159/000114922PMID 342247.
  18. ^ Koskiniemi M, Van Vleymen B, Hakamies L, Lamusuo S, Taalas J (March 1998). “Piracetam relieves symptoms in progressive myoclonus epilepsy: a multicentre, randomised, double blind, crossover study comparing the efficacy and safety of three dosages of oral piracetam with placebo”Journal of Neurology, Neurosurgery, and Psychiatry64 (3): 344–348. doi:10.1136/jnnp.64.3.344PMC 2169975PMID 9527146.
  19. ^ Fedi M, Reutens D, Dubeau F, Andermann E, D’Agostino D, Andermann F (May 2001). “Long-term efficacy and safety of piracetam in the treatment of progressive myoclonus epilepsy”Archives of Neurology58 (5): 781–786. doi:10.1001/archneur.58.5.781PMID 11346373.
  20. Jump up to:a b c Gouliaev AH, Senning A (May 1994). “Piracetam and other structurally related nootropics”. Brain Research. Brain Research Reviews19 (2): 180–222. doi:10.1016/0165-0173(94)90011-6PMID 8061686S2CID 18122566.
  21. ^ “Piracetam Material Safety Sheet” (PDF). Spectrum.
  22. ^ Ahmed AH, Oswald RE (March 2010). “Piracetam defines a new binding site for allosteric modulators of alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptors”Journal of Medicinal Chemistry53 (5): 2197–203. doi:10.1021/jm901905jPMC 2872987PMID 20163115.
  23. ^ Giurgea CE (January 1982). “The nootropic concept and its prospective implications”. Drug Development Research2 (5): 441–446. doi:10.1002/ddr.430020505ISSN 1098-2299S2CID 145059666.
  24. Jump up to:a b Winnicka K, Tomasiak M, Bielawska A (2005). “Piracetam–an old drug with novel properties?”. Acta Poloniae Pharmaceutica62(5): 405–9. PMID 16459490.
  25. ^ Müller WE, Eckert GP, Eckert A (March 1999). “Piracetam: novelty in a unique mode of action”. Pharmacopsychiatry32 (Suppl 1): 2–9. doi:10.1055/s-2007-979230PMID 10338102.
  26. Jump up to:a b Grau M, Montero JL, Balasch J (1987). “Effect of Piracetam on electrocorticogram and local cerebral glucose utilization in the rat”. General Pharmacology18 (2): 205–11. doi:10.1016/0306-3623(87)90252-7PMID 3569848.
  27. ^ Nickolson VJ, Wolthuis OL (October 1976). “Effect of the acquisition-enhancing drug piracetam on rat cerebral energy metabolism. Comparison with naftidrofuryl and methamphetamine”. Biochemical Pharmacology25 (20): 2241–4. doi:10.1016/0006-2952(76)90004-6PMID 985556.
  28. ^ Tacconi MT, Wurtman RJ (1986). “Piracetam: physiological disposition and mechanism of action”. Advances in Neurology43: 675–85. PMID 3946121.
  29. ^ Yeh HH, Yang YH, Ko JY, Chen SH (July 2006). “Rapid determination of piracetam in human plasma and cerebrospinal fluid by micellar electrokinetic chromatography with sample direct injection”. J Chromatogr A1120 (1–2): 27–34. doi:10.1016/j.chroma.2005.11.071PMID 16343512.
  30. ^ Bravo-Martínez J, Arenas I, Vivas O, Rebolledo-Antúnez S, Vázquez-García M, Larrazolo A, García DE (October 2012). “A novel CaV2.2 channel inhibition by piracetam in peripheral and central neurons”. Exp Biol Med (Maywood)237 (10): 1209–18. doi:10.1258/ebm.2012.012128PMID 23045722.
  31. ^ Li JJ, Corey EJ (2013). Drug Discovery: Practices, Processes, and Perspectives. John Wiley & Sons. p. 276. ISBN 9781118354469.
  32. ^ Schmidt D, Shorvon S (2016). The End of Epilepsy?: A History of the Modern Era of Epilepsy Research 1860-2010. Oxford University Press. p. 69. ISBN 9780198725909.
  33. ^ Medew J (1 October 2009). “Call for testing on ‘smart drugs'”. Fairfax Media. Retrieved 29 May 2014.
  34. ^ “Erowid Piracetam Vault: Legal Status”.
  35. ^ Jann Bellamy (26 September 2019). “FDA proposes ban on curcumin and other naturopathic favorites in compounded drugs”Science-Based Medicine.
  36. ^http://www.mhra.gov.uk/home/groups/spcpil/documents/spcpil/con1547788739542.pdf
  37. ^ “Nootropil Tablets 800 mg”(emc).
  38. ^ “UCB’s piracetam approved in Japan”The Pharma Letter. 25 November 1999.
  39. ^ “Guidance Document on the Import Requirements for Health Products under the Food and Drugs Act and its Regulations (GUI-0084)”. Health Canada / Health Products and Food Branch Inspectorate. 1 June 2010. Retrieved 15 December 2019.

External links

Gouliaev AH, Senning A (May 1994). “Piracetam and other structurally related nootropics”. Brain Research. Brain Research Reviews19 (2): 180–222. doi:10.1016/0165-0173(94)90011-6PMID 8061686S2CID 18122566.

Clinical data
Trade namesBreinox, Dinagen, Lucetam, Nootropil, Nootropyl, Oikamid, Piracetam and many others
AHFS/Drugs.comInternational Drug Names
Routes of
administration
By mouth, parenteral, or vaporized
ATC codeN06BX03 (WHO)
Legal status
Legal statusAU: S4 (Prescription only)CA: UnscheduledUK: POM (Prescription only)US: Unscheduled (Not permitted as drug or supplement[1])
Pharmacokinetic data
Bioavailability~100%
Onset of actionSwiftly following administration. Food delays time to peak concentration by 1.5 h approximately to 2–3 h since dosing.[2]
Elimination half-life4–5 h
ExcretionUrinary
Identifiers
showIUPAC name
CAS Number7491-74-9 
PubChem CID4843
IUPHAR/BPS4288
DrugBankDB09210
ChemSpider4677 
UNIIZH516LNZ10
KEGGD01914 
ChEMBLChEMBL36715 
CompTox Dashboard (EPA)DTXSID5044491 
ECHA InfoCard100.028.466 
Chemical and physical data
FormulaC6H10N2O2
Molar mass142.158 g·mol−1
3D model (JSmol)Interactive image
Melting point152 °C (306 °F)
showSMILES
showInChI
  (verify)

///////////UCB 6215, Nootropic, PIRACETAM

wdt-8

NEW DRUG APPROVALS

ONE TIME

$10.00

MEROPENEM


Thumb
Meropenem | C17H25N3O5S - PubChem
Meropenem
Meropenem.svg

Meropenem

CAS number96036-03-2

IUPAC Name(4R,5S,6S)-3-{[(3S,5S)-5-(dimethylcarbamoyl)pyrrolidin-3-yl]sulfanyl}-6-[(1R)-1-hydroxyethyl]-4-methyl-7-oxo-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid

WeightAverage: 383.463
Monoisotopic: 383.151491615

Chemical FormulaC17H25N3O5S

  • Antibiotic SM 7338
  • ICI 194660
  • SM 7338

CAS Registry Number: 96036-03-2 
CAS Name: (4R,5S,6S)-3-[[(3S,5S)-5-[(Dimethylamino)carbonyl]-3-pyrrolidinyl]thio]-6-[(1R)-1-hydroxyethyl]-4-methyl-7-oxo-1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid 
Additional Names: (1R,5S,6S)-2-[(3S,5S)-5-(dimethylaminocarbonyl)pyrrolidin-3-ylthio]-6-[(R)-1-hydroxyethyl]-1-methylcarbapen-2-em-3-carboxylic acid 
Molecular Formula: C17H25N3O5S 
Molecular Weight: 383.46 
Percent Composition: C 53.25%, H 6.57%, N 10.96%, O 20.86%, S 8.36% 
Literature References: Carbapenem antibiotic. Prepn: M. Sunagawa et al.,EP126587; M. Sunagawa, US4943569 (1984, 1990 both to Sumitomo). 
Structure-activity study: M. Sunagawa et al.,J. Antibiot.43, 519 (1990).Crystal structure: K. Yanagi et al.,Acta Crystallogr.C48, 1737 (1992).HPLC determn in serum and bronchial secretions: M. Ehrlich et al., J. Chromatogr. B751, 357 (2001). Pharmacokinetics: R. Wise et al.,Antimicrob. Agents Chemother.34, 1515 (1990).Series of articles on antimicrobial activity, metabolism: J. Antimicrob. Chemother.24, Suppl. A, 1-320 (1989); and clinical performance: ibid.36, Suppl. A, 1-223 (1995).Review of clinical experience in intensive care: M. Hurst, H. M. Lamb, Drugs59, 653-680 (2000). 
Derivative Type: Trihydrate 
CAS Registry Number: 119478-56-7 
Manufacturers’ Codes: ICI-194660; SM-7338 
Trademarks: Meronem (AstraZeneca); Meropen (Sumitomo); Merrem (AstraZeneca) 
Properties: White to pale yellow crystalline powder. Sparingly sol in water; very slightly sol in hydrated ethanol. Practically insol in acetone, ether. 
Therap-Cat: Antibacterial. 
Keywords: Antibacterial (Antibiotics); ?Lactams; Carbapenems.

Product Ingredients

INGREDIENTUNIICASINCHI KEY
Meropenem sodiumNot Available211238-34-5UBQRNADYCUXRBD-NACOAMSHSA-N
Meropenem trihydrateFV9J3JU8B1119478-56-7CTUAQTBUVLKNDJ-OBZXMJSBSA-N

International/Other BrandsAronem (ACI) / Aropen (Aristopharma) / Carbanem (Sanofi-Aventis) / Erope (Lincoln) / Fulspec (Acme) / I-penam (Incepta) / Merenz (Admac) / Merofit (FHC) / Meronem (AstraZeneca) / Meronis (Neiss) / Meropen (Swiss Parenterals) / Merotec (Zuventus) / Merrem I.V. (AstraZeneca) / Monan (AstraZeneca) / Ropenem (Drug International) / Zeropenem (Sanofi-Aventis)

Synthesis Reference

Yoon Seok Song, Sung Woo Park, Yeon Jung Yoon, Hee Kyoon Yoon, Seong Cheol Moon, Byung Goo Lee, Soo Jin Choi, Sun Ah Jun, “METHOD FOR PREPARING MEROPENEM USING ZINC POWDER.” U.S. Patent US20120065392, issued March 15, 2012.

US20120065392

SYN

Carbapenem antibiotic. Prepn: M. Sunagawa et al., EP 126587; M. Sunagawa, US 4943569 (1984, 1990 both to Sumitomo). Structure-activity study: M. Sunagawa et al., J. Antibiot. 43, 519 (1990).

File:Meropenem synthesis.svg

SYN

https://patents.google.com/patent/WO2012062035A1/enCarbapenem, a type of β-lactam antibiotic, is known for its broad spectrum of antibacterial activity and strong antibacterial activity, such as meropenem (Me r0 p e nem), imine South (Imipenem) and Biabenem, etc., play an important role in the cure of severe infections.

Figure imgf000003_0001

Meropenem Imipenem For the synthetic methods of the Peinan type, the previous studies have mainly synthesized the corresponding Peinan side chain compound and the parent nucleus MAP, respectively, and then condensed and removed the protecting group to obtain the Peinan product. Such as US patentsUSP4933333, starting from 4-acetoxyazetidinone (4AA), obtained a matrix MAP after several steps of reaction. The mother nucleus is then condensed and deprotected from the side chain to obtain meropenem. However, this method is cumbersome, the synthesis step is long, and the total yield is low, and the noble metal catalyst is inevitably used in the synthesis of the compound (9).

Figure imgf000003_0002

MAP (10) Meropenem The Chinese invention patent document CN200810142137.5 has introduced a method for synthesizing meropenem.

Figure imgf000004_0001

 (XII) (I)(TBD S = Si (CH 3 ) 2 C (CH 3) 3; PNB = p-N0 2 -C 6 H 4 CH 2; PNZ = 2 -C 6 H 4 CH 2 OCO N0 p-) This method of Scheme Short, easy to operate, easy to get raw materials, but there are some areas for improvement.

Figure imgf000004_0002

Example 11) (3R, 4S)-3-[(R)-l-(tert-butyldimethylsilyloxy)ethyl]-4-[(2,S, 4’R)- 1- (allyl Synthesis of oxycarbonylxiaodimethylaminocarbonylpyrrolidinothio]-2-azetidinone (II) In a 500 ml reaction flask, add 22.6 g (0.075 mol) of (3S,4S)-3-[( R) l-(tert-Butyldimethylsilyloxy)ethyl]-4-[(R)-1-carbonylethyl]-2-azetidinone (IV), 17.1 g (0.083 mol) Dicyclohexylcarbodiimide (DCC) in 100 ml of acetone and 0.76 g of 4-dimethylaminopyridine (DMAP), 20.3 g (0.078 mol) of (2S, 4R)-2-dimethylamine was added dropwise with stirring. A solution of carbonyl-4-mercapto (i-propoxycarbonyl)pyrrolidine (V) in 125 ml of acetone was reacted at room temperature for 14 hours. Filtration, collecting the filtrate, concentrating, adding 200 ml of toluene thereto, using 200 ml of a 5 % acetic acid solution, 200 ml of a saturated sodium hydrogencarbonate solution and 150 ml of saturation Washed with brine, dried over anhydrous magnesium sulfate and evaporated to dryness <mjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjj 4-[(2,8, 4, ) small (propoxycarbonyl dimethyl dimethylaminocarbonyl)pyrrolidinyl]-2-azetidinone (II), directly without further treatment Invest in the next step.1H-NMR (400 MHz, CDC 13): </ RTI> <RTIgt; m), 2.816-2.849 (lH, s), 2.935-2.953 (3H, m), 3.027-079 (3H, d), 3.378-3.401 (lH, m), 3.792-3.796 (1H, d), 3.807- 3.953 (lH, m), 4.042-4.160 (3H, m), 4.492-4.570 (2H, m), 4.670-4.739 (lH, m), 5.164-5.295 (1H, m), 5.807-5.921 (lH, m ), 6.214(1H, s). Example 22) (31,48)-3-[(1 )-1-(tert-butyldimethylsilyloxy)ethyl]-4-[(2,8,4,1 )- 1- (allyl Synthesis of oxycarbonyl-1-dimethylaminocarbonylpyrrolidinothio]-1-(zincpropoxyl)-2-azetidinone (III) In a 1000 ml reaction flask, add 34.8 g (0.064) Mol) (3R, 4S)-3-[(R)-l-(tert-butyldimethylsilyloxy)ethyl]-4-[(2,S, 4,R)-1-(allyl Oxycarbonyl-1-pyrimidinylcarbonyl)pyrrolidinylthio]-2-azetidinone (11), 15.0 ml of triethylamine and 350 ml of toluene, control temperature below -10 °C, add 18.9 g (0.128 mol) p-nitrobenzyl chloroacetate (VI), heated to 0 ° C (-20 ° 5 ° C can be) reaction l ~ 3h. Then slowly add 250 ml of ice water and stir for 10 min. The layers were static and the organic phase was washed three times with saturated sodium bicarbonate solution, 200 ml each time. Dry over anhydrous magnesium sulfate, filtered, and evaporated to dryness to give white crystals, 4,7g (0.0622mol, yield 97.3%) (3R, 4S)-3-[(R) small (tert-butyldimethylsilyloxy)ethyl ]-4-[(2,S, 4,R)-1-(allyloxycarbonyldimethyldimethylaminocarbonyl)pyrrolidinylsulfur]sodium (sweetoxypropanoyl)-2-azetidinone (III), the product was directly put into the next step without further purification.Mp: 33-34 °C1H-NMR (300 MHz, CDC 13):0.819(9H, s), 1.167(3H, d), 1.188(4H, d), 1.693(5H, s), 1.850-1.926(1H, m), 2.631-2.700(1H, m), 2.941-2.960( 3H,d), 3.029-3.080(3H,d), 3.357-3.433(lH, m), 3.506-3.545(2H, m), 3.918-3.968(1H, m), 4.054-4.123 (2H, m), 4.270-4.291(lH, m), 4.391(lH,s), 4.518-4.568(2H, m), 4.588-4.779(3H, m), 5.178-5.416(3H, m), 5.861-5.982(2H,m ). Example 33) (5R,6S,8R,2’S, 4,S)-[(R)-1-(tert-butyldimethylsilyloxy)ethyl]-3-[4-(1-allyloxycarbonyl) -1- dimethylaminocarbonylpyrrolidinothio]-6-(1-allyloxycarbonylethoxy)-1-azabicyclo[3.2.0]-hept-2-en-7-one- Synthesis of 2-carboxylate (W) In a 500 ml reaction flask, 40; 7 g (0.0622 mol) of (3R, 4S)-3-[(R)-l-(tert-butyldimethylsilyloxy) was added. Ethyl]-4-[(2,S,4,R)-1-(indolyloxycarbonyl-1-dimethylaminocarbonyl)pyrrolidinylsulfate]small (sweetoxypropanoyl)-2-nitrogen Heterocyclic butanone (III) and 150 ml of toluene, 22 ml of trimethyl phosphite (furrowing lg of hydroquinone) were added under nitrogen. After reacting at 60 ° C for 16 hours, the solvent was evaporated under reduced pressure. It was recrystallized by adding 300 ml of ethyl acetate, and the solid was collected, and vacuum-dried at 40 ° C to obtain 32.8 g (0.0528 mol, yield: 85.0%) (5R, 6S, 8R, 2’S, 4,S)-[(R)- 1-(tert-Butyldimethylsilyloxy)ethyl]-3-[4-(1-allyloxycarbonyl-1-dimethylaminocarbonyl)pyrrolidinyl] -6-(1-ene Propoxycarbonyl ethoxy) small azabicyclo[3.2.0]-hept-2-en-7-one-2-carboxylate (oxime).1H-NMR (300 MHz, CDC 13):0.82(9H, s), 1.24(6H, d), 1.26(3H, s), 1.36(3H, s), 1.94(1H, m), 2.69(1 H, m), 2.97-3.11(6H, m ), 3.15-3.74(4H, m), 4.35(2H,m), 4.37-4.67(5H, m), 5.24-5.28(4H, m), 5.84(1H, m). Example 44) (5R, 6S, 8R, 2, S, 4’S)-[(R)小(hydroxy)ethyl]-3-[4-(1-allyloxycarbonylsuccinylcarbonyl)pyrrolidinyl Synthesis of thio]-6-(1-allyloxycarbonylethoxy)-1-azabicyclo[3.2.0]-hept-2-en-7-one-2-carboxylate (Vffl) at room temperature , in a 2000ml reaction flask, add 32.8g (0.0528mol) (5R,6S,8R,2’S,4,S)-[(R)-1-(tert-butyldimethylsilyloxy)ethyl] 3-[4-(1-allyloxycarbonyl-1-dimethylaminocarbonyl)pyrrolidinyl]-6-(1-indolyloxycarbonylethoxy)-1-azabicyclo[3.2.0 -Hept-2-ene-7-one-2-carboxylate (W), 27.4 ml of acetic acid, 41.3 g of fluorohydrogenamine and 1000 ml of dichloromethane, stirred at room temperature for 48 h. After completion of the reaction, 500 ml of a saturated aqueous solution of sodium hydrogencarbonate was added to the reaction mixture, and the mixture was stirred for 10 minutes, and the methylene chloride layer was separated and dried over anhydrous magnesium sulfate to give a white solid (26.2 g (0.0517 mol, yield 98.0). %) (5R, 6S, 8R, 2’S, 4’S)-[(R)小(hydroxy)ethyl]-3-[4-(1-allyloxycarbonylsuccinylcarbonyl)pyr Rhodium thio] -6-(l-allyloxycarbonylethoxy)-1-azabicyclo[3. 2. 0]-hept-2-en-7-one-2-carboxylate (ring The product was directly charged to the next step without further purification.1H-NMR (300 MHz, CDC 13):1.26(3H, s), 1.36(3H, s), 1.94(1H, m), 2.67(1H, m), 2.97-3.11(6H, m), 3.2-3.7(4H, m) ; 4.25(2H, m), 4.47-4.87 (5H, m), 5.15-5.50 (4H, m), 5.94 (2H, m). Example 55) (5R,6S,8R,2,S,4,S)-3-[4-dimethylaminocarbonyl)pyrrolidinyl]-6-(l-hydroxyethyl)-1-aza Synthesis of bicyclo[3.2.0]-hept-2-en-7-one-2-carboxylate (I) To the reaction flask, 26.2 g (0.0517 mol) (5R, 6S, 8R, 2’S, 4’S) was added. – [(R)-l-(hydroxy)ethyl]-3-[4-(1-allyloxycarbonyl-1-dimethylaminocarbonyl)pyrrolidinyl] -6-(1-allyloxy Carbonyl ethoxy)-1-azabicyclo[3. 2. 0]-hept-2-en-7-one-2-carboxylate (VDI), 21.3 g (0.152 mol) dimethylcyclohexane The ketone and 550 ml of ethyl acetate were heated to 30 ° C, and a solution of 1.0 g (0.865 mmol) of tetratriphenylphosphine palladium in 150 ml of dichloromethane was added dropwise thereto, and the mixture was reacted at room temperature for 3 h under nitrogen atmosphere. After adding 300 ml of water to the reaction mixture, the aqueous layer was separated, the aqueous layer was washed with ethyl acetate, and then, 500 ml of tetrahydrofuran was added dropwise with stirring in an ice bath, and the crystals were stirred, and the crystals were collected and dried in vacuo to give pale yellow crystals of 13.4 g (0.0352 md, Yield 68.1%) (5R,6S,8R,2,S,4,S)-3-[4-(2-dimethylaminocarbonyl)pyrrolidinylthio]-6-(1-hydroxyethyl) 1-Azabicyclo[3.2.0]-hept-2-en-7-one-2-carboxylic acid trihydrate (I)-Meropectin.IR max KBr cm- 1 : 1755, 1627, 1393, 1252, 1130NMR (D20, 300Hz): 1.25 (3H, d), 1.81-1.96 (1H, m), 2.96 (3H, s), 3.03 (3H, s), 3.14-3.20 (3H, m), 3.31-3.41 (2H, m), 3.62- 3.72 (1H, m), 3.90-4.00 (1H, m), 4.14-4.26 (2H, m), 4.63 (1H, t). Example 6 6) (5R,6S,8R,2’S,4’S)-3-[4-(2-Dimethylaminocarbonyl)pyrrolidinylthio]-6-(l-hydroxyethyl)-1-azabicyclo[ Synthesis of 3.2.0]-hept-2-en-7-one-2-carboxylate (I)21.3 g (0.152 mol) of dimethylcyclohexanedione in Example 5 was replaced with 45.1 g (0.155 mol) of tributyltin hydride, and 0.125 g (0.108 mmol) of tetrakistriphenylphosphine palladium was added dropwise, and the other amount was added. And the same method, the obtained 16.2g (0.0426mol, 82.5%) (5R,6S,8R,2’S,4’S)-3-[4-(2-dimethylaminocarbonyl)pyrrolidinyl Sulfur]-6-(l-hydroxyethyl)-1-azabicyclo[3.2.0]-hept-2-en-7-one-2-carboxylic acid trihydrate (1) ~ meropenem. Example 7 7) (5R,6S,8R,2,S,4,S)-3-[4-(2-dimethylaminocarbonyl)pyrrolidinyl]-6-(1-hydroxyethyl)-1- Synthesis of azabicyclo[3.2.0]-hept-2-en-7-one-2-carboxylate (I) To the reaction flask, 26.2 g (0.0517 mol) of (5R, 6S, 8R, 2, S, 4’S)-[(R)-l-(hydroxy)ethyl]-3-[4-(1-allyl was added) Oxycarbonyl-1-ylaminocarbonylcarbonylpyrrolidinothio]-6-(1-allyloxycarbonylethoxy)azaabicyclo[3. 2.]-hept-2-ene-7- Ketone-2-carboxylate 01), 6.0 g (0.0387 mol) of N, N-dimethylbarbituric acid and 500 ml of dichloromethane, and 6.0 g (5.2 mmol) of tetratriphenylphosphine was added dropwise thereto. A solution of palladium in 100 ml of dichloromethane was reacted at room temperature for 5 h under nitrogen. After adding 300 ml of water to the reaction mixture, the aqueous layer was separated, and the aqueous layer was washed with ethyl acetate. THF was evaporated and evaporated, and the crystals were evaporated, and crystals were collected, and the crystals were dried in vacuo to give 15.7 g (0.0413 mol, yield: 80.1%). 5R, 6S, 8R, 2,S,4,S) – 3-[4-(2-Dimethylaminocarbonyl)pyrrolidinylthio]-6-(1-hydroxyethyl)-1-azabicyclo [3. 2. 0] -Hept-2-ene-7-keto-2-carboxylic acid trihydrate (I)-Meropectin. 
ClaimsHide Dependent 

Rights requesta synthetic method of meropenem, characterized in that the specific reaction route of the synthetic method

Figure imgf000011_0001

 The reaction steps are as follows:1) The compound of the formula (IV) and the compound of the formula (V) are dissolved in an organic solvent and then subjected to a condensation reaction to obtain a compound of the formula (Π), the reaction time is 2 to 24 hours, and the reaction temperature is 0 to 40 ° C. ;2) The compound of the formula (Π) and the compound of the formula (VI) are dissolved in toluene, ethyl acetate or tetrahydrofuran and reacted with a base to form a compound of the formula (III), and the reaction time is ! ~ 3 hours, the reaction temperature is -20~5 °C;3) The compound of the formula (III) is dissolved in cyclohexanyl, n-glyoxime, n-octyl, toluene or xylene, and a Wittig ring-closing reaction is carried out under the action of an organophosphorus reagent to obtain a compound of the formula (VD), the organophosphorus reagent Is triphenylphosphine, tri-n-butylphosphine, triethyl phosphite or trimethyl phosphite;4) The compound of the formula (VII) is dissolved in methanol, tetrahydrofuran, acetone, n-pentane, n-hexane, diethyl ether, acetonitrile, dichloromethane, chloroform or ethyl acetate to hydrolyze the silyl ether bond under the action of an acid to obtain a formula (W). a compound; the acid is dilute hydrochloric acid, hydrofluoric acid, tetrabutylammonium fluoride, benzyltributylammonium fluoride, hydrofluoric hinge or vinegar The acid, the molar ratio of the acid to the compound of the formula (W) is 5 to 15: 1; the temperature of the hydrolysis reaction is 0 to 40 ° C, and the reaction time is 8 to 24 hours;5) a compound of the formula (W) dissolved in one or more of methanol, ethanol, tert-butanol, isobutanol, isopropanol, tetrahydrofuran, dioxanthene, acetone, dichloromethane, chloroform and water After the solvent is formed, the allylic group is hydrogenated by a palladium catalyst to obtain the target product (1). The molar ratio of the palladium catalyst to the compound of the formula 1) is 0.0001 to 0.5:1; the reaction temperature is 0 to 40 ° C. , the reaction time is 2~24h.2. A method for synthesizing meropenem according to claim 1, wherein the molar ratio of the compound of the formula (IV) to the compound of the formula (V) is 1.05 to 1.0: 1, the condensing agent and The molar ratio of the compound of the formula (IV) is 1.50 to 1.05:1.The method for synthesizing meropenem according to claim 1 or 2, wherein the condensing agent is a carbodiimide reagent or hydrazine, Ν’-carbonyldiimidazole; and the organic solvent is acetone. , acetonitrile, toluene, tetrahydrofuran, chloroform or dimethylformamide.The method for synthesizing meropenem according to claim 1, wherein the molar ratio of the compound of the formula (VI) to the compound of the formula (VI) is from 1.5 to 2.5:1, the base and the The molar ratio of the compound of the formula (VI) is from 1.2 to 2:1.The method for synthesizing meropenem according to claim 1, wherein the molar ratio of the organophosphorus reagent to the compound of formula (III) in step 3) is 2-8: 1; The reaction temperature is 25 to 100 £ ^, and the reaction time is 10 to 24 hours.The method for synthesizing meropenem according to claim 3, wherein the carbodiimide reagent is dicyclohexylcarbodiimide, diisopropylcarbodiimide or 1-( 3-dimethylaminopropyl)-3-ethylcarbodiimide.7. A method for synthesizing meropenem according to claim 1, wherein the base in step 2) is an inorganic base or an organic base; when it is an inorganic base, it is sodium hydroxide, sodium carbonate or Sodium bicarbonate; when it is an organic base, it is pyridine, triethylamine, diisopropylethylamine or 2,6-lutidine.The method for synthesizing meropenem according to claim 1, wherein the palladium catalyst is palladium acetate, palladium chloride, palladium nitrate, bistriphenylphosphine palladium chloride or tetrakistriphenylphosphine. palladium.9. A method for synthesizing meropenem according to claim 1, wherein the protecting group acceptor in step 5) is morpholine, dimethylcyclohexanedione, tributyltin hydride, N, N-dimethylbarbituric acid, -ethylhexanoic acid or hexanoic acid. 
 SYN 

Reference: Nadenik, Peter; Storm, Ole; Kremminger, Peter. Meropenem intermediate in crystalline form. WO 2005118586. (Assignee Sandoz AG, Switz)

SYN 2

Reference: Nishino, Keita; Koga, Teruyoshi. Improved process for producing carbapenem compound. WO 2007111328. (Assignee Kaneka Corporation, Japan)

SYN 3

Reference: Manca, Antonio; Monguzzi, Riccardo Ambrogio. Process for synthesizing carbapenem using Raney nickel. EP 2141167. (Assignee ACS Dobfar S.p.A., Italy)

SYN 4 

Reference: Tseng, Wei-Hong; Chang, Wen-Hsin; Chang, Chia-Mao; Yeh, Chia-Wei; Kuo, Yuan-Liang. Improved process for the preparation of carbapenem using carbapenem intermediates and recovery of carbapenem. EP 2388261. (Assignee Savior Lifetec Corp., Taiwan)

STR5 

Reference: Gnanaprakasam, Andrew; Ganapathy, Veeramani; Syed Ibrahim, Shahul Hameed; Karthikeyan, Murugesan; Sivasamy, Thangavel; Michael, Sekar Jeyaraj; Arulmoli, Thangavel; Das, Gautam Kumar. Preparation of meropenem trihydrate. WO 2012160576. (Assignee Sequent Anti Biotics Private Limited, India)

SYN 6 

Reference: Gnanprakasam, Andrew; Ganapathy, Veeramani; Syed Ibrahim, Shahul Hameed; Karthikeyan, Murugesan; Sivasamy, Thangavel; Sekar, Jeyaraj; Arulmoli, Thangavel. Preparation of meropenem trihydrate. IN 2011CH01780. (Assignee Sequent Scientific Limited, India)

SYN7 

Reference: Senthikumar, Udayampalayam Palanisamy; Sureshkumar, Kanagaraj; Babu, Kommoju Nagesh; Sudhan, Henry Syril; Kamaraj, Ponraj Pravin; Suresh, Thangaiyan. An improved process for the preparation of carbapenem antibiotic. WO 2013150550. (Assignee Orchid Chemicals & Pharmaceuticals Limited, India)

SYN 8 

Reference: Ong, Winston Zapanta; Nowak, Pawel Wojciech; Kim, Jinsoo; Enlow, Elizabeth M.; Bourassa, James; Cu, Yen; Popov, Alexey; Chen, Hongming. Meropenem derivatives and uses thereof. WO 2014144285. (Assignee Kala Pharmaceuticals, Inc., USA)

SYN9 

Reference: Cookson, James; McNair, Robert John; Satoskar, Deepak Vasant. Preparation of a carbapenem antibiotic by hydrogenation in the presence of a heterogeneous catalyst. WO 2015145161. (Assignee Johnson Matthey Public Limited Company, UK)

SYN 10 

Reference: Gruenewald, Elena; Weidlich, Stephan; Jantke, Ralf. Process for the deprotection of a carbapenem by heterogeneous catalytic hydrogenation with hydrogen in the presence of an organic amine. WO 2018010974. (Assignee Evonik Degussa GmbH, Germany)

SYN 11 

Some improvements in total synthesis of meropenem; Hu, Lai-Xing; Liu, Jun; Jin, Jie; Zhongguo Yiyao Gongye Zazhi; Volume 31; Issue 7; Pages 290-292; Journal; 2000 
synhttps://www.researchgate.net/figure/Synthesis-of-MRPD-starting-from-meropenem_fig9_283306781

Synthesis of MRPD starting from meropenem.

////////////////////////// 

wdt-16

join me on Linkedin

Anthony Melvin Crasto Ph.D – India | LinkedIn

join me on Researchgate

RESEARCHGATE

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

join me on Facebook

Anthony Melvin Crasto Dr. | Facebook

join me on twitter

Anthony Melvin Crasto Dr. | twitter

+919321316780 call whatsaapp

EMAIL. amcrasto@amcrasto

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

Meropenem is an ultra-broad spectrum injectable antibiotic used to treat a wide variety of infections, including meningitis and pneumonia. It is a beta-lactam and belongs to the subgroup of carbapenem, similar to imipenem and ertapenem. Meropenem was originally developed by Sumitomo Pharmaceuticals. It is marketed outside Japan by AstraZeneca with the brand names Merrem and Meronem. Other brand names include Zwipen (India, Marketed by Nucleus) Mepem (Taiwan) Meropen (Japan, Korea) and Neopenem (NEOMED India) . It gained FDA approval in July 1996. It penetrates well into many tissues and body fluids including the cerebrospinal fluid, bile, heart valves, lung, and peritoneal fluid.

Meropenem, sold under the brandname Merrem among others, is an intravenous β-lactam antibiotic used to treat a variety of bacterial infections.[1] Some of these include meningitisintra-abdominal infectionpneumoniasepsis, and anthrax.[1]

Common side effects include nausea, diarrhea, constipation, headache, rash, and pain at the site of injection.[1] Serious side effects include Clostridium difficile infectionseizures, and allergic reactions including anaphylaxis.[1] Those who are allergic to other β-lactam antibiotics are more likely to be allergic to meropenem as well.[1] Use in pregnancy appears to be safe.[1] It is in the carbapenem family of medications.[1] Meropenem usually results in bacterial death through blocking their ability to make a cell wall.[1] It is more resistant to breakdown by β-lactamase producing bacteria.[1]

Meropenem was patented in 1983.[2] It was approved for medical use in the United States in 1996.[1] It is on the World Health Organization’s List of Essential Medicines.[3] The World Health Organization classifies meropenem as critically important for human medicine.[4]

Medical uses

The spectrum of action includes many Gram-positive and Gram-negative bacteria (including Pseudomonas) and anaerobic bacteria. The overall spectrum is similar to that of imipenem, although meropenem is more active against Enterobacteriaceae and less active against Gram-positive bacteria. It works against extended-spectrum β-lactamases, but may be more susceptible to metallo-β-lactamases.[5] Meropenem is frequently given in the treatment of febrile neutropenia. This condition frequently occurs in patients with hematological malignancies and cancer patients receiving anticancer drugs that suppress bone marrow formation. It is approved for complicated skin and skin structure infections, complicated intra-abdominal infections and bacterial meningitis.

In 2017 the FDA granted approval for the combination of meropenem and vaborbactam to treat adults with complicated urinary tract infections.[6]

Administration

Meropenem is administered intravenously as a white crystalline powder to be dissolved in 5% monobasic potassium phosphate solution. Dosing must be adjusted for altered kidney function and for haemofiltration.[7]

As with other ß-lactams antibiotics, the effectiveness of treatment depends on the amount of time during the dosing interval that the meropenem concentration is above the minimum inhibitory concentration for the bacteria causing the infection.[8] For ß-lactams, including meropenem, prolonged intravenous administration is associated with lower mortality than bolus intravenous infusion in persons with whose infections are severe, or caused by bacteria that are less sensitive to meropenem, such as Pseudomonas aeruginosa.[8][9]

Side effects

The most common adverse effects are diarrhea (4.8%), nausea and vomiting (3.6%), injection-site inflammation (2.4%), headache (2.3%), rash (1.9%) and thrombophlebitis (0.9%).[10] Many of these adverse effects were observed in severely ill individuals already taking many medications including vancomycin.[11][12] Meropenem has a reduced potential for seizures in comparison with imipenem. Several cases of severe hypokalemia have been reported.[13][14] Meropenem, like other carbapenems, is a potent inducer of multidrug resistance in bacteria.

Pharmacology

Mechanism of action

Meropenem is bactericidal except against Listeria monocytogenes, where it is bacteriostatic. It inhibits bacterial cell wall synthesis like other β-lactam antibiotics. In contrast to other beta-lactams, it is highly resistant to degradation by β-lactamases or cephalosporinases. In general, resistance arises due to mutations in penicillin-binding proteins, production of metallo-β-lactamases, or resistance to diffusion across the bacterial outer membrane.[10] Unlike imipenem, it is stable to dehydropeptidase-1, so can be given without cilastatin.

In 2016, a synthetic peptide-conjugated PMO (PPMO) was found to inhibit the expression of New Delhi metallo-beta-lactamase, an enzyme that many drug-resistant bacteria use to destroy carbapenems.[15][16]

Society and culture

Meropenem vial

Trade names

CountryNameMaker
IndiaInzapenumDream India
  Aurobindo Pharma
 PenmerBiocon
 MeronirNirlife
 MerowinStrides Acrolab
 AktimerAktimas Biopharmaceuticals
 NeopenemNeomed
 MexopenSamarth life sciences
 MeropeniaSYZA Health Sciences LLP
 IvpenemMedicorp Pharmaceuticals
 Merofit 
 LykapiperLyka Labs
 WinmeroParabolic Drugs
Bangladesh
 MerojectEskayef Pharmaceuticals Ltd.
 MeroconBeacon Pharmaceuticals
IndonesiaMerofenKalbe
BrazilZylpenAspen Pharma
Japan, KoreaMeropen 
AustraliaMerem 
TaiwanMepem 
GermanyMeronem 
NigeriaZironemLyn-Edge Pharmaceuticals
USMeronemAstraZeneca
MerosanSanbe Farma
 MerobatInterbat
 Zwipen 
 Carbonem 
 RonemOpsonin Pharma, BD
 Neopenem 
 MeroconContinental
 CarnemLaderly Biotech
 PenroBosch
 MerozaGerman Remedies
 MerotrolLupin)
 MeromerOrchid Chemicals
 MepenoxBioChimico
 MeromaxEurofarma
 RopenMacter
 mirageadwic
 MeropexApex Pharma Ltd.
 MerostarkylHefny Pharma Group[17]

References

  1. Jump up to:a b c d e f g h i j “Meropenem”. The American Society of Health-System Pharmacists. Retrieved 8 December 2017.
  2. ^ Fischer, Janos; Ganellin, C. Robin (2006). Analogue-based Drug Discovery. John Wiley & Sons. p. 497. ISBN 9783527607495.
  3. ^ World Health Organization (2019). World Health Organization model list of essential medicines: 21st list 2019. Geneva: World Health Organization. hdl:10665/325771. WHO/MVP/EMP/IAU/2019.06. License: CC BY-NC-SA 3.0 IGO.
  4. ^ World Health Organization (2019). Critically important antimicrobials for human medicine (6th revision ed.). Geneva: World Health Organization. hdl:10665/312266ISBN 9789241515528.
  5. ^ AHFS Drug Information (2006 ed.). American Society of Health-System Pharmacists. 2006.
  6. ^ Commissioner, Office of the (24 March 2020). “Press Announcements – FDA approves new antibacterial drug”http://www.fda.gov.
  7. ^ Bilgrami, I; Roberts, JA; Wallis, SC; Thomas, J; Davis, J; Fowler, S; Goldrick, PB; Lipman, J (July 2010). “Meropenem dosing in critically ill patients with sepsis receiving high-volume continuous venovenous hemofiltration” (PDF). Antimicrobial Agents and Chemotherapy54 (7): 2974–8. doi:10.1128/AAC.01582-09PMC 2897321PMID 20479205.
  8. Jump up to:a b Yu Z, Pang X, Wu X, Shan C, Jiang S (2018). “Clinical outcomes of prolonged infusion (extended infusion or continuous infusion) versus intermittent bolus of meropenem in severe infection: A meta-analysis”PLOS ONE13 (7): e0201667. Bibcode:2018PLoSO..1301667Ydoi:10.1371/journal.pone.0201667PMC 6066326PMID 30059536.
  9. ^ Vardakas KZ, Voulgaris GL, Maliaros A, Samonis G, Falagas ME (January 2018). “Prolonged versus short-term intravenous infusion of antipseudomonal β-lactams for patients with sepsis: a systematic review and meta-analysis of randomised trials”. Lancet Infect Dis18 (1): 108–120. doi:10.1016/S1473-3099(17)30615-1PMID 29102324.
  10. Jump up to:a b Mosby’s Drug Consult 2006 (16 ed.). Mosby, Inc. 2006.
  11. ^ Erden, M; Gulcan, E; Bilen, A; Bilen, Y; Uyanik, A; Keles, M (7 March 2013). “Pancytopenýa and Sepsýs due to Meropenem: A Case Report” (PDF). Tropical Journal of Pharmaceutical Research12 (1). doi:10.4314/tjpr.v12i1.21.
  12. ^ “Meropenem side effects – from FDA reports”. eHealthMe.
  13. ^ Margolin, L (2004). “Impaired rehabilitation secondary to muscle weakness induced by meropenem”. Clinical Drug Investigation24(1): 61–2. doi:10.2165/00044011-200424010-00008PMID 17516692S2CID 44484294.
  14. ^ Bharti, R; Gombar, S; Khanna, AK (2010). “Meropenem in critical care – uncovering the truths behind weaning failure”Journal of Anaesthesiology Clinical Pharmacology26 (1): 99–101.
  15. ^ “New molecule knocks out superbugs’ immunity to antibiotics”newatlas.com. 20 January 2017. Retrieved 2017-01-25.
  16. ^ K., Sully, Erin; L., Geller, Bruce; Lixin, Li; M., Moody, Christina; M., Bailey, Stacey; L., Moore, Amy; Michael, Wong; Patrice, Nordmann; M., Daly, Seth (2016). “Peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) restores carbapenem susceptibility to NDM-1-positive pathogens in vitro and in vivo”Journal of Antimicrobial Chemotherapy72 (3): 782–790. doi:10.1093/jac/dkw476PMC 5890718PMID 27999041.
  17. ^ “Hefny Pharma Group”hefnypharmagroup.info. Retrieved 2018-05-22.

External links

  • “Meropenem”Drug Information Portal. U.S. National Library of Medicine.
Clinical data
Trade namesMerrem, others
AHFS/Drugs.comMonograph
Pregnancy
category
AU: B2
Routes of
administration
Intravenous
ATC codeJ01DH02 (WHO)
Legal status
Legal statusAU: S4 (Prescription only)UK: POM (Prescription only)US: ℞-only
Pharmacokinetic data
Bioavailability100%
Protein bindingApproximately 2%
Elimination half-life1 hour
ExcretionRenal
Identifiers
showIUPAC name
CAS Number119478-56-7 
PubChem CID441130
DrugBankDB00760 
ChemSpider389924 
UNIIFV9J3JU8B1
KEGGD02222 
ChEBICHEBI:43968 
ChEMBLChEMBL127 
PDB ligandMEM (PDBeRCSB PDB)
CompTox Dashboard (EPA)DTXSID7045526 
ECHA InfoCard100.169.299 
Chemical and physical data
FormulaC17H25N3O5S
Molar mass383.46 g·mol−1
3D model (JSmol)Interactive image
showSMILES
showInChI
  (verify)

Patent

Publication numberPriority datePublication dateAssigneeTitleUS4888344A *1986-07-301989-12-19Sumitomo Pharmaceuticals Company, LimitedCarbapenem compound in crystalline form, and its production and useCN101348486A *2008-08-292009-01-21深圳市海滨制药有限公司Preparation of meropenemCN101962383A *2010-11-122011-02-02上海巴迪生物医药科技有限公司Synthesis method of meropenemFamily To Family CitationsJPS6475488A *1987-09-171989-03-22Sumitomo PharmaProduction of beta-lactam compound* Cited by examiner, † Cited by third party

 

Publication numberPriority datePublication dateAssigneeTitleFamily To Family CitationsCN101962383A *2010-11-122011-02-02上海巴迪生物医药科技有限公司Synthesis method of meropenemCN102250096B *2011-09-052016-04-06江西华邦药业有限公司A kind of preparation method of meropenemCN104072523B *2014-07-142017-10-24上海上药新亚药业有限公司The preparation method of BiapenemCN108191869A *2018-01-222018-06-22重庆天地药业有限责任公司The purification process of Meropenem 
PublicationPublication DateTitleEP0007973B11984-02-01Process for the preparation of thienamycin and intermediatesUS4631150A1986-12-23Process for the preparation of penemsWO2012062035A12012-05-18Synthesis method for meropenemWO2010022590A12010-03-04Method for preparation of meropenemUS4443373A1984-04-17Process for the production of antibiotic penemsWO2008035153A22008-03-27Process for the preparation of beta-lactam antibioticEP0167154B11990-01-03Process for preparing 4-acetoxy-3-hydroxyethylazetizin-2-one derivativesKR101059339B12011-08-24Method for preparing carbapenem compound for oral administrationKR100886347B12009-03-03Process for stereoselective preparation of 4-BMA using a chiral auxiliaryUS4841043A1989-06-20Stereoselective synthesis of 1-β-alkyl carbapenem antibiotic intermediatesUS4772683A1988-09-20High percentage beta-yield synthesis of carbapenem intermediatesJP2000344774A2000-12-12Production of carbapenem compoundAU745980B22002-04-11Titanium catalyzed preparation of carbapenem intermediatesUS5700930A1997-12-234-substituted azetidinones as precursors to 2-substituted-3-carboxy carbapenem antibiotics and a method of producing themJP2002338572A2002-11-27Method for producing carbapenemsJP3684339B22005-08-17Method for producing carbapenem compoundsEP0066301B11986-01-22Intermediates for the preparation of thienamycin and process for preparing the sameWO2001053305A12001-07-26Processes for the preparation of carbapenem derivativesAU737502B22001-08-23Preparation of beta-methyl carbapenem intermediatesJP3213734B22001-10-02New β-lactam compoundsJP2004107289A2004-04-08Method for producing vinyl sulfide compoundJPH085853B21996-01-24Lactam compound and its manufacturing methodJPH0827168A1996-01-30Carbapenem intermediate fieldEP0204440A11986-12-10Azetidine derivatives productionWO1994021638A11994-09-29Process for the preparation of condensed carbapeneme derivatives

 

ApplicationPriority dateFiling dateTitleCN 2010105416652010-11-122010-11-12Synthesis method of meropenemCN201010541665.52010-11-12
Nmrhttps://www.researchgate.net/figure/1HNMR-spectra-of-meropenem-hydrolysis-catalyzed-by-NDM-1-Ecoli-cells-Only-1H-signals-of_fig3_272515470

1H NMR spectra of meropenem hydrolysis catalyzed by NDM-1 E. coli cells. Only 1H signals of methyl groups are shown. Signals from meropenem and the hydrolyzed product are colored in green and red, respectively.
NMR spectra monitoring meropenem hydrolysis catalyzed by NDM-1. a¹H NMR spectrum of hydrolyzed meropenem recorded before and 6 or 20 min after NDM-1 addition to the reaction system. b Part of a ROESY spectrum of the hydrolysis product. Diagonal and cross peaks are shown in blue and red, respectively. Proton signal assignments are labeled beside the peaks. The chemical shifts of H2, H1, H5, and H10 are highlighted by dashed lines

NMRNMR spectra monitoring meropenem hydrolysis catalyzed by NDM-1. a¹H NMR spectrum of hydrolyzed meropenem recorded before and 6 or 20 min after NDM-1 addition to the reaction system. b Part of a ROESY spectrum of the hydrolysis product. Diagonal and cross peaks are shown in blue and red, respectively. Proton signal assignments are labeled beside the peaks. The chemical shifts of H2, H1, H5, and H10 are highlighted by dashed linesSEEhttps://www.mdpi.com/1420-3049/23/11/2738/htm

Molecules 23 02738 g001 550

Figure 1. FT-IR spectra of unirradiated and irradiated (25 kGy) meropenem.

Molecules 23 02738 g002 550

Figure 2. Raman spectra of unirradiated and irradiated (A-25 kGy) meropenem.

Molecules 23 02738 g006 550

Figure 6. XRPD diffractograms of unirradiated and irradiated (25 kGy) meropenem.

Molecules 23 02738 g007 550

Figure 7. Differential scanning calorimetry (DSC) curves of non-irradiated and irradiated (A-25 kGy, B-400 kGy) meropenem. The arrows indicate the changes in the DSC spectrum after irradiation.

Molecules 23 02738 g009 550

Figure 9. FT-IR spectra of unirradiated and irradiated (400 kGy) meropenem. The arrows indicate the changes in the FT-IR spectrum after irradiation.

Molecules 23 02738 g010 550

Figure 10. Raman spectra of unirradiated and irradiated (400 kGy) meropenem. The arrow indicates the change in the Raman spectrum after irradiation.

//////////////MeropenemMerrem,  intravenous β-lactam antibiotic, bacterial infections,  meningitisintra-abdominal infectionpneumoniasepsis,  anthrax, Antibiotic SM 7338, ICI 194660, SM 7338, ANTIBACTERIALS

[H][C@]1([C@@H](C)O)C(=O)N2C(C(O)=O)=C(S[C@@H]3CN[C@@H](C3)C(=O)N(C)C)[C@H](C)[C@]12[H]

NEW DRUG APPROVALS

ONE TIME

$10.00