| FDA Orange Book Patents: 1 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9339551 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
Home » Articles posted by DR ANTHONY MELVIN CRASTO Ph.D (Page 86)
Author Archives: DR ANTHONY MELVIN CRASTO Ph.D
Ifetroban イフェトロバン
![]()
Ifetroban イフェトロバン
3-[2-({(1S,2R,3S)-3-[4-(Pentylcarbamoyl)-1,3-oxazol-2-yl]-7-oxabicyclo[2.2.1]hept-2-yl}methyl)phenyl]propanoic acid
- Molecular FormulaC25H32N2O5
- Average mass440.532 Da
- 143443-90-7;
Ifetroban is a potent and selective thromboxane receptor antagonist.[1]
Ifetroban has been used in trials studying the treatment of Skin Diseases, Autoimmune Diseases, Pathologic Processes, Scleroderma, Limited, and Scleroderma, Diffuse, among others.
This compound belongs to the class of organic compounds known as phenylpropanoic acids. These are compounds with a structure containing a benzene ring conjugated to a propanoic acid.
- OriginatorBristol-Myers Squibb
- DeveloperBristol-Myers Squibb; Cumberland Pharmaceuticals; Vanderbilt-Ingram Cancer Center
- ClassAntiasthmatics; Antihypertensives; Antiplatelets; Heterocyclic bicyclo compounds; Oxazoles; Small molecules
- Mechanism of ActionThromboxane A2 receptor antagonists
- Phase IIAsthma; Hepatorenal syndrome; Portal hypertension; Solid tumours; Systemic scleroderma
- DiscontinuedCoronary thrombosis; Peripheral vascular disorders; Thrombosis
- 12 Dec 2018Phase-II clinical trials in Solid tumours (Metastatic disease, Late-stage disease, Second-line therapy or greater, Recurrent) in USA (PO) (NCT03694249)
- 13 Nov 2018Efficacy and adverse events data from a phase II trial in Portal hypertension released by Cumberland Pharmaceuticals
- 03 Oct 2018Vanderbilt-Ingram Cancer Center and Cumberland Pharmaceuticals plans a phase II trial for Solid tumours (Metastatic disease, Late-stage disease, Second-line therapy or greater, Recurrent) (PO, capsule) (NCT03694249)
Ifetroban sodium
- Molecular FormulaC25H31N2NaO5
- Average mass462.514 Da
- Monoisotopic mass462.213074 Da

SYN
BMS-180291 sodium salt was prepared from optically active 7-oxabicyclo[2.2.1]heptane lactol (I): The interphenylene side chain was introduced by deprotonation of (I) with ethylmagnesium bromide (0.95 eq.) followed by treatment with excess aryl Grignard (II) to afford crystalline diol (III). The extraneous benzylic hydroxyl group in (III) was removed by reduction with hydrogen in the presence of Pearlman’s catalyst to give alcohol (IV). Transformation of the alpha-side chain silyloxy carbinol of (IV) to a carboxymethyl ester was accomplished by initial protection of the omega-side chain alcohol as the acetate (Ac2O/py) followed by oxidation under Jones conditions and then exposure of the resulting crude acetate-acid to methanolic hydrogen chloride to afford crystalline alcohol-ester (V). Oxidation of (V) under Jones conditions furnished acid-ester (VI). The oxazole side chain was introduced into (VI) via serine-derived amino alcohol (VII). Standard coupling of acid (VI) with (VII) mediated by water-soluble carbodiimide (EDAC) gave amide (VIII). Acyclic side chain intermediate (VIII) was converted into oxazole (X) in three steps by mesylation followed by treatment with triethylamine to furnish cyclized oxazoline (IX). Dehydrogenation of (IX) employing a novel oxidative protocol (1) involving treatment with a mixture of copper (II) bromide and 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU) in chloroform/ethyl acetate solvent yielded oxazole (X). Saponification of (X) followed by acidification afforded (BMS-180291) as a white solid which could be purified by recrystallization from acetonitrile. The water-soluble sodium salt (XI) was available as a precipitate from BMS-18091 by treatment with sodium methoxide/methanol in acetone.

SYN
The interphenylene side chain was introduced by deprotonation of (I) with ethylmagnesium bromide (0.95 eq.) followed by treatment with excess aryl Grignard (II) to afford crystalline diol (III). The extraneous benzylic hydroxyl group in (III) was removed by reduction with hydrogen in the presence of Pearlman’s catalyst to give alcohol (IV). Transformation of the alpha-side chain silyloxy carbinol to a carboxy methyl ester was accomplished by initial protection of the omega-side chain alcohol as the acetate (Ac2O/pyr) followed by oxidation under Jones conditions and then exposure of the resulting crude acetate-acid to methanolic hydrogen chloride to afford crystalline alcohol-ester (V). Oxidation of (V) under Jones conditions furnished acid-ester (VI). The oxazole side chain was introduced into (VI) via serine-derived amino alcohol (VII). Standard coupling of acid (VI) with (VII) mediated by water-soluble carbodiimide (EDAC) gave amide (VIII). Acyclic side chain intermediate (VIII) was converted into oxazole (X) in three steps by mesylation followed by treatment with triethylamine to furnish cyclized oxazoline (IX). Dehydrogenation of (IX) employing a novel oxidative protocol involving treatment with a mixture of copper (II) bromide and 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU) in chloroform/ethyl acetate solvent yielded oxazole (X). Saponification of (X) followed by acidification afforded (XI) (BMS-180291) as a white solid which could be purified by recrystallization from acetonitrile. The water-soluble sodium salt was available as a precipitate from (XI) by treatment with sodium methoxide/methanol in acetone.

SYN
| Org Process Res Dev 1997,1(1),14 |
The synthesis of [1S-(1alpha,2alpha,3alpha,4alpha)]-2-[2-[2-(methoxycarbonyl)ethyl]benzyl]-7-oxabicyclo[2.2.1]heptane-3-carboxylic acid (VI), a key intermediate in the synthesis of 203961 [see scheme 20396101a] has been presented: This compound has been obtained by two similar ways: 1) The condensation of L-valinol (XII) with anhydride (XXII) catalyzed by oxalic acid gives imide (XIII), which is treated with ethylmagnesium chloride, the Grignard reagent (XIV) and NaBH4 yielding intermediate (XV). This intermediate, without isolation, is treated with HCl in THF to afford the substituted benzaldehyde (XVI), which is condensed with trimethyl phosphonoacetate (XVII) and DBU in acetonitrile giving the propenoic ester (XVIII). Finally, this compound is submitted to a simultaneous reduction and hydrogenolysis with H2 over a Pearlman catalyst in methanol to provide the target of [1S-(1alpha,2alpha,3alpha,4alpha)]-2-[2-[2-(methoxycarbonyl)ethyl]benzyl]-7-oxabicyclo[2.2.1]heptane-3-carboxylic acid (VI). 2) The preceding reaction sequence can also be performed using (S)-2-phenylglycinol (XIX) instead of the L-valinol (XII) yielding the previously reported benzaldehyde (XVI) through the imide (XX) and the nonisolated intermediate (XXI).
References
- ^ Dockens, RC; Santone, KS; Mitroka, JG; Morrison, RA; Jemal, M; Greene, DS; Barbhaiya, RH (August 2000). “Disposition of Radiolabeled Ifetroban in Rats, Dogs, Monkeys, and Humans”(PDF). Drug Metabolism and Disposition. 28 (8): 973–80. PMID 10901709. Retrieved 5 October 2016.
| Identifiers | |
|---|---|
| CAS Number | |
| PubChem CID | |
| ChemSpider | |
| UNII | |
| KEGG | |
| ChEMBL | |
| Chemical and physical data | |
| Formula | C25H32N2O5 |
| Molar mass | 440.53 g/mol |
| 3D model (JSmol) | |
////////Ifetroban, BMS 18029, BMS 180291, BMS 180291A, BMS-180291-02, Boxaban, CPI 211, Hepatoren, Portaban, Vasculan, イフェトロバン
CCCCCNC(=O)C1=COC(=N1)C2C3CCC(C2CC4=CC=CC=C4CCC(=O)O)O3
Aceclofenac, ацеклофенак , أسيكلوفيناك , 醋氯芬酸 , アセクロフェナク
Aceclofenac
アセクロフェナク
- Molecular FormulaC16H13Cl2NO4
- Average mass354.185 Da



Characterization of Aceclofenac by 1H NMR spectroscopy
1H NMR (400 MHz, DMSO-d6) δ (ppm) 3.896 (s, 2H, Aliphatic –CH2), 4.634 (s, 2H, Aliphatic –CH2), 6.279 (d J= 8.00HZ, 1H, Aromatic), 6.887 (t, J = 7.2 Hz, 1H), 6.936 (s, 1H, NH), 7.039(t, J = 7.6 Hz, 1H, Aromatic), 7.225 (t J= 8.00 HZ, 1H, Aromatic), 7.260 (d J= 8.00 HZ, 1H, Aromatic), 7.537 (d J= 8.4HZ, 2H, Aromatic), 13.076 (s, 1H, Carboxylic acid) …https://www.sciencedirect.com/science/article/pii/S2214180417301290




https://www.dea.gov/sites/default/files/pr/microgram-journals/2014/mj11-1_29-41.pdf
Aceclofenac is a nonsteroidal anti-inflammatory drug (NSAID) analog of diclofenac. It is used for the relief of pain and inflammation in rheumatoid arthritis, osteoarthritis and ankylosing spondylitis.
Aceclofenac (C16H13Cl2NO4), chemically [(2-{2, 6-dichlorophenyl) amino} phenylacetooxyacetic acid], is a crystalline powder with a molecular weight of 354.19. It is practically insoluble in water with good permeability. It is metabolized in human hepatocytes and human microsomes to form [2-(2′,6′-dichloro-4′-hydroxy- phenylamino) phenyl] acetoxyacetic acid as the major metabolite, which is then further conjugated. According to the Biopharmaceutical Classification System (BCS) drug substances are classified to four classes upon their solubility and permeability. Aceclofenac falls under the BCS Class II, poorly soluble and highly permeable drug.[1]
Aceclofenac works by inhibiting the action of cyclooxygenase (COX) that is involved in the production of prostaglandins (PG) which is accountable for pain, swelling, inflammation and fever. The incidence of gastric ulcerogenicity of aceclofenac has been reported to be significantly lower than that of the other frequently prescribed NSAIDs, for instance, 2-folds lesser than naproxen, 4-folds lesser than diclofenac, and 7-folds lesser than indomethacin.
Aceclofenac should not be given to people with porphyria or breast-feeding mothers, and is not recommended for children. It should be avoided near term in a pregnant woman because of the risk of having a patent ductus arteriosus in the neonate.
![]()
SYN
Manufacturing Process for Aceclofenac
Stage-1
T Butanol and Chloro Acetyl Chloride react in presence of NN Dimethyl Aniline at low temperature. After reaction
organics mass wash with water and sodium bicarbonate solution to get stage-1
Stage-2
Stage-I react with Diclofenac Sodium in presence of TBAB in Toluene media, further react with formic acid and
reaction mass quenching in water and product is isolated by filtration. Finally Crude Aceclofenac purified in ethyl
acetate and charcoal. Pure product isolated by filtration.


SYN’
EP 0119932; US 4548952
Alkylation of the sodium salt of diclofenac (I) with benzyl bromoacetate (II) in hot DMF yielded the (arylacetoxy)acetate (III). Subsequent hydrogenolysis of the benzyl ester of (III) in the presence of Pd/C gave the title carboxylic acid. Alternatively, the benzyl ester group of (III) was cleaved by means of the combination of chlorotrimethylsilane and sodium iodide. This method of selective ester hydrolysis with in situ generated iodotrimethylsilane was also applied to the corresponding methyl (IV) and tert-butyl (V) esters. In a related procedure, tert-butyl ester (V) was prepared by alkylation of diclofenac (VI) with tert-butyl bromoacetate (VII) in the presence of tertiary amines. Selective cleavage of the tert-butyl ester group of (V) was then performed by treatment with either trifluoroacetic or formic acid.

SYN
| ES 2046141 |
Aceclofenac was prepared by selective hydrolysis of other labile ester precursors. Alkylation of diclofenac sodium (I) with tetrahydropyranyl chloroacetate (IX), prepared by protection of chloroacetic acid (VIII) with dihydropyran, furnished the tetrahydropyranyl ester of aceclofenac (X), which was then deprotected by treatment with HCl. Similarly, the preparation of aceclofenac was reported by acidic hydrolysis of the analogous tetrahydrofuranyl ester (XI).

References
- ^ Karmoker, J.R.; Sarkar, S.; Joydhar, P.; Chowdhury, S.F. (2016). “Comparative in vitro equivalence evaluation of some Aceclofenac generic tablets marketed in Bangladesh” (PDF). The Pharma Innovation Journal. 5: 3–7. Retrieved 2016-09-01.
- Sources
- British National Formulary 55, March 2008; ISBN 978-0-85369-776-3 p. 537
References
-
- EP 119 932 (Prodes; appl. 19.3.1984; E-prior. 21.3.1983).
- US 4 548 952 (Prodes; 22.10.1985; appl. 15.3.1984; E-prior. 21.3.1983).
-
Alternative synthesis:
- ES 2 020 146 (Prodesfarma; appl. 29.5.1990).
-
- ATC:M01AB16
- Use:non-steroidal anti-inflammatory, analgesic, non-selective cyclooxigenase inhibitor
- Chemical name:2-[(2,6-dichlorophenyl)amino]benzeneacetic acid carboxymethyl ester
- Formula:C16H13Cl2NO4
- MW:354.19 g/mol
- CAS-RN:89796-99-6
- InChI Key:MNIPYSSQXLZQLJ-UHFFFAOYSA-N
- InChI:InChI=1S/C16H13Cl2NO4/c17-11-5-3-6-12(18)16(11)19-13-7-2-1-4-10(13)8-15(22)23-9-14(20)21/h1-7,19H,8-9H2,(H,20,21)
- LD50:121 mg/kg (M, p.o.)
| Clinical data | |
|---|---|
| Trade names | Hifenac, Cincofen, Zerodol, Nacsiv, Acenac, others |
| AHFS/Drugs.com | International Drug Names |
| Routes of administration |
oral, topical |
| ATC code | |
| Legal status | |
| Legal status |
|
| Identifiers | |
| CAS Number | |
| PubChem CID | |
| ChemSpider | |
| UNII | |
| KEGG | |
| ChEBI | |
| ChEMBL | |
| ECHA InfoCard | 100.169.686 |
| Chemical and physical data | |
| Formula | C16H13Cl2NO4 |
| Molar mass | 353.02161 g/mol |
| 3D model (JSmol) | |
//////////Aceclofenac, ацеклофенак , أسيكلوفيناك , 醋氯芬酸 , アセクロフェナク
Diclofenac Sodium
Diclofenac Sodium
15307-79-6; Sodium diclofenac; Diclofenac sodium salt; Voltaren; Solaraze
| Molecular Formula: | C14H10Cl2NNaO2 |
|---|---|
| Molecular Weight: | 318.129 g/mol |
Diclofenac, sold under the trade names Voltaren among others, is a nonsteroidal anti-inflammatory drug (NSAID) used to treat pain and inflammatory diseases such as gout.[3] It is taken by mouth or applied to the skin.[3] Improvements in pain typically occur within half an hour and last for as much as eight hours.[3] It is also available in combination with misoprostol in an effort to decrease stomach problems.[4]
Common side effects include abdominal pain, gastrointestinal bleeding, nausea, dizziness, headache, and swelling.[3] Serious side effects may include heart disease, stroke, kidney problems, and stomach ulceration.[4][3] Use is not recommended in the third trimester of pregnancy.[3] It is likely safe during breastfeeding.[4] It is believed to work by decreasing the production of prostaglandin.[5] It blocks both cycloxygenase-1 (COX-1) and cycloxygenase-2 (COX-2).[3]
Diclofenac was patented in 1965 by Ciba-Geigy and came into medical use in the United States in 1988.[3][6] It is available as a generic medication.[3] In the United States the wholesale cost per dose is less than US$0.15 as of 2018.[7] In 2016 it was the 78th most prescribed medication in the United States with more than 9 million prescriptions.[8] It is available as both a sodium and a potassium salt.[4]
Medical uses
Diclofenac is used to treat pain, inflammatory disorders, and dysmenorrhea.[9]
Pain
Inflammatory disorders may include musculoskeletal complaints, especially arthritis, rheumatoid arthritis, polymyositis, dermatomyositis, osteoarthritis, dental pain, temporomandibular joint (TMJ) pain, spondylarthritis, ankylosing spondylitis, gout attacks,[10] and pain management in cases of kidney stones and gallstones. An additional indication is the treatment of acute migraines.[11] Diclofenac is used commonly to treat mild to moderate postoperative or post-traumatic pain, in particular when inflammation is also present,[10] and is effective against menstrual pain and endometriosis.
Diclofenac is also available in topical forms and has been found to be useful for osteoarthritis but not other types of long-term musculoskeletal pain.[12]
It may also help with actinic keratosis, and acute pain caused by minor strains, sprains, and contusions (bruises).[13]
In many countries,[14] eye drops are sold to treat acute and chronic nonbacterial inflammation of the anterior part of the eyes (e.g., postoperative states). Diclofenac eye drops have also been used to manage pain for traumatic corneal abrasion.[15]
Diclofenac is often used to treat chronic pain associated with cancer, in particular if inflammation is also present (Step I of the World Health Organization (WHO) scheme for treatment of chronic pain).[16] Diclofenac can be combined with opioids if needed such as a fixed combination of diclofenac and codeine.
-
Voltaren (diclofenac) 50 mg enteric coatedtablets
-
Arthrotec (diclofenac and misoprostol) 50 mg tablets
-
Sintofarm (diclofenac) for suppositoryadministration
Contraindications
- Hypersensitivity against diclofenac
- History of allergic reactions (bronchospasm, shock, rhinitis, urticaria) following the use of other NSAIDs such as aspirin
- Third-trimester pregnancy
- Active stomach and/or duodenal ulceration or gastrointestinal bleeding
- Inflammatory bowel disease such as Crohn’s disease or ulcerative colitis
- Severe insufficiency of the heart (NYHA III/IV)
- Pain management in the setting of coronary artery bypass graft (CABG) surgery
- Severe liver insufficiency (Child-Pugh Class C)
- Severe renal insufficiency (creatinine clearance <30 ml/min)
- Caution in patients with pre-existing hepatic porphyria, as diclofenac may trigger attacks
- Caution in patients with severe, active bleeding such as cerebral hemorrhage
- NSAIDs in general should be avoided during dengue fever, as it induces (often severe) capillary leakage and subsequent heart failure.
- Caution in patients with fluid retention or heart failure
- Can lead to onset of new hypertension or worsening of pre-existing hypertension
- Can cause serious skin adverse events such as exfoliative dermatitis, Stevens-Johnson Syndrome (SJS), and toxic epidermal necrolysis (TEN), which can be fatal[17]
Adverse effects
Diclofenac consumption has been associated with significantly increased vascular and coronary risk in a study including coxib, diclofenac, ibuprofen and naproxen.[18] Upper gastrointestinal complications were also reported.[18] Major adverse cardiovascular events (MACE) were increased by about a third by diclofenac, chiefly due to an increase in major coronary events.[18] Compared with placebo, of 1000 patients allocated to diclofenac for a year, three more had major vascular events, one of which was fatal.[18] Vascular death was increased significantly by diclofenac.[18]
Heart
In 2013, a study found major vascular events were increased by about a third by diclofenac, chiefly due to an increase in major coronary events.[18] Compared with placebo, of 1000 people allocated to diclofenac for a year, three more had major vascular events, one of which was fatal.[18] Vascular death was increased by diclofenac (1·65).[18]
Following the identification of increased risks of heart attacks with the selective COX-2 inhibitor rofecoxib in 2004, attention has focused on all the other members of the NSAIDs group, including diclofenac. Research results are mixed, with a meta-analysis of papers and reports up to April 2006 suggesting a relative increased rate of heart disease of 1.63 compared to nonusers.[19] Professor Peter Weissberg, Medical Director of the British Heart Foundation said, “However, the increased risk is small, and many patients with chronic debilitating pain may well feel that this small risk is worth taking to relieve their symptoms”. Only aspirin was found not to increase the risk of heart disease; however, this is known to have a higher rate of gastric ulceration than diclofenac. In Britain the Medicines and Healthcare Products Regulatory Agency (MHRA) said in June 2013 that the drug should not be used by people with serious underlying heart conditions—people who had suffered heart failure, heart disease or a stroke were advised to stop using it completely.[20] As of January 15, 2015 the MHRA announced that diclofenac will be reclassified as a prescription-only medicine (POM) due to the risk of cardiovascular adverse events.[21]
A subsequent large study of 74,838 Danish users of NSAIDs or coxibs found no additional cardiovascular risk from diclofenac use.[22] A very large study of 1,028,437 Danish users of various NSAIDs or coxibs found the “Use of the nonselective NSAID diclofenac and the selective cyclooxygenase-2 inhibitor rofecoxib was associated with an increased risk of cardiovascular death (odds ratio, 1.91; 95% confidence interval, 1.62 to 2.42; and odds ratio, 1.66; 95% confidence interval, 1.06 to 2.59, respectively), with a dose-dependent increase in risk.”[23]
Diclofenac is similar in COX-2 selectivity to celecoxib.[24]
Gastrointestinal
- Gastrointestinal complaints are most often noted. The development of ulceration and/or bleeding requires immediate termination of treatment with diclofenac. Most patients receive a gastro-protective drug as prophylaxis during long-term treatment (misoprostol, ranitidine 150 mg at bedtime or omeprazole 20 mg at bedtime).
Liver
- Liver damage occurs infrequently, and is usually reversible. Hepatitis may occur rarely without any warning symptoms and may be fatal. Patients with osteoarthritis more often develop symptomatic liver disease than patients with rheumatoid arthritis. Liver function should be monitored regularly during long-term treatment. If used for the short-term treatment of pain or fever, diclofenac has not been found more hepatotoxic than other NSAIDs.
- As of December 2009, Endo, Novartis, and the US FDA notified healthcare professionals to add new warnings and precautions about the potential for elevation in liver function tests during treatment with all products containing diclofenac sodium.[25]
- Cases of drug-induced hepatotoxicity have been reported in the first month, but can occur at any time during treatment with diclofenac. Postmarketing surveillance has reported cases of severe hepatic reactions, including liver necrosis, jaundice, fulminant hepatitis with and without jaundice, and liver failure. Some of these reported cases resulted in fatalities or liver transplantation.
- Physicians should measure transaminases periodically in patients receiving long-term therapy with diclofenac. Based on clinical trial data and postmarketing experiences, transaminases should be monitored within 4 to 8 week after initiating treatment with diclofenac.
Kidney
- NSAIDs “are associated with adverse renal [kidney] effects caused by the reduction in synthesis of renal prostaglandins“[26] in sensitive persons or animal species, and potentially during long-term use in nonsensitive persons if resistance to side effects decreases with age. However, this side effect cannot be avoided merely by using a COX-2 selective inhibitor because, “Both isoforms of COX, COX-1 and COX-2, are expressed in the kidney… Consequently, the same precautions regarding renal risk that are followed for nonselective NSAIDs should be used when selective COX-2 inhibitors are administered.”[26] However, diclofenac appears to have a different mechanism of renal toxicity.[citation needed]
- Studies in Pakistan showed diclofenac caused acute kidney failure in vultures when they ate the carcasses of animals that had recently been treated with it. Drug-sensitive species and individual humans are initially assumed to lack genes expressing specific drug detoxification enzymes.[27]
Mental health
- Mental health side effects have been reported. These symptoms are rare, but exist in significant enough numbers to include as potential side effects. These include depression, anxiety, irritability, nightmares, and psychotic reactions.[28]
Mechanism of action
The primary mechanism responsible for its anti-inflammatory, antipyretic, and analgesic action is thought to be inhibition of prostaglandin synthesis by inhibition of the transiently expressed prostaglandin-endoperoxide synthase-2 (PGES-2) also known as cycloxygenase-2 (COX-2). It also appears to exhibit bacteriostatic activity by inhibiting bacterial DNA synthesis.[29]
Inhibition of prostaglandin synthesis occurs systemically resulting in undesirable symptoms such as irritation of the gastric epithelium.[citation needed] This is the main side effect of diclofenac. Diclofenac inhibits COX-2 with 20 times greater potency than the constitutively expressed isoenzyme COX-1[30] and has, therefore, a somewhat lower incidence of gastrointestinal complaints than noted with aspirin which inhibits COX-1 to a greater extent.
The action of one single dose is much longer (6 to 8 hr) than the very short 1.2–2 hr half-life of the drug would indicate. This could be partly because it persists for over 11 hours in synovial fluids.[31]
Diclofenac may also be a unique member of the NSAIDs. Some evidence indicates it inhibits the lipoxygenase pathways, thus reducing formation of the leukotrienes(also pro-inflammatory autacoids). It also may inhibit phospholipase A2 as part of its mechanism of action. These additional actions may explain its high potency – it is the most potent NSAID on a broad basis.[32]
Marked differences exist among NSAIDs in their selective inhibition of the two subtypes of cyclooxygenase, COX-1 and COX-2. Much pharmaceutical drug design has attempted to focus on selective COX-2 inhibition as a way to minimize the gastrointestinal side effects of NSAIDs such as aspirin. In practice, use of some COX-2 inhibitors with their adverse effects has led to massive numbers of patient family lawsuits alleging wrongful death by heart attack, yet other significantly COX-selective NSAIDs, such as diclofenac, have been well tolerated by most of the population.\
Besides the COX-inhibition, a number of other molecular targets of diclofenac possibly contributing to its pain-relieving actions have recently been identified. These include:
- Blockage of voltage-dependent sodium channels (after activation of the channel, diclofenac inhibits its reactivation also known as phase inhibition)[citation needed]
- Blockage of acid-sensing ion channels (ASICs)[33]
- Positive allosteric modulation of KCNQ- and BK-potassium channels (diclofenac opens these channels, leading to hyperpolarization of the cell membrane)
Ecological effects
Use of diclofenac for animals is controversial due to toxicity when eaten by scavenging birds that eat dead animals; the drug has been banned for veterinary use in many countries.
Use of diclofenac in animals has been reported to have led to a sharp decline in the vulture population in the Indian subcontinent – a 95% decline by 2003[34] and a 99.9% decline by 2008. The mechanism is presumed to be renal failure;[35] however, toxicity may be due to direct inhibition of uric acid secretion in vultures.[36] Vultures eat the carcasses of livestockthat have been administered veterinary diclofenac, and are poisoned by the accumulated chemical,[37] as vultures do not have a particular enzyme to break down diclofenac. At a meeting of the National Wildlife Board in March 2005, the Government of India announced it intended to phase out the veterinary use of diclofenac.[38] Meloxicam is a safer alternative to replace use of diclofenac.[39] It is more expensive than diclofenac, but the price is coming down as more pharmaceutical companies begin to manufacture it.
Steppe eagles have the same vulnerability to diclofenac as vultures and may also fall victim to it.[40] Diclofenac has been shown also to harm freshwater fish species such as rainbow trout.[41][42][43][44] In contrast, New World vultures, such as the turkey vulture, can tolerate at least 100 times the level of diclofenac that is lethal to Gyps species.[45]
“The loss of tens of millions of vultures over the last decade has had major ecological consequences across the Indian Subcontinent that pose a potential threat to human health. In many places, populations of feral dogs (Canis familiaris) have increased sharply from the disappearance of Gyps vultures as the main scavenger of wild and domestic ungulatecarcasses. Associated with the rise in dog numbers is an increased risk of rabies“[39] and casualties of almost 50,000 people.[46] The Government of India cites this as one of the major consequences of a vulture species extinction.[38] A major shift in the transfer of corpse pathogens from vultures to feral dogs and rats could lead to a disease pandemic, causing millions of deaths in a crowded country like India, whereas vultures’ digestive systems safely destroy many species of such pathogens. Vultures are long-lived and slow to breed. They start breeding only at the age of six and only 50% of young survive. Even if the government ban is fully implemented, it will take several years to revive the vulture population.[47]
The loss of vultures has had a social impact on the Indian Zoroastrian Parsi community, who traditionally use vultures to dispose of human corpses in Towers of Silence, but are now compelled to seek alternative methods of disposal.[39]
Despite the vulture crisis, diclofenac remains available in other countries including many in Europe.[48] It was controversially approved for veterinary use in Spain in 2013 and continues to be available, despite Spain being home to around 90% of the European vulture population and an independent simulation showing that the drug could reduce the population of vultures by 1-8% annually. Spain’s medicine agency presented simulations suggesting that the number of deaths would be quite small.[49][50]
Formulations and trade names
The name “diclofenac” derives from its chemical name: 2-(2,6-dichloranilino) phenylacetic acid. Diclofenac was first synthesized by Alfred Sallmann and Rudolf Pfister and introduced as Voltaren by Ciba-Geigy (now Novartis) in 1973, now by Glaxo SmithKline.[51]
In the United Kingdom, United States, India, and Brazil diclofenac may be supplied as either the sodium or potassium salt; in China, it is most often supplied as the sodium salt, while in some other countries it is only available as the potassium salt.
Pennsaid is a minimally systemic prescription topical lotion formulation of 1.5% w/w diclofenac sodium, which is approved in the US, Canada and other countries for osteoarthritis of the knee.
Flector Patch, a minimally systemic topical patch formulation of diclofenac, is indicated for acute pain due to minor sprains, strains, and contusions. The patch has been approved in many other countries outside the US under different brand names.
Voltaren and Voltarol contain the sodium salt of diclofenac. In the United Kingdom, Voltarol can be supplied with either the sodium salt or the potassium salt, while Cataflam, sold in some other countries, is the potassium salt only. However, Voltarol Emulgel contains diclofenac diethylammonium, in which a 1.16% concentration is equivalent to a 1% concentration of the sodium salt. In 2016 Voltarol was one of the biggest selling branded over-the-counter medications sold in Great Britain, with sales of £39.3 million.[52]
Diclofenac is available in stomach acid-resistant formulations (25 and 50 mg), fast-disintegrating oral formulations (25 and 50 mg), powder for oral solution (50 mg), slow- and controlled-release forms (75, 100 or 150 mg), suppositories (50 and 100 mg), and injectable forms (50 and 75 mg).
Diclofenac is also available over-the-counter in some countries: 12.5 mg diclofenac as potassium salt in Switzerland (Voltaren dolo), the Netherlands (Voltaren K), and preparations containing 25 mg diclofenac as the potassium salt in Germany (various trade names), New Zealand, Australia, Japan, (Voltaren Rapid), and Sweden (Voltaren T and Diclofenac T). Diclofenac as potassium salt can be found throughout the Middle East in 25 mg and 50 mg doses (Cataflam).
Solaraze (3% diclofenac sodium gel) is topically applied, twice a day for three months, to manage the skin condition known as actinic or solar keratosis. Parazone-DP is a combination of diclofenac potassium and paracetamol, manufactured and supplied by Ozone Pharmaceuticals and Chemicals, Gujarat, India. It is sold in Uruguay alone or, in combination with orphenadrine to treat muscle spasms/pain due to injuries (Dicloflex Ion).
On 14 January 2015, diclofenac oral preparations were reclassified as prescription-only medicines in the UK. The topical preparations are still available without prescription.[53]
Diclofenac formulations are available worldwide under many different trade names.[1]
![]() |
|
Title: Diclofenac
CAS Registry Number: 15307-86-5
CAS Name: 2-[(2,6-Dichlorophenyl)amino]benzeneacetic acid
Additional Names: [o-(2,6-dichloroanilino)phenyl]acetic acid
Trademarks: Motifene (Sankyo)
Molecular Formula: C14H11Cl2NO2
Molecular Weight: 296.15
Percent Composition: C 56.78%, H 3.74%, Cl 23.94%, N 4.73%, O 10.80%
Literature References: Prepn: NL 6604752; A. Sallmann, R. Pfister, US 3558690 (1966, 1971 both to Geigy). Pharmacology: Renaud, Lecompte, Thromb. Diath. Haemorrh. 24, 577 (1970), C.A. 74, 86215m (1971); Krupp et al., Experientia 29, 450 (1973). HPLC determn in plasma and urine: J. Godbillon et al., J. Chromatogr. 338, 151 (1985). Symposium on pharmacology and clinical experience: Semin. Arthritis Rheum. 15, Suppl. 1, 57-110 (1985); on pharmacology, efficacy and safety: Am. J. Med. 80, Suppl. 4B, 1-87 (1986). Comprehensive description: C. M. Adeyeye, P-K. Li, Anal. Profiles Drug Subs. 19, 123-144 (1990). Review of clinical trials in actinic keratosis: D. C. Peters, R. H. Foster, Drugs Aging 14, 313-319 (1999).
Properties: Crystals from ether-petr ether, mp 156-158°.
Melting point: mp 156-158°
Derivative Type: Diethylammonium salt
CAS Registry Number: 78213-16-8
Trademarks: Voltarol (Novartis)
Molecular Formula: C14H11Cl2NO2.C4H11N
Molecular Weight: 369.29
Percent Composition: C 58.54%, H 6.00%, Cl 19.20%, N 7.59%, O 8.66%
Derivative Type: Sodium salt
CAS Registry Number: 15307-79-6
Manufacturers’ Codes: GP-45840
Trademarks: Allvoran (TAD); Benfofen (Sanofi-Synthelabo); Dealgic (Pharmacia); Deflamat (Sankyo); Delphinac (Riemser); Dicloflex (Dexcel); Diclomax (Provalis); Diclophlogont (Azupharma); Dicloreum (Alfa); Duravolten (Dura); Ecofenac (Ecosol); Effekton (Teofarma); Lexobene (Merckle); Neriodin (Nagase); Novapirina (Novartis); Primofenac (Streuli); Prophenatin (Nipro); Rewodina (AWD); Rhumalgan (Sandoz); Voldal (Novartis); Voltaren (Novartis); Xenid (RPG)
Molecular Formula: C14H10Cl2NNaO2
Molecular Weight: 318.13
Percent Composition: C 52.86%, H 3.17%, Cl 22.29%, N 4.40%, Na 7.23%, O 10.06%
Properties: Crystals from water, mp 283-285°. uv max (methanol) 283 nm (e 1.05 ´ 105); (phosphate buffer, pH 7.2) 276 nm (e1.01 ´ 105). Soly at 25°C (mg/ml): deionized water (pH 5.2) >9; methanol >24; acetone 6; acetonitrile <1; cyclohexane <1; HCl (pH 1.1) <1; phosphate buffer (pH 7.2) 6. pKa 4. Partition coefficient (N-octanol/aq. buffer): 13.4. LD50 in mice, rats (mg/kg): ~390, 150 orally (Krupp).
Melting point: mp 283-285°
pKa: pKa 4
Log P: Partition coefficient (N-octanol/aq. buffer): 13.4
Absorption maximum: uv max (methanol) 283 nm (e 1.05 ´ 105); (phosphate buffer, pH 7.2) 276 nm (e 1.01 ´ 105)
Toxicity data: LD50 in mice, rats (mg/kg): ~390, 150 orally (Krupp)
Derivative Type: Potassium salt
CAS Registry Number: 15307-81-0
Manufacturers’ Codes: CGP-45840B
Trademarks: Cataflam (Novartis)
Molecular Formula: C14H10Cl2KNO2
Molecular Weight: 334.24
Percent Composition: C 50.31%, H 3.02%, Cl 21.21%, K 11.70%, N 4.19%, O 9.57%
Therap-Cat: Anti-inflammatory.
Keywords: Anti-inflammatory (Nonsteroidal); Arylacetic Acid Derivatives.
|
Synthesis
![]()

The mechanism begins with the condensation of hydrazine onto a ketone (details not shown) to give a hydrazone. Under basic conditions, this hydrazone is deprotonated at nitrogen to give an anionic intermediate. In this case, the negative charge can be delocalized onto oxygen, resulting in an enolate structure. Typically, the negative charge is only shared between a nitrogen and carbon, so this substrate gives a particularly stable intermediate. Protonation of the enolate at carbon gives the first C-H bond necessary to form the product. A second deprotonation at nitrogen gives a similar flow of electrons to form another enolate structure, this time with cleavage of the C-N bond and release of nitrogen gas. Another C-protonation gives the lactam precursor to diclofenac. Cleavage of the amide with hydroxide (details not shown) gives the target.
Manufacturing Process
2, 6-Dichlorophenol is reacted with MMCA, Aniline and Chloro Acetyl Chloride and AlCl3 to yield (2, 6 –
Dichlorophenol) Indolinone is hydrolyzed using isopropyl alcohol and sodium hydroxide to give crude Diclofenac
Sodium. This on purification using deminerlised water and isopropyl alcohol gives the pure Diclofenac Sodium






CLIP





References
- ^ Jump up to:a b “Diclofenac”. Drugs.com. Retrieved 22 December 2018.
- ^ Mujib sayyad (August 23, 2018). “Diclofenac Oral Uses, Dosage, Side Effects And Composition”. Medicine Reviews Agency.
- ^ Jump up to:a b c d e f g h i “Diclofenac epolamine Monograph for Professionals”. Drugs.com. AHFS. Retrieved 22 December 2018.
- ^ Jump up to:a b c d British national formulary : BNF 74 (74 ed.). British Medical Association. 2017. pp. 1033–1035. ISBN 978-0857112989.
- ^ Mosby’s Drug Reference for Health Professions. Elsevier Health Sciences. 2017. p. 398. ISBN 9780323566827.
- ^ Fischer, Janos (2006). Analogue-based drug discovery. Wiley-VCH. p. 517. ISBN 3527312579.
- ^ “NADAC as of 2018-12-19”. Centers for Medicare and Medicaid Services. Retrieved 22 December 2018.
- ^ “The Top 300 of 2019”. clincalc.com. Retrieved 22 December 2018.
- ^ “Diclofenac Epolamine”. The American Society of Health-System Pharmacists. Retrieved 3 April 2011.
- ^ Jump up to:a b “RUFENAL”. Birzeit Pharmaceutical Company. Archived from the original on 2011-05-26.
- ^ “Patient Site – CAMBIA (diclofenac potassium) for oral solution”. cambiarx.
- ^ Dutta, NK; Mazumdar, K; Dastidar, SG; Park, JH (October 2007). “Activity of diclofenac used alone and in combination with streptomycin against Mycobacterium tuberculosis in mice”. International journal of antimicrobial agents. 30 (4): 336–40. doi:10.1016/j.ijantimicag.2007.04.016. PMID 17644321.
- ^ “Diclofenac (Topical Application Route) Description and Brand Names – Mayo Clinic”. http://www.mayoclinic.com.
- ^ cbg-meb.nl, SPC Netherlands
- ^ Wakai A, Lawrenson JG, Lawrenson AL, Wang Y, Brown MD, Quirke M, Ghandour O, McCormick R, Walsh CD, Amayem A, Lang E, Harrison N (2017). “Topical non-steroidal anti-inflammatory drugs for analgesia in traumatic corneal abrasions”. Cochrane Database Syst Rev. 5: CD009781. doi:10.1002/14651858.CD009781.pub2. PMID 28516471.
- ^ “WHO – WHO’s cancer pain ladder for adults”. http://www.who.int.
- ^ “Diclofenac Potassium”. Drugs.com. Drugsite Trust. Retrieved 2015-11-15.
- ^ Jump up to:a b c d e f g h Bhala, N.; Emberson, J.; et al. (2013). “Vascular and upper gastrointestinal effects of non-steroidal anti-inflammatory drugs: meta-analyses of individual participant data from randomised trials”. The Lancet. 382 (9894): 769–779. doi:10.1016/S0140-6736(13)60900-9. PMC 3778977. PMID 23726390.
- ^ Kearney PM, Baigent C, Godwin J, Halls H, Emberson JR, Patrono C (2006). “Do selective cyclo-oxygenase-2 inhibitors and traditional non-steroidal anti-inflammatory drugs increase the risk of atherothrombosis? Meta-analysis of randomised trials”. BMJ. 332(7553): 1302–8. doi:10.1136/bmj.332.7553.1302. PMC 1473048. PMID 16740558.
- ^ “Heart risk warning over painkiller”. 29 June 2013 – via http://www.bbc.co.uk.
- ^ “Press release: Diclofenac tablets now only available as a prescription medicine”. Medicines and Healthcare Products Regulatory Agency. January 14, 2015. Retrieved January 14, 2015.
- ^ Solomon DH, Avorn J, Stürmer T, Glynn RJ, Mogun H, Schneeweiss S (2006). “Cardiovascular outcomes in new users of coxibs and nonsteroidal antiinflammatory drugs: high-risk subgroups and time course of risk”. Arthritis Rheum. 54 (5): 1378–89. doi:10.1002/art.21887. PMID 16645966.
- ^ Fosbøl EL, Folke F, Jacobsen S, Rasmussen JN, Sørensen R, Schramm TK, Andersen SS, Rasmussen S, Poulsen HE, Køber L, Torp-Pedersen C, Gislason GH (2010). “Cause-Specific Cardiovascular Risk Associated With Nonsteroidal Antiinflammatory Drugs Among Healthy Individuals”. Circ Cardiovasc Qual Outcomes. 3 (4): 395–405. doi:10.1161/CIRCOUTCOMES.109.861104. PMID 20530789.
- ^ FitzGerald GA, Patrono C (2001). “The coxibs, selective inhibitors of cyclooxygenase-2”. N Engl J Med. 345 (6): 433–42. doi:10.1056/NEJM200108093450607. PMID 11496855.
- ^ “fda.gov”.
- ^ Jump up to:a b Brater DC (2002). “Renal effects of cyclooxygyenase-2-selective inhibitors”. J Pain Symptom Manage. 23 (4 Suppl): S15–20, discussion S21–3. doi:10.1016/S0885-3924(02)00370-6. PMID 11992745.
- ^ Becker, Rachel. “Cattle drug threatens thousands of vultures”. Nature. doi:10.1038/nature.2016.19839.
- ^ “Diclofenac Side Effects”. Drugs.com. Retrieved 21 January 2013.
- ^ Dastidar SG, Ganguly K, Chaudhuri K, Chakrabarty AN (2000). “The anti-bacterial action of diclofenac shown by inhibition of DNA synthesis”. Int. J. Antimicrob. Agents. 14 (3): 249–51. doi:10.1016/S0924-8579(99)00159-4. PMID 10773497.
- ^ Cryer, B.; Feldman, M. (1998). “Cyclooxygenase-1 and Cyclooxygenase-2 Selectivity of Widely Used Nonsteroidal Anti-Inflammatory Drugs”. The American Journal of Medicine. 104(5): 413–421. doi:10.1016/S0002-9343(98)00091-6.
- ^ Fowler PD, Shadforth MF, Crook PR, John VA (1983). “Plasma and synovial fluid concentrations of diclofenac sodium and its major hydroxylated metabolites during long-term treatment of rheumatoid arthritis”. Eur. J. Clin. Pharmacol. 25 (3): 389–94. doi:10.1007/BF01037953. PMID 6628528.
- ^ Scholer. Pharmacology of Diclofenac Sodium. Am J of Medicine Volume 80 April 28, 1986
- ^ Voilley N, de Weille J, Mamet J, Lazdunski M: Nonsteroid anti-inflammatory drugs inhibit both the activity and the inflammation-induced expression of acid-sensing ion channels in nociceptors. J Neurosci. 2001 Oct 15;21(20):8026-33.
- ^ Oaks JL, Gilbert M, Virani MZ, Watson RT, Meteyer CU, Rideout BA, Shivaprasad HL, Ahmed S, Chaudhry MJ, Arshad M, Mahmood S, Ali A, Khan AA (2004). “Diclofenac residues as the cause of vulture population decline in Pakistan”. Nature. 427 (6975): 630–3. Bibcode:2004Natur.427..630O. doi:10.1038/nature02317. PMID 14745453.
- ^ Swan, Gerry E.; Cuthbert, Richard; Quevedo, Miguel; Green, Rhys E.; Pain, Deborah J.; Bartels, Paul; Cunningham, Andrew A.; Duncan, Neil; Meharg, Andrew A.; Oaks, J. Lindsay; Parry-Jones, Jemima; Shultz, Susanne; Taggart, Mark A.; Verdoorn, Gerhard; Wolter, Kerri (2006-06-22). “Toxicity of diclofenac to Gyps vultures”. Biology Letters. 2 (2): 279–282. doi:10.1098/rsbl.2005.0425. ISSN 1744-9561. PMC 1618889. PMID 17148382.
- ^ Naidoo V, Swan GE (August 2008). “Diclofenac toxicity in Gyps vulture is associated with decreased uric acid excretion and not renal portal vasoconstriction”. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 149 (3): 269–74. doi:10.1016/j.cbpc.2008.07.014. PMID 18727958.
- ^ “Vet drug ‘killing Asian vultures‘“. BBC News. 2004-02-28.
- ^ Jump up to:a b “Saving the Vultures from Extinction” (Press release). Press Information Bureau, Government of India. 2005-05-16. Retrieved 2006-05-12.
- ^ Jump up to:a b c Swan G, Naidoo V, Cuthbert R, Green RE, Pain DJ, Swarup D, Prakash V, Taggart M, Bekker L, Das D, Diekmann J, Diekmann M, Killian E, Meharg A, Patra RC, Saini M, Wolter K (2006). “Removing the threat of diclofenac to critically endangered Asian vultures”. PLoS Biol. 4 (3): e66. doi:10.1371/journal.pbio.0040066. PMC 1351921. PMID 16435886.
- ^ Phadnis, Mayuri (May 28, 2014). “Eagles fall prey to vulture-killing chemical”. Pune Mirror. Retrieved May 28, 2014.
- ^ Schwaiger J, Ferling H, Mallow U, Wintermayr H, Negele RD (2004). “Toxic effects of the non-steroidal anti-inflammatory drug diclofenac. Part I: Histopathological alterations and bioaccumulation in rainbow trout”. Aquat. Toxicol. 68 (2): 141–150. doi:10.1016/j.aquatox.2004.03.014. PMID 15145224.
- ^ Triebskorn R, Casper H, Heyd A, Eikemper R, Köhler HR, Schwaiger J (2004). “Toxic effects of the non-steroidal anti-inflammatory drug diclofenac. Part II: Cytological effects in liver, kidney, gills and intestine of rainbow trout (Oncorhynchus mykiss)”. Aquat. Toxicol. 68(2): 151–166. doi:10.1016/j.aquatox.2004.03.015. PMID 15145225.
- ^ Schwaiger & Triebskorn (2005). UBA-Berichte 29/05: 217-226.
- ^ Triebskorn R, Casper H, Scheil V, Schwaiger J (2007). “Ultrastructural effects of pharmaceuticals (carbamazepine, clofibric acid, metoprolol, diclofenac) in rainbow trout (Oncorhynchus mykiss) and common carp (Cyprinus carpio)”. Anal Bioanal Chem. 387 (4): 1405–16. doi:10.1007/s00216-006-1033-x. PMID 17216161.
- ^ Rattner BA, Whitehead MA, Gasper G, Meteyer CU, Link WA, Taggart MA, Meharg AA, Pattee OH, Pain DJ (2009). “Apparent tolerance of turkey vultures (Cathartes aura) to the non-steroidal anti-inflammatory drug diclofenac”. Environmental Toxicology and Chemistry. 27 (11): 2341–2345. doi:10.1897/08-123.1. PMID 18476752.
- ^ Walker, Matt (August 6, 2008). “Rabies tragedy follows loss of India’s vultures”. New Scientist.
- ^ Choudhary, Srishti (August 29, 2016). “‘Decline in vulture population has given rise to diseases’: Dr Vibhu Prakash”. The Indian Express. Retrieved December 12, 2018.
- ^ “E-010588/2015: answer given by Mr Andriukaitis on behalf of the Commission”. European Parliament. Retrieved 2 May 2016.
- ^ Becker, Rachel. “Cattle drug threatens thousands of vultures”. Nature. Retrieved 2 May2016.
- ^ International, BirdLife. “Vulture killing drug now available on EU market”. http://www.birdlife.org.
- ^ Altman, R; Bosch, B; Brune, K; Patrignani, P; Young, C (May 2015). “Advances in NSAID development: evolution of diclofenac products using pharmaceutical technology”. Drugs. 75(8): 859–77. doi:10.1007/s40265-015-0392-z. PMID 25963327.
- ^ “A breakdown of the over-the-counter medicines market in Britain in 2016”. Pharmaceutical Journal. 28 April 2017. Retrieved 29 May 2017.
- ^ “Oral diclofenac presentations with legal status ‘P’ – reclassified to POM – GOV.UK”. http://www.gov.uk.
External links
References
-
- US 3 558 690 (Geigy; 26.1.1971; CH-prior. 8.4.1965, 25.2.1966, 30.3.1966, 20.12.1967).
- DAS 1 543 639 (Ciba-Geigy; appl. 7.4.1966; CH-prior. 8.4.1965).
- DAS 1 793 592 (Ciba-Geigy; appl. 7.4.1966; CH-prior. 8.4.1965).
- US 3 652 762 (Ciba-Geigy; 28.3.1972; prior. 9.12.1968, 29.9.1969, 14.4.1970).
- US 3 778 470 (Geigy; 11.12.1973; appl. 2.10.1970; prior. 4.4.1966).
- CH 492 679 (Geigy; appl. 30.3.1966).
-
Alternative synthesis:
- DOS 2 613 838 (Ikeda Mohando; appl. 31.3.1976; J-prior. 31.3.1975).
| Clinical data | |
|---|---|
| Trade names | Cataflam, Voltaren, others[1] |
| AHFS/Drugs.com | Monograph |
| MedlinePlus | a689002 |
| Pregnancy category |
|
| Routes of administration |
By mouth, rectal, intramuscular, intravenous(renal- and gallstones), topical |
| ATC code | |
| Legal status | |
| Legal status | |
| Pharmacokinetic data | |
| Protein binding | More than 99% |
| Metabolism | Liver, oxidative, primarily by CYP2C9, also by CYP2C8, CYP3A4, as well as conjugative by glucuronidation (UGT2B7) and sulfation;[2] no active metabolites exist |
| Elimination half-life | 1.2–2 hr (35% of the drug enters enterohepatic recirculation) |
| Excretion | 40% biliary 60% urine |
| Identifiers | |
| CAS Number | |
| PubChem CID | |
| IUPHAR/BPS | |
| DrugBank | |
| ChemSpider | |
| UNII | |
| KEGG | |
| ChEBI | |
| ChEMBL | |
| PDB ligand | |
| ECHA InfoCard | 100.035.755 |
| Chemical and physical data | |
| Formula | C14H11Cl2NO2 |
| Molar mass | 296.148 g/mol |
| 3D model (JSmol) | |
Diclofenac
-
- ATC:M01AB05; M02AA15; S01BC03
- Use:anti-inflammatory, antirheumatic
- Chemical name:2-[(2,6-dichlorophenyl)amino]benzeneacetic acid
- Formula:C14H11Cl2NO2
- MW:296.15 g/mol
- CAS-RN:15307-86-5
- InChI Key:DCOPUUMXTXDBNB-UHFFFAOYSA-N
- InChI:InChI=1S/C14H11Cl2NO2/c15-10-5-3-6-11(16)14(10)17-12-7-2-1-4-9(12)8-13(18)19/h1-7,17H,8H2,(H,18,19)
- EINECS:239-348-5
- LD50:170 mg/kg (M, p.o.);
62.5 mg/kg (R, p.o.)
Monosodium salt
- Formula:C14H10Cl2NNaO2
- MW:318.14 g/mol
- CAS-RN:15307-79-6
- EINECS:239-346-4
- LD50:116 mg/kg (M, i.v.); 390 mg/kg (M, p.o.);
117 mg/kg (R, i.v.); 150 mg/kg (R, p.o.)
//////////////Diclofenac Sodium
C1=CC=C(C(=C1)CC(=O)[O-])NC2=C(C=CC=C2Cl)Cl.[Na+]
Diclofenac Sodium
| FDA Orange Book Patents: 2 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9339552 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 3 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9415029 |
| Expiration | Jul 10, 2029 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 4 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9370501 |
| Expiration | Jul 10, 2029 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 5 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9375412 |
| Expiration | Jul 10, 2029 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 6 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 8946292 |
| Expiration | Mar 22, 2027 |
| Applicant | JAVELIN PHARMS INC |
| Drug Application | N022396 (Prescription Drug: DYLOJECT. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 7 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9168305 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 8 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9168304 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 9 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9220784 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 10 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 6407079 |
| Expiration | Jun 18, 2019 |
| Applicant | JAVELIN PHARMS INC |
| Drug Application | N022396 (Prescription Drug: DYLOJECT. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 11 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 8252838 |
| Expiration | Apr 21, 2028 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 12 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 8618164 |
| Expiration | Jul 10, 2029 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 13 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 8546450 |
| Expiration | Aug 9, 2030 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 14 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 8217078 |
| Expiration | Jul 10, 2029 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 15 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 8563613 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 16 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 8871809 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 17 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9066913 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 18 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 8741956 |
| Expiration | Jul 10, 2029 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 19 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9101591 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 20 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9132110 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
| FDA Orange Book Patents: 21 of 21 (FDA Orange Book Patent ID) | |
|---|---|
| Patent | 9539335 |
| Expiration | Oct 17, 2027 |
| Applicant | HZNP |
| Drug Application | N204623 (Prescription Drug: PENNSAID. Ingredients: DICLOFENAC SODIUM) |
Clotrimazole
![]()
Clotrimazole
- Molecular FormulaC22H17ClN2
- Average mass344.837 Da
![]() |
|
Title: Clotrimazole
CAS Registry Number: 23593-75-1
CAS Name: 1-[(2-Chlorophenyl)diphenylmethyl]-1H-imidazole
Additional Names: 1-(o-chloro-a,a-diphenylbenzyl)imidazole; 1-[a-(2-chlorophenyl)benzhydryl]imidazole; 1-[(o-chlorophenyl)diphenylmethyl]imidazole; diphenyl-(2-chlorophenyl)-1-imidazolylmethane; 1-(o-chlorotrityl)imidazole
Manufacturers’ Codes: FB-5097; Bay b 5097
Trademarks: Canesten (Bayer); Canifug (Wolff); Empecid (Bayer-Takeda); Gyne-Lotrimin (Schering-Plough); Lotrimin (Schering-Plough); Mono-Baycuten; Mycelex-G (Miles); Mycofug (Hermal); Mycosporin (Bayer); Pedisafe (Sagitta); Rimazole (Cheil Sugar); Tibatin (Dak); Trimysten
Molecular Formula: C22H17ClN2
Molecular Weight: 344.84
Percent Composition: C 76.63%, H 4.97%, Cl 10.28%, N 8.12%
Literature References: Prepn: K. H. Buechel et al., ZA 6805392; eidem, US 3705172 (1969, 1972 both to Bayer). Pharmacology: Plempel et al., Antimicrob. Agents Chemother. 1969, 271; eidem, Dtsch. Med. Wochenschr. 94, 1356 (1969). Clinical findings: Oberste-Lehn et al., ibid. 1365. Series of articles on prepn, toxicology, pharmacokinetics, clinical studies: Arzneim.-Forsch. 22,1260-1272, 1276-1299 (1972). Toxicity: D. Tettenborn, ibid. 1276. Comprehensive description: J. G. Hoogerheide, B. E. Wyka, Anal. Profiles Drug Subs. 11, 225-255 (1982).
Properties: Crystals, mp 147-149°. A weak base, slightly sol in water, benzene, toluene; sol in acetone, chloroform, ethyl acetate, DMF. Hydrolyzes rapidly upon heating in aq acids. LD50 in male mice, rats (mg/kg): 923, 708 orally (Tettenborn).
Melting point: mp 147-149°
Toxicity data: LD50 in male mice, rats (mg/kg): 923, 708 orally (Tettenborn)
Derivative Type: Hydrochloride
Molecular Formula: C22H17ClN2.HCl
Molecular Weight: 381.30
Percent Composition: C 69.30%, H 4.76%, Cl 18.60%, N 7.35%
Properties: mp 159°.
Melting point: mp 159°
Therap-Cat: Antifungal.
Therap-Cat-Vet: Antifungal.
Keywords: Antifungal (Synthetic); Imidazoles.
|
Clotrimazole, sold under the brand name Canesten among others, is an antifungal medication.[1] It is used to treat vaginal yeast infections, oral thrush, diaper rash, pityriasis versicolor, and types of ringworm including athlete’s foot and jock itch.[1] It can be taken by mouth or applied as a cream to the skin or in the vagina.[1]
Common side effects when taken by mouth include nausea and itchiness.[1] When applied to the skin common side effects include redness and burning.[1] In pregnancy, use on the skin or in the vagina is believed to be safe.[1] There is no evidence of harm when used by mouth during pregnancy but this has been less well studied.[1] When used by mouth, greater care should be taken in those with liver problems.[1] It is in the azole class of medications and works by disrupting the cell membrane.[1]
Clotrimazole was discovered in 1969.[2] It is on the World Health Organization’s List of Essential Medicines, the most effective and safe medicines needed in a health system.[3] It is available as a generic medication.[1] The wholesale cost in the developing world as of 2014 is 0.20–0.86 USD per 20 gram tube of cream.[4] In the United States a course of treatment typically costs less than 25 USD.[5]
It is commonly available without a prescription in various dosage forms, such as a cream, vaginal tablet, or as a prescription troche or throat lozenge (prescription only). Topically, clotrimazole is used for vulvovaginal candidiasis (yeast infection) or yeast infections of the skin. For vulvovaginal candidiasis (yeast infection), clotrimazole tablets and creams are inserted into the vagina. Troche or throat lozenge preparations are used for oropharyngeal candidiasis (oral thrush) or prophylaxis against oral thrush in neutropenic patients.
Clotrimazole is usually used 5 times daily for 14 days for oral thrush, twice daily for 2 to 8 weeks for skin infections, and once daily for 3 or 7 days for vaginal infections.[6]
Clotrimazole may be compounded with a glucocorticoid, such as betamethasone, in a topical cream for the treatment of tinea corporis (ringworm), tinea cruris (jock itch) and tinea pedis (athlete’s foot). Although FDA approved, clotrimazole-betamethasone combination cream is not the preferred treatment for dermatophyte infections due to increased side effects from the topical glucocorticoid. Although temporary relief and partial suppression of symptoms may be observed with the combination therapy, glucocorticoids can elicit an immunosuppressive response and rebound effect that results in more severe infection typically requiring systemic antifungal agents to treat the disease. Combination creams are best avoided in order to improve treatment outcome, reduce the possibility of skin atrophy associated with prolonged topical glucocorticoid use, and to limit the cost of treatment. It can be effective in treating chronic paronychia. The preferred treatment of tinea infections is therefore with clotrimazole monotherapy.[7]
Topical and oral clotrimazole can be used in both adults and children.
Additionally, clotrimazole may be used to treat the sickling of cells (related to sickle cell anemia).[8][9]
Pregnancy
Small amounts of clotrimazole may be absorbed systemically following topical and vaginal administration. However, this may still be used to treat yeast infections in pregnant women.[10]
Side effects
Side effects of the oral formulation include itching, nausea, and vomiting. >10% of patients using the oral formulation may have abnormal liver function tests. Side effects include rash, hives, blisters, burning, itching, peeling, redness, swelling, pain or other signs of skin irritation.[1] For this reason, liver function tests should be monitored periodically when taking the oral clotrimazole (troche). When used to treat vulvovaginal candidiasis (yeast infection), <10% of patient have vulvar or vaginal burning sensation. <1% of patients have the following side effects: Burning or itching of penis of sexual partner; polyuria; vulvar itching, soreness, edema, or discharge [6][11][12]
Clotrimazole creams and suppositories contain oil which may weaken latex condoms and diaphragms.[10]
Drug interactions
There are no known significant drug interactions with topical clotrimazole. However, with oral (troche) clotrimazole, there are multiple interactions as the medication is a CYP450 enzyme inhibitor, primarily CYP3A4. Thus, any medication that is metabolized by the CYP3A4 enzyme will potentially have elevated levels when oral clotrimazole is used. The prescribing physician should be aware of any medication the patient is taking prior to starting oral clotrimazole. Certain medications should not be taken with oral clotrimazole.[11]
Mechanism of action
Clotrimazole works by inhibiting the growth of individual Candida or fungal cells by altering the permeability of the fungal cell wall. It binds to phospholipids in the cell membrane and inhibits the biosynthesis of ergosterol and other sterols required for cell membrane production.[12][11] Clotrimazole may be fungistatic (slow fungal growth) or fungicidal (result in fungal cell death).[1]
Society and culture
Clotrimazole (Canesten) antifungal cream
It is available as a generic medication.[1] The wholesale cost in the developing world as of 2014 is 0.20–0.86 USD per 20gm tube of cream.[4]In the United States a course of treatment typically costs less than 25 USD.[5] In 2016 Canesten was one of the biggest selling branded over-the-counter medications sold in Great Britain, with sales of £39.2 million.[13]
syn


CLIP
Fig 4. Open Babel bond-line chemical structure with annotated hydrogens.
Click to toggle size.
Spectrum Plot
Fig 5. 1H NMR spectrum of C22H17ClN2 in CDCL3 at 400 MHz.

Figure 7. 2D 13 C13 C refocused INADEQUATE spectrum of clotrimazole showing intramolecular contacts among 13 C resonances as marked in the molecular structure on the right. The full spectrum is included in the Figure S4. The 2D spectrum was acquired in 17 hr at 106 K on 400 MHz, 384 scans per increment, 2 s recycle delay and 80 t 1 increments of a 27.7 ?s.


2D 13C-13C refocused INADEQUATE spectrum of clotrimazole showing intramolecular contacts among 13C resonances as marked in the molecular structure on the right. The 2D spectrum was acquired in 17 hr at 106 K on 400 MHz.

PATENT
https://patents.google.com/patent/CN105566156A/en
The object of the present invention is to provide a method for synthesizing a pharmaceutical Clotrimazole intermediate o-chlorobenzonitrile, comprising the steps of:
[0004] (i) in a reaction container equipped with a stirrer, a thermometer, a distillation apparatus, was added o-chlorobenzyl alcohol (2) 3. lmol, aniline (3) 3.6-3 · 9mol, nitromethane burning 310ml, chloro cuprous 1 · 56mol, hook are mixed, controlling the stirring speed 110-160rpm, the solution temperature increased to 110-115 ° C, 3-5h the reaction, the solution temperature increased to 130-135 ° C, the reaction 2-3h, solution temperature increased to 190-195 ° C, the reaction 90-120min, reducing the solution temperature to 15-20 ° C, was added 700 ml of saline solution, sodium bisulfite solution, 130ml, distilled under reduced pressure to collect 130-135 ° C fraction , washed with triethylamine in toluene and recrystallized to give crystals of o-chlorobenzonitrile (1).
[0005] wherein the mass fraction of nitromethane according to step (i) is 60-65%, of the salt solution in step (i) is ammonium nitrate, potassium iodide to any one of the steps of (i) mass fraction of sodium hydrogen sulfite solution was 40-45%, which pressure in the vacuum distillation of step (i) is 1.6-1.7kPa, triethylamine mass fraction of said step (i) is 70-75%, step (i) in toluene of the mass fraction of 90-95%. Throughout the reaction using the following reaction formula:
[0006
[0007 “not as good as Wu Ming 1 point Shi Bian: J Cheng less
A slave I anti Day “* 1, section A, J array low reaction temperature and reaction time, the reaction yield improved.
Detailed ways
[0008] The following examples with reference to specific embodiments of the present invention is further described:
Clotrimazole synthesis kinds drug intermediates of o-chlorobenzonitrile – [0009]
[0010] Example 1:
[0011] In a reaction vessel fitted with a stirrer, a thermometer, a distillation apparatus, was added o-chlorobenzyl alcohol (2) 3. Lmol, aniline (3) 3.6111〇1, mass fraction of 60% nitromethane 3,101,111 chloride cuprous 1.56111 〇1, mixing, stirring speed control lOrpm 1, the solution temperature increased to 110 ° C, the reaction 3h, the solution temperature increased to 130 ° C, the reaction 2h, the solution temperature is raised to 190 ° (:, reaction 9011 ^ 11, reducing the solution temperature to 15 ° (:, 7,001,111 ammonium nitrate solution was added, the mass fraction of 40% sodium bisulfite solution was 130ml, 1.6kPa vacuum distillation, collecting the fraction 130-135 ° C, mass fraction of 70 washed% triethylamine, 90% toluene to a mass fraction of recrystallized to give crystals of o-chlorobenzonitrile 308.02g, yield 72%.
[0012] Example 2:
[0013] In a reaction vessel fitted with a stirrer, a thermometer, a distillation apparatus, was added o-chlorobenzyl alcohol (2) 3. Lmol, aniline (3) 3.7111〇1, mass fraction of 62% nitromethane 31〇1111, 1.56111〇1 cuprous chloride, mixed, controlling the stirring speed of 130 rpm, the temperature was raised to 112 ° C, the reaction 4h, the solution temperature increased to 132 ° C, the reaction 2h, the solution temperature increased to 192 ° C, the reaction llOmin, reducing the solution temperature to 17 ° C, 700 ml of a solution of potassium iodide was added, the mass fraction of 42% sodium bisulfite solution 130ml, 1.65kPa vacuum distillation, collecting the fraction 130-135 ° C, mass fraction of 72% triethylamine washed, recrystallized from toluene to 92% mass fraction, to obtain crystals of o-chlorobenzonitrile 337.96g, yield 79%.
[0014] Example 3:
[0015] In a reaction vessel fitted with a stirrer, a thermometer, a distillation apparatus, was added o-chlorobenzyl alcohol (2) 3. Lmol, aniline (3) 3.9111〇1, mass fraction of 65% nitromethane 31〇1111, 1.56111 〇1 cuprous chloride, mixed, controlling stirring speed 160 rpm, temperature was raised to 115 ° C, the reaction 5h, the solution temperature increased to 135 ° C, the reaction 3h, the solution temperature increased to 195 ° C, the reaction 120min, reducing the solution temperature to 20 ° C, was added 700 ml of a solution of ammonium nitrate, 45% mass fraction of sodium bisulfite solution was 130ml, 1.7kPa vacuum distillation, collecting the fraction 130-135 ° C, mass fraction of 75% triacetyl amine scrubbing, 95%, recrystallized from toluene to a mass fraction to obtain crystals of o-chlorobenzonitrile 350.80g, yield 82%.
PATENT
https://patents.google.com/patent/US5091540A/en
Clotrimazole, i.e. 1-(o.Cl-α,α-diphenylbenzyl)imidazole, of formula: ##STR1## is a known antimycotic for human use, and a fungicide useful against plant pathogenic fungi.
Methods for its preparation are described in various patents. In particular, U.S. Pat. No. 3,929,820 describes a process starting from chlorophenyldiphenyl methylchloride and imidazole in the presence of a neutralizing agent, such as triethylamine, in a polar organic solvent. The process is strictly limited by the use, as the medium for the reaction in question, of a solvent falling within the given definition, i.e. having a dielectric constant of at least 4.5 and preferably between 15 and 50. In all the examples of the implementation of the process according to the patent in question, acetonitrile (D=37.5) is used as solvent.
EXAMPLE
900 g of benzene and 117.5 g of aluminium chloride are placed in a 2 liter flask fitted with a reflux condenser, stirrer and drying tube.
The mixture is cooled to 0° C. and a solution of 150 g of o.chlorobenzotrichloride in 150 g of benzene is added while maintaining a temperature not exceeding 15° C. The mixture is heated carefully under reflux for 4 hours. HCl is evolved.
The reaction mixture is then cooled to ambient temperature and slowly poured into 300 g of concentrated hydrochloric acid and 800 g of ice, so as not to exceed 25° C. The aqueous layer is then separated and discarded.
The benzene solution is washed with a solution of 230 g of sodium chloride in 800 g of water. The benzene phase is separated and dried over anhydrous sodium sulphate for 1 hour, and then filtered.
45 g of imidazole in 70 g of triethylamine are added to the filtrate and the mixture heated for 3 hours at 45°-50° C. It is then cooled to ambient temperature and 500 g of water are added while stirring. The aqueous layer is separated and discarded, and the benzene phase washed with 200 g of water. The benzene layer is separated and evaporated to dryness under vacuum.
The residue is dissolved in 250 g of ethyl acetate while stirring. 250 g of water are added and the solution titrated to calculate the exact quantity of nitric acid to add.
The solution is cooled to 15° C. and the calculated nitric acid quantity is quickly added. Stirring is halted when precipitation commences, and the system left until precipitation is complete.
The product is centrifuged and washed with 300 g of ethyl acetate and then with 300 g of water.
The moist product is placed into the reaction flask and 300 g of water, 450 g of methylene chloride, 5 g of triethylamine and 110 g of 30% sodium hydroxide are added. The mixture is stirred until a solution forms and the solution then left until the phases separate.
The aqueous phase is washed with 100 g of methylene chloride, and the pooled organic phases are washed twice with 200 g of water each time.
The solution in methylene chloride is treated with YMS decolorizing carbon and filtered, the filter then being washed with methylene chloride which si recovered by distillation. The residue is taken up in 100 g of acetone and redistilled to completely eliminate the methylene chloride.
The residue is taken up in 900 g of acetone and heated to 50° C. to obtain a complete solution. YMS decolorizing carbon and triethylamine are added, the mixture filtered and washed with acetone. Part of the acetone is then removed by distillation, reducing the volume to about 500 c.c. The mixture is cooled to 0° C. and, after five hours, the product is centrifuged and washed with 100 g of acetone. It is dried at 60° C., to obtain 150 g of final product.
References
- ^ Jump up to:a b c d e f g h i j k l m American Society of Health-System Pharmacists (8 February 2016). “Clotrimazole Monograph for Professionals”. http://www.drugs.com. Archived from the original on 28 October 2016. Retrieved 28 October 2016.
- ^ Walker, S. R. (2012). Trends and Changes in Drug Research and Development. Springer Science & Business Media. p. 109. ISBN 9789400926592. Archived from the original on 2016-09-14.
- ^ “WHO Model List of Essential Medicines (19th List)” (PDF). World Health Organization. April 2015. Archived (PDF) from the original on 13 December 2016. Retrieved 8 December 2016.
- ^ Jump up to:a b “Clotrimazole”. International Drug Price Indicator Guide. Archived from the original on 10 May 2017. Retrieved 28 October 2016.
- ^ Jump up to:a b Tarascon Pharmacopoeia 2016 Professional Desk Reference Edition. Jones & Bartlett Publishers. 2016. p. 176. ISBN 9781284095302. Archived from the original on 2016-10-28.
- ^ Jump up to:a b “Clotrimazole: MedlinePlus Drug Information”. The American Society of Health-System Pharmacists, Inc. Archived from the original on 18 April 2014. Retrieved 17 April2014.
- ^ Moriarty, B; Hay, R; Morris-Jones, R (10 July 2012). “The diagnosis and management of tinea”. BMJ (Clinical research ed.). 345: e4380. doi:10.1136/bmj.e4380. PMID 22782730.
- ^ Marieb & Hoehn, (2010). Human Anatomy and Physiology, p. 643. Toronto: Pearson
- ^ Rodgers, Griffin. “Hydroxyurea and other disease-modifying therapies in sickle cell disease”. UpToDate. Archived from the original on 15 April 2014. Retrieved 14 April2014.
- ^ Jump up to:a b “Diseases Characterized by Vaginal Discharge”. CDC. Archived from the original on 28 April 2014. Retrieved 17 April 2014.
- ^ Jump up to:a b c “Clotrimazole”. DrugBank. Archived from the original on 17 April 2014. Retrieved 17 April 2014.
- ^ Jump up to:a b “Clotrimazole (Oral)”. Lexicomp Online. Archived from the original on 23 January 2015. Retrieved 17 April 2014.
- ^ “A breakdown of the over-the-counter medicines market in Britain in 2016”. Pharmaceutical Journal. 28 April 2017. Retrieved 29 May 2017.
/////////////clotrimazole
READ
ANTHONY MELVIN CRASTO
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO …..FOR BLOG HOME CLICK HERE
amcrasto@gmail.com
CALL +919323115463 INDIA
//////////////
SELETALISIB, селеталисиб , سيلستاليسيب , 司来利塞 ,

SELETALISIB
CAS 1362850-20-1
UCB-5857 , Plaque psoriasis,Sjoegren’s syndrome,Immunodeficiency disorders
PHASE 3 UCB
23H14ClF3N6O , 482.85
Phosphatidylinositol 3 kinase delta (PI3Kδ) inhibitors
| Pyrido[3,2-d]pyrimidin-4-amine, N-[(1R)-1-[8-chloro-2-(1-oxido-3-pyridinyl)-3-quinolinyl]-2,2,2-trifluoroethyl]- |
3-{8-chloro-3-[(1R)-2,2,2-trifluoro-1-({pyrido[3,2-d]pyrimidin-4-yl}amino)ethyl]quinolin-2-yl}pyridin-1-ium-1-olate
Seletalisib has been used in trials studying the treatment and basic science of Primary Sjogren’s Syndrome.
- Originator UCB
- Class Anti-inflammatories; Small molecules
- Mechanism of Action Immunomodulators; Phosphatidylinositol 3 kinase delta inhibitors
- Phase III Immunodeficiency disorders
- Phase II Sjogren’s syndrome
- No development reported Plaque psoriasis
- 05 Dec 2017 UCB Celltech terminates a phase II trial in Sjogren’s syndrome in France, Spain, United Kingdom, Greece, Sweden, Italy, due to enrolment challenges (PO) (NCT02610543) (EudraCT2014-004523-51)
- 04 Nov 2017 No recent reports of development identified for phase-I development in Plaque-psoriasis in United Kingdom (PO, Capsule)
- 14 Jun 2017 Pharmacokinetics and pharmacodynamics data from Preclinical and Clinical studies in Immunodeficiency disorders presented at the 18th Annual Congress of the European League Against Rheumatism (EULAR-2017)
SYN
US 9029392
https://patents.google.com/patent/US9029392B2/en
Example 27 N—{(R)-1-[8-Chloro-2-(pyridin-3-yl)quinolin-3-yl]-2,2,2-trifluoroethyl}N-(1-oxypyrido-[3,2-d]pyrimidin-4-yl)amine
A stirred solution of Example 1 (955 mg, 2.05 mmol) in DCM (40 mL) was cooled to 0° C. MCPBA (410 mg, 1.84 mmol) was added and the mixture was allowed to warm slowly to r.t. over 3 h. The reaction mixture was partitioned between DCM and saturated aqueous NaHCO3 solution. The aqueous phase was extracted with further DCM and the combined organic fractions were washed with brine, dried Na2SO4) and evaporated in vacuo. The residue was purified by column chromatography (SiO2, 3-60% MeOH in EtOAc) to give the title compound (39 mg, 4%) as a yellow solid. δH (DMSO-d6) 9.64-9.52 (m, 1H), 9.30 (s, 1H), 9.06 (dd, J 4.2, 1.3 Hz, 1H), 8.78-8.71 (m, 2H), 8.67 (dd, J 4.9, 1.6 Hz, 1H), 8.64 (s, 1H), 8.16-8.01 (m, 4H), 7.75-7.69 (m, 1H), 7.52 (ddd, J 7.8, 4.9, 0.7 Hz, 1H), 6.65-6.52 (m, 1H). LCMS (ES+) 483 (M+H)+, RT 1.87 minutes.

AND

PATENT
WO 2012032334
PATENT
WO 2015181053
WO 2015181055
WO 2016170014
PATENT
A SPECIFIC TRIFLUOROETHYL QUINOLINE ANALOGUE FOR USE IN THE TREATMENT OF APDS
The present invention relates to the new therapeutic use of a known chemical compound. More particularly, the present invention concerns the use of a specific substituted quinoline derivative comprising a fluorinated ethyl side-chain in the treatment of activated phosphoinositide 3 -kinase delta syndrome (APDS).
N- {(R)- 1 -[8-Chloro-2-(l -oxypyridin-3-yl)quinolin-3-yl]-2,2,2-trifiuoroethyl} -pyrido[3,2-JJpyrimidin-4-ylamine is specifically disclosed in WO 2012/032334. The compounds described in that publication are stated to be of benefit as pharmaceutical agents, especially in the treatment of adverse inflammatory, autoimmune, cardiovascular, neurodegenerative, metabolic, oncological, nociceptive and ophthalmic conditions.
There is no specific disclosure or suggestion in WO 2012/032334, however, that the compounds described therein might be beneficial in the treatment of APDS.
Activated phosphoinositide 3-kinase delta syndrome (APDS), also known as
PASLI (pi ΙΟδ-activating mutation causing senescent T cells, lymphadenopathy and immunodeficiency), is a serious medical condition that impairs the immune system.
APDS patients generally have reduced numbers of white blood cells (lymphopenia), especially B cells and T cells, compromising their propensity to recognise and attack invading microorganisms, such as viruses and bacteria, and thereby prevent infection. Individuals affected with APDS develop recurrent infections, particularly in the lungs, sinuses and ears. Recurrent respiratory tract infections may gradually lead to bronchiectasis, a condition which damages the passages leading from the windpipe to the lungs (bronchi) and can cause breathing problems. APDS patients may also suffer from chronic active viral infections, including Epstein-Barr virus infections and cytomegalovirus infections.
APDS has also been associated with abnormal clumping of white blood cells, which can lead to enlarged lymph nodes (lymphadenopathy). Alternatively, the white blood cells can build up to form solid masses (nodular lymphoid hyperplasia), usually in the moist lining of the airways or intestines. Whilst lymphadenopathy and nodular lymphoid hyperplasia are benign (noncancerous), APDS also increases the risk of developing a form of cancer called B cell lymphoma.
APDS is a disorder of childhood, typically arising soon after birth. However, the precise prevalence of APDS is currently unknown.
Phosphoinositide 3-kinase delta (ΡΒΚδ) is a lipid kinase which catalyses the generation of phosphatidylinositol 3,4,5-trisphosphate (PIP3) from phosphatidylinositol 4,5-bisphosphate (PIP2). PI3K5 activates signalling pathways within cells, and is specifically found in white blood cells, including B cells and T cells. PI3K5 signalling is involved in the growth and division (proliferation) of white blood cells, and it helps direct B cells and T cells to mature (differentiate) into different types, each of which has a distinct function in the immune system.
APDS is known to occur in two variants, categorised as APDSl and APDS2.
APDSl is associated with a heterozygous gain-of- function mutation in the PIK3CD gene encoding the PI3K5 protein; whereas APDS2 is associated with loss-of-function frameshift mutations in the regulatory PIK3R1 gene encoding the p85a regulatory subunit of class I phosphoinositide 3-kinase (PI3K) peptides. Both mutations lead to hyperactivated PI3K signalling. See I. Angulo et ah, Science, 2013, 342, 866-871; C.L. Lucas et ah, Nature Immunol, 2014, 15, 88-97; and M-C. Deau et al, J. Clin. Invest., 2014, 124, 3923-3928.
There is currently no effective treatment available for APDS. Because of the seriousness of the condition, and the fact that it arises in infancy, the provision of an effective treatment for APDS would plainly be a highly desirable objective.
It has now been found that N-{(R)-l-[8-chloro-2-(l-oxypyridin-3-yl)quinolin-3-yl]- 2,2,2-trifluoroethyl}pyrido[3,2-(i]pyrimidin-4-ylamine is capable of inhibiting the elevation of PI3K signalling in T cells (lymphocytes) from both APDSl and APDS2 patients in the presence or absence of T cell receptor activation.
The present invention accordingly provides N-{(R)-l-[8-chloro-2-(l-oxypyridin-3-yl)quinolinB-yl]-2,2,2-trifluoroethyl}pyrido[3,2-JJpyrimidin-4-ylamine of formula (A):
or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prevention of APDS.
The present invention also provides a method for the treatment and/or prevention of APDS, which method comprises administering to a patient in need of such treatment an effective amount of N-{(R)-l-[8-chloro-2-(l-oxypyridin-3-yl)quinolin-3-yl]-2,2,2-trifluoro-ethyl}pyrido[3,2-(i]pyrimidin-4-ylamine of formula (A) as depicted above, or a pharmaceutically acceptable salt thereof. The present invention also provides the use of N-{(R)-l-[8-chloro-2-(l-oxypyridin-3-yl)quinolin-3-yl]-2,2,2-trifluoroethyl}pyrido[3,2-JJpyrimidin-4-ylamine of formula (A) as depicted above, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment and/or prevention of APDS.
PAPER
Journal of Pharmacology and Experimental Therapeutics (2017), 361(3), 429-440.
http://jpet.aspetjournals.org/content/361/3/429
///////////////SELETALISIB, PHASE 3, UCB, селеталисиб , سيلستاليسيب , 司来利塞 ,
[O-][N+]1=CC(=CC=C1)C1=NC2=C(Cl)C=CC=C2C=C1[C@@H](NC1=NC=NC2=CC=CN=C12)C(F)(F)F
READ
ANTHONY MELVIN CRASTO
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO …..FOR BLOG HOME CLICK HERE
amcrasto@gmail.com
CALL +919323115463 INDIA
Omadacycline tosylate
Omadacycline tosylate
728.8521, C29H40N4O7. C7H8O3S
CAS: 1075240-43-5
389139-89-3 FREE FORM
FDA 2018/10/3, Nuzyra
オマダサイクリントシル酸塩;
UNII-5658Y89YCD
Omadacycline has been used in trials studying the treatment of Bacterial Pneumonia, Bacterial Infections, Community-Acquired Infections, and Skin Structures and Soft Tissue Infections. Omadacycline represents a significant advance over the well-known tetracycline family, and has been shown to be highly effective in animal models at treating increasingly problematic, clinically prevalent infections caused by gram-positive bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), and by gram-negative, atypical and anaerobic bacteria, including those resistant to currently available classes of antibiotics and known to cause diseases such as pneumonias, urinary tract infections, skin diseases and blood-borne infections in both the hospital and community settings.
Omadacycline (formerly known as PTK-0796)[1] is a broad spectrum antibiotic belonging to the aminomethylcycline subclass[2] of tetracycline antibiotics. In the United States, it was approved in October 2018 for the treatment of community-acquired bacterial pneumonia and acute skin and skin structure infections.
In vitro studies
In vitro studies have shown that omadacycline has activity against a broad range of Gram-positive and select Gram-negativepathogens.[3] Omadacycline has potent in vitro activity against Gram-positive aerobic bacteria including methicillin-resistant Staphylococcus aureus (MRSA), pencillin-resistant and multi-drug resistant Streptococcus pneumoniae, and vancomycin-resistant Enterococcus. Omadacycline also has antimicrobial activity against common Gram-negative aerobes, some anaerobes, and atypical bacteria such as Legionella and Chlamydia.[4] This activity translated to potent efficacy for omadacycline in an in vivo systemic infection model in mice.[5]
Additional in vitro and in vivo studies of omadacycline metabolism, disposition, and drug interactions show that omadacycline is metabolically stable (i.e., it does not undergo significant biotransformation) and neither inhibits nor interacts with metabolizing enzymes or transporters.[6]
Mechanism of action
The mechanism of action of omadacycline is similar to that of other tetracyclines – inhibition of bacterial protein synthesis. Omadacycline has activity against bacterial strains expressing the two main forms of tetracycline resistance (efflux and ribosomal protection).[7]
Clinical trials
A phase 2 study was conducted comparing the safety and efficacy of omadacycline to linezolid for the treatment of complicated skin and skin structure infections. Patients were randomized at 11 sites in the US to receive either omadacycline 100 mg intravenously once daily with an option to transition to 200 mg orally once daily or linezolid 600 mg intravenously twice daily with an option to transition to 600 mg orally twice daily. The results indicated that omadacycline is well-tolerated and has the potential to be an effective treatment in patients with complicated skin and skin structure infections.[8]
In June 2013, the US Food and Drug Administration (FDA) designated the intravenous and oral formulations of omadacycline as a qualified infectious disease product in the treatment of acute bacterial skin and skin structure infections and community-acquired bacterial pneumonia.[9]
A 650 patient phase 3 registration study comparing omadacycline to linezolid for the treatment of acute bacterial skin and skin structure infections began in June 2015.[10][11]Omadacycline met the primary efficacy endpoint of early clinical response with statistical non-inferiority (10% margin) compared to linezolid, and was generally safe and well-tolerated. The most common treatment-emergent adverse events were gastrointestinal side effects (18.0% for omadacycline vs. 15.8% for linezolid).[12]
A 750 patient phase 3 study comparing omadacycline to moxifloxacin for the treatment of community-acquired bacterial pneumonia began in November 2015.[13] Omadacycline was statistically non-inferior to moxifloxacin at the early clinical response, 72 to 120 hours after therapy was initiated.[14]
In May 2016, a phase 1b study of omadacycline in urinary tract infection was initiated.[15]
In August 2016, a second phase 3 study of omadacycline was initiated in patients with acute bacterial skin and skin structure infections, comparing the efficacy and safety of once-daily, oral omadacycline to that of twice-daily, oral linezolid.[16] In July 2017, analysis of the data showed that all of the primary and secondary endpoints required for submission to the FDA and EMA were met. This was the third phase 3 registration study of omadacycline with favorable results.[17]
Discovery
Omadacycline was invented at Tufts University School of Medicine by a research team led by Mark L. Nelson with Mohamed Ismail while at Tufts and Kwasi Ohemeng and Laura Honeyman at Paratek Pharmaceuticals, Boston. The team applying their chemistry methods to the tetracycline scaffolds created over 3000 new derivatives, leading to the novel third generation compounds omadacycline and sarecycline. 18[18]
PAPERS
Tetrahedron Letters (2008), 49(42), 6095-6100

PATENTS
WO 2009120389
WO 2009111064
WO 2017165729
WO 2018026987
WO 2018085216
SYNTHESIS BY PHARMACODIA WEBSITE
Omadacycline, www.pharmacodia.com
REF Omadacycline, www.pharmacodia.com
References
- Jump up^ Boggs, Jennifer. “Antibiotic Firm Paratek Joins IPO Queue; Aiming for $92M”. bioworld.com. Clarivate Analytics. Retrieved October 17, 2017.
- Jump up^ Honeyman, Laura; Ismail, Mohamed; Nelson, Mark L.; Bhatia, Beena; Bowser, Todd E.; Chen, Jackson; Mechiche, Rachid; Ohemeng, Kwasi; Verma, Atul K.; Cannon, E. Pat; MacOne, Ann; Tanaka, S. Ken; Levy, Stuart (2015). “Structure-Activity Relationship of the Aminomethylcyclines and the Discovery of Omadacycline”. Antimicrobial Agents and Chemotherapy. 59 (11): 7044–7053. doi:10.1128/AAC.01536-15. PMC 4604364. PMID 26349824.
- Jump up^ Tanaka, S. Ken (20 June 2016). “In Vitro and In Vivo Assessment of Cardiovascular Effects with Omadacycline”. Antimicrobial Agents and Chemotherapy. 60 (9): 5247–53. doi:10.1128/AAC.00320-16. PMC 4997885. PMID 27324778.
- Jump up^ Villano, Stephen (19 August 2016). “Omadacycline: development of a novel aminomethylcycline antibiotic for treating drug-resistant bacterial infections”. Future Microbiology. 11: 1421–1434. doi:10.2217/fmb-2016-0100. Retrieved 24 August 2016.
- Jump up^ MacOne, A. B.; Caruso, B. K.; Leahy, R. G.; Donatelli, J.; Weir, S.; Draper, M. P.; Tanaka, S. K.; Levy, S. B. (February 2014). “In Vitro and in Vivo Antibacterial Activities of Omadacycline, a Novel Aminomethylcycline”. Antimicrobial Agents and Chemotherapy. 58 (2): 1127–1135. doi:10.1128/AAC.01242-13. PMC 3910882. PMID 24295985.
- Jump up^ Flarakos, Jimmy (8 August 2016). “Clinical disposition, metabolism and in vitro drug–drug interaction properties of omadacycline”. Xenobiotica: 1–15. doi:10.1080/00498254.2016.1213465.
- Jump up^ Draper, M. P.; Weir, S.; MacOne, A.; Donatelli, J.; Trieber, C. A.; Tanaka, S. K.; Levy, S. B. (March 2014). “Mechanism of Action of the Novel Aminomethylcycline Antibiotic Omadacycline”. Antimicrobial Agents and Chemotherapy. 58 (3): 1279–1283. doi:10.1128/AAC.01066-13. PMC 3957880. PMID 24041885.
- Jump up^ Noel, G. J.; Draper, M. P.; Hait, H.; Tanaka, S. K.; Arbeit, R. D. (November 2012). “A Randomized, Evaluator-Blind, Phase 2 Study Comparing the Safety and Efficacy of Omadacycline to Those of Linezolid for Treatment of Complicated Skin and Skin Structure Infections”. Antimicrobial Agents and Chemotherapy. 56 (11): 5650–5654. doi:10.1128/AAC.00948-12. PMC 3486554. PMID 22908151.
- Jump up^ “Paratek Pharmaceuticals Announces FDA Grant of Qualified Infectious Disease Product (QIDP) Designation for Its Lead Product Candidate, Omadacycline”. prnewsire.com. PR Newswire. January 3, 2013. Retrieved October 17, 2017.
- Jump up^ Seiffert, Don (2015). “Paratek presents new trial data for antibiotic as late-stage trials continue”. bizjournals.com. American City Business Journals. Retrieved October 17,2017.
- Jump up^ “Omadacycline Versus Linezolid for the Treatment of ABSSSI (EudraCT #2013-003644-23)”. clinicaltrials.gov. Retrieved 2015-10-13.
- Jump up^ “Paratek Announces that Omadacycline Met All Primary and Secondary Efficacy Outcomes Designated by FDA and EMA in a Phase 3 Study in Acute Bacterial Skin Infections; Omadacycline was Generally Safe and Well-Tolerated”. finance.yahoo.com. Retrieved 3 July 2016.
- Jump up^ “Omadacycline vs Moxifloxacin for the Treatment of CABP (EudraCT #2013-004071-13)”. clinicaltrials.gov. Retrieved 2015-10-13.
- Jump up^ “Paratek Announces Positive Phase 3 Study of Omadacycline in Community-Acquired Bacterial Pneumonia”. http://www.globenewswire.com. April 3, 2017. Retrieved 16 May 2017.
- Jump up^ “Paratek Initiates Phase 1b Study of Omadacycline in Urinary Tract Infection”. globenewswire.com. May 2, 2016. Retrieved 3 July 2016.
- Jump up^ “Paratek Initiates Phase 3 Study of Oral-only Omadacycline in ABSSSI”. globenewswire.com. August 15, 2016. Retrieved 15 August 2016.
- Jump up^ “Paratek Announces Phase 3 Study of Oral-Only Dosing of Omadacycline Met All Primary and Secondary FDA and EMA Efficacy Endpoints in Acute Bacterial Skin Infections”. http://www.globenewswire.com. July 17, 2017. Retrieved 19 July 2017.
- Jump up^ Ref: Mark L. Nelson and Kwasi Ohemeng: 4-dedimethylamino tetracycline compounds, United States (US) patent number 7,056,902 (2006)
| Clinical data | |
|---|---|
| Trade names | Nuzyra |
| Identifiers | |
| CAS Number | |
| PubChem CID | |
| ChemSpider | |
| UNII | |
| KEGG | |
| Chemical and physical data | |
| Formula | C29H40N4O7 |
| Molar mass | 556.66 g·mol−1 |
| 3D model (JSmol) | |
/////////////FDA 2018, Nuzyra, Omadacycline tosylate, Omadacycline, オマダサイクリントシル酸塩 ,PTK-0796, PTK 0796
CC1=CC=C(C=C1)S(O)(=O)=O.[H][C@@]12CC3=C(C=C(CNCC(C)(C)C)C(O)=C3C(=O)C1=C(O)[C@]1(O)C(=O)C(C(N)=O)=C(O)[C@@H](N(C)C)[C@]1([H])C2)N(C)C
Golvatinib, ゴルバチニブ
Golvatinib
E-7050, cas 928037-13-2
1-N’-[2-fluoro-4-[2-[[4-(4-methylpiperazin-1-yl)piperidine-1-carbonyl]amino]pyridin-4-yl]oxyphenyl]-1-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide
| Molecular Formula: | C33H37F2N7O4 |
|---|---|
| Molecular Weight: | 633.701 g/mol |
-
N’-[2-fluoro-4-[2-[[4-(4-methylpiperazin-1-yl)piperidine-1-carbonyl]amino]pyridin-4-yl]oxyphenyl]-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamideUNII:516Z3YP58E
-
Originator Eisai Co Ltd
- Class Amides; Antineoplastics; Cyclopropanes; Fluorobenzenes; Piperazines; Piperidines; Pyridines; Small molecules
- Mechanism of Action Angiogenesis inhibitors; Proto oncogene protein c met inhibitors; Vascular endothelial growth factor receptor-2 antagonists
- Discontinued Gastric cancer; Glioblastoma; Head and neck cancer; Liver cancer; Malignant melanoma; Solid tumours
- 15 Nov 2013Eisai completes enrolment in its phase Ib/II trial for Head and neck cancer (second-line combination therapy, late-stage disease) in USA, United Kingdom, South Korea & Ukraine (NCT01332266)
- 14 Nov 2013Phase-I/II clinical trials in liver cancer (first-line combination therapy, late-stage disease) in Italy & Ukraine (PO)
- 01 Jul 2013Eisai completes a phase I trial in Solid tumours in Japan (NCT01428141)
Golvatinib is an orally bioavailable dual kinase inhibitor of c-Met (hepatocyte growth factor receptor) and VEGFR-2 (vascular endothelial growth factor receptor-2) tyrosinekinases with potential antineoplastic activity. c-Met/VEGFR kinase inhibitor E7050 binds to and inhibits the activities of both c-Met and VEGFR-2, which may inhibit tumor cell growth and survival of tumor cells that overexpress these receptor tyrosine kinases. c-Met and VEGFR-2 are upregulated in a variety of tumor cell types and play important roles in tumor cell growth, migration and angiogenesis.
Method for producing a phenoxy pyridine derivative (3)
The present invention, hepatocyte growth factor receptor (Hepatocyte growth factor receptor; hereinafter, abbreviated as “HGFR”) inhibitory action, antitumor action, anti-tumor agents with such angiogenesis inhibitory activity and cancer metastasis inhibitory action, a cancer metastasis suppressing the method for producing a useful phenoxy pyridine derivatives as agents.
Patent Document 1 has a HGFR inhibitory activity, anti-tumor agents, useful phenoxy pyridine derivative as an angiogenesis inhibitor or cancer metastasis inhibitor has been disclosed.
(In the formula, R 1, .R 2 and R 3 means such as 3-10 membered non-aromatic heterocyclic group, .R 4, R 5, R 6 and R 7 which represents a hydrogen atom, same or different, a hydrogen atom, a halogen atom, .R 8 to mean a C 1-6 alkyl group, .R 9 to mean a hydrogen atom or the like is and 3-10 membered non-aromatic heterocyclic group meaning .n is .X to mean 1 to 2 integer, it refers to a group or a nitrogen atom represented by the formula -CH =.)
As a method for producing the phenoxy pyridine derivative, to the Example 48 of Patent Document 1, N, N-dimethylformamide, triethylamine and benzotriazol-1-yloxytris (dimethylamino) or lower in the presence of a phosphonium hexafluorophosphate discloses that perform the reaction.
Patent Document 2, as a manufacturing method suitable for industrial mass synthesis of the phenoxy pyridine derivative in the presence a condensing agent, production method of reacting an aniline derivative with a carboxylic acid derivative.
(In the formula, R 1, is .R 2, R 3, R 4 and R 5, which means such good azetidin-1-yl group which may have a substituent, the same or different and each represents a hydrogen atom or fluorine It refers to an atom .R 6 means a hydrogen atom or a fluorine atom.)
Patent Document 3, another manufacturing method of the phenoxy pyridine derivative, there is disclosed the manufacturing method shown in the following scheme.
(In the formula, R 1 means a 4- (4-methylpiperazin-1-yl) piperidin-1-yl group or a 3-hydroxy-1-yl group .R 2, R 3, R 4 and R 5 are the same or different, represents a hydrogen atom or a fluorine atom. However, among R 2, R 3, R 4 and R 5, 2 or 3 is a hydrogen atom .R 6 is a hydrogen atom or .R 7 to mean a fluorine atom, .Ar which means a protecting group for the amino group means a phenyl group.)
International Publication No. WO 2007/023768 International Publication No. WO 2008/026577 International Publication No. WO 2009/104520
[Formula
1 H-NMR Spectrum (DMSO-d 6) .Delta. (Ppm): 1.22-1.33 (2H, m), 1.54-1.63 (4H, m), 1.68-1.78 (2H, m), 2.12 (3H , S), 2.12-2.40 (5H, m), 2.40-2.60 (4H, m), 2.68-2.78 (2H, m), 4.06-4.14 (2H, t, J = 8 Hz), 7.22 (2H, m), 6.60 (1H, dd, J = 2.4 Hz, 5.6 Hz), 7.00 (1 H, dd, J = 2.4 Hz, 11.2 Hz), 7.40 (1 H, s), 7.61 (2 H, dd, J = 5.2 Hz, 8 Hz), 7.93 J = 8.8 Hz), 8.13 (1 H, d, J = 5.6 Hz), 9.21 (1 H, s), 9.90 (1 H, brs), 10.55 (1 H, brs).
///////////////Golvatinib, phase 2, ゴルバチニブ ,
CN1CCN(CC1)C2CCN(CC2)C(=O)NC3=NC=CC(=C3)OC4=CC(=C(C=C4)NC(=O)C5(CC5)C(=O)NC6=CC=C(C=C6)F)F
Savolitinib
CAS 1313725-88-0, Molecular Formula, C17-H15-N9, Molecular Weight, 345.3685
1H-1,2,3-Triazolo(4,5-b)pyrazine, 1-((1S)-1-imidazo(1,2-a)pyridin-6-ylethyl)-6-(1-methyl-1H-pyrazol-4-yl)-
- AZD-6094
- AZD6094
- HMPL-504
- HMPL504
- Savolitinib
- Savolitinib [INN]
- UNII-2A2DA6857R
- Volitinib
- HM 5016504
Hutchison China MediTech (Chi-Med), Cancer, kidney (renal cell carcinoma, papillary)
A c-Met kinase inhibitor with antineoplastic activity.
NCI: volitinib. An orally bioavailable inhibitor of the c-Met receptor tyrosine kinase with potential antineoplastic activity. Volitinib selectively binds to and inhibits the activation of c-Met in an ATP-competitive manner, and disrupts c-Met signal transduction pathways. This may result in cell growth inhibition in tumors that overexpress the c-Met protein. C-Met encodes the hepatocyte growth factor receptor tyrosine kinase and plays an important role in tumor cell proliferation, survival, invasion, and metastasis, and tumor angiogenesis; this protein is overexpressed or mutated in a variety of cancers.(NCI Thesaurus)
Savolitinib is an experimental small molecule inhibitor of c-Met. It is being investigated for the treatment of cancer by AstraZeneca.[1] It is in phase II clinical trials for adenocarcinoma, non-small cell lung cancer, and renal cell carcinoma.[2]
Savolitinib is a first-in-class inhibitor of c-Met in phase III clinical development at at Hutchison China MediTech (Chi-Med) and AstraZeneca for the treatment of patients with MET-driven, unresectable and locally advanced or metastatic papillary renal cell carcinoma. Phase II trials are also under way for the oral treatment of locally advanced or metastatic pulmonary sarcomatoid carcinoma. AstraZeneca is conducting phase II clinical trials for the treatment of non-small cell lung cancer. Phase I/II trials are ongoing at Samsung Medical Center for the second-line treatment of advanced gastric adenocarcinoma patients with MET amplification.
In 2011, the drug was licensed to AstraZeneca by at Hutchison China MediTech (Chi-Med) for worldwide codevelopment and marketing rights for the treatment of cancer.

SYNTHESIS

PAPER
Journal of Organic Chemistry (2018),
A multidisciplinary approach covering synthetic, physical, and analytical chemistry, high-throughput experimentation and experimental design, process engineering, and solid-state chemistry is used to develop a large-scale (kilomole) Suzuki–Miyaura process. Working against clear criteria and targets, a full process investigation and optimization package is described highlighting how and why key decisions are made in the development of large-scale pharmaceutical processes.
Process Design and Optimization in the Pharmaceutical Industry: A Suzuki–Miyaura Procedure for the Synthesis of Savolitinib








PAPER
Journal of Medicinal Chemistry (2014), 57(18), 7577-7589
HGF/c-Met signaling has been implicated in human cancers. Herein we describe the invention of a series of novel triazolopyrazine c-Met inhibitors. The structure–activity relationship of these compounds was investigated, leading to the identification of compound 28, which demonstrated favorable pharmacokinetic properties in mice and good antitumor activities in the human glioma xenograft model in athymic nude mice.
Discovery of (S)-1-(1-(Imidazo[1,2-a]pyridin-6-yl)ethyl)-6-(1-methyl-1H-pyrazol-4-yl)-1H-[1,2,3]triazolo[4,5-b]pyrazine (Volitinib) as a Highly Potent and Selective Mesenchymal–Epithelial Transition Factor (c-Met) Inhibitor in Clinical Development for Treatment of Cancer
Preparation of (S)-2-(4-(1-(1-(pyrazolo[1,5-a]pyridin-5-yl)ethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-pyrazol-1-yl)ethanol (30) and (R)-2-(4-(1-(1-(pyrazolo[1,5-a]pyridin-5-yl)ethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-pyrazol-1-yl)ethanol (31)
PATENT
WO 2018175251
WO 2018055029
WO 2018024608
WO 2016087680
WO 2016081773
PATENT
JP 2016069348
PATENT
The present invention provides a triazolopyrazine derivatives, the chemical name (S) -1- (l_ (imidazo [l, 2_a] pyrazin-6-yl) ethane-yl) -6-α _ -1H- pyrazol-4-yl-methyl) -1Η- [1,2,3] triazolo [4,5-b] pyrazine, of formula (I), the
[0005] This compound is an inhibitor of the activity c -Me t, may be used for treatment or prevention of inhibition of c -Me t sensitive cancers. In the Chinese patent CN 102906092A (W02011 / 079804), discloses the synthesis and use triazolopyrazine derivatives. Prepared by repeating the above patent, the compound powder obtained by detecting an amorphous state. As those skilled in the art, although amorphous higher solubility and dissolution rate than polymorph in most cases, but it is unstable, hygroscopic, readily converted to stable crystalline form.Thus, without the presence of processing stability and poor storage stability shaped, and in the production process, the smaller the bulk density of the particles of amorphous, high surface free energy, are likely to cause aggregation, poor flowability, and a series of powerful elastic deformation of the formulation problem seriously affecting the clinical value of amorphous Drug triazolopyrazine derivatives.
PATENT
PATENT
WO 2014174478
CN 102127096
PATENT
References
- ^ https://searchusan.ama-assn.org/usan/documentDownload?uri=%2Funstructured%2Fbinary%2Fusan%2Fsavolitinib.pdf
- ^ adisinsight.springer.com/drugs/800035893
| Clinical data | |
|---|---|
| Synonyms | Volitinib |
| Identifiers | |
| CAS Number | |
| ChemSpider | |
| KEGG | |
| Chemical and physical data | |
| Formula | C17H15N9 |
| Molar mass | 345.37 g·mol−1 |
| 3D model (JSmol) | |
///////////////Savolitinib, Phase III, AZD-6094, AZD6094, HMPL-504, HMPL504, UNII-2A2DA6857R, Volitinib, HM 5016504
C[C@@H](c1ccc2nccn2c1)n3c4c(ncc(n4)c5cnn(c5)C)nn3
In some embodiments, the c-Met inhibitor is ARQ197 (Tivantinib). Tivantinib has the IUPAC name (3R,4R)-3-(5,6-Dihydro-4H-pyrrolo[3,2,1-ij]quinolin-1-yl)-4-(1H-indol-3-yl)-2,5-pyrrolidinedione and the following chemical structure:
[0058] In some embodiments, the c-Met inhibitor is EMD1214063 (MSC2156119J; Tepotinib).
Tepotinib has the IUPAC name 3-(1-(3-(5-((1-methylpiperidin-4-yl)methoxy)pyrimidin-2-yl)benzyl)-1,6-dihydro-6-oxopyridazin-3-yl)benzonitrile and the following chemical structure:
[0059] In some embodiments, the c-Met inhibitor is GSK/1363089/XL880 (Foretinib). Foretinib has the IUPAC name N1’-[3-fluoro-4-[[6-methoxy-7-(3-morpholinopropoxy)-4-quinolyl]oxy]phenyl]-N1-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide and the following chemical structure:
[0060] In some embodiments, the c-Met inhibitor is XL184 (Cabozantinib). Cabozantinib has the IUPAC name N-(4-((6,7-Dimethoxyquinolin-4-yl)oxy)phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide and the following chemical structure:
[0061] In some embodiments, the c-Met inhibitor is HMPL-504/AZD6094/volitinib (Savolitinib). Volitinib has the IUPAC name (S)-1-(1-(imidazo[1,2-a]pyridin-6-yl)ethyl)-6-(1-methyl-1H-pyrazol-4-yl)-1H-[1,2,3]triazolo[4,5-b]pyrazine and the following chemical structure:
[0062] In some embodiments, the c-Met inhibitor is MSC2156119J (EMD 1214063, Tepotinib).
Tepotinib has the IUPAC name Benzonitrile, 3-[1,6-dihydro-1-[[3-[5-[(1-methyl-4-piperidinyl)methoxy]-2-pyrimidinyl]phenyl]methyl]-6-oxo-3-pyridazinyl]- and the following chemical structure:
[0063] In some embodiments, the c-Met inhibitor is LY2801653 (Merestinib). Merestinib has the IUPAC name N-(3-fluoro-4-{[1-methyl-6-(1H-pyrazol-4-yl)-1H-indazol-5 yl]oxy}phenyl)-1-(4-fluorophenyl)-6-methyl-2-oxo-1,2-dihydropyridine-3-carboxamide and the following chemical structure:
[0064] In some embodiments, the c-Met inhibitor is AMG 337. AMG 337 has the IUPAC name 7-methoxy-N-((6-(3-methylisothiazol-5-yl)-[1,2,4]triazolo[4,3-b]pyridazin-3-yl)methyl)-1,5-naphthyridin-4-amine and the following chemical structure:
[0065] In some embodiments, the c-Met inhibitor is INCB28060 (Capmatinib). Capmatinib has the IUPAC name 2-fluoro-N-methyl-4-[7-(quinolin-6-ylmethyl)imidazo[1,2-b][1,2,4]triazin-2-yl]benzamide and the following chemical structure:
[0066] In some embodiments, the c-Met inhibitor is AMG 458. AMG 458 has the IUPAC name 1-(2-hydroxy-2-methylpropyl)-N-(5-((7-methoxyquinolin-4-yl)oxy)pyridin-2-yl)-5-methyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide and the following chemical structure:
[0067] In some embodiments, the c-Met inhibitor is PF-04217903. PF-04217903 has the IUPAC name 2-(4-(1-(quinolin-6-ylmethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-pyrazol-1-yl)ethanol and the following chemical structure:
[0068] In some embodiments, the c-Met inhibitor is PF-02341066 (Crizotinib). Crizotinib has the IUPAC name (R)-3-(1-(2,6-dichloro-3-fluorophenyl)ethoxy)-5-(1-(piperidin-4-yl)-1H-pyrazol-4-yl)pyridin-2-amine and the following chemical structure:
[0069] In some embodiments, the c-Met inhibitor is E7050 (Golvatinib). Golvatinib has the IUPAC name N-(2-fluoro-4-((2-(4-(4-methylpiperazin-1-yl)piperidine-1-carboxamido)pyridin-4-yl)oxy)phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide and the following chemical structure:
[0070] In some embodiments, the c-Met inhibitor is MK-2461. MK-2461 has the IUPAC name N-((2R)-1,4-Dioxan-2-ylmethyl)-N-methyl-N’-[3-(1-methyl-1H-pyrazol-4-yl)-5-oxo-5H-benzo[4,5]cyclohepta[1,2-b]pyridin-7-yl]sulfamide and the following chemical structure:
[0071] In some embodiments, the c-Met inhibitor is BMS-777607. BMS-777607 has the IUPAC name N-(4-((2-amino-3-chloropyridin-4-yl)oxy)-3-fluorophenyl)-4-ethoxy-1-(4-fluorophenyl)-2-oxo-1,2-dihydropyridine-3-carboxamide and the following chemical structure:
[0072] In some embodiments, the c-Met inhibitor is JNJ-38877605. JNJ-38877605 has the IUPAC name 6-(difluoro(6-(1-methyl-1H-pyrazol-3-yl)-[1,2,4]triazolo[4,3-b]pyridazin-3-yl)methyl)quinoline and the following chemical structure:
Epitinib
Epitinib succinate; HMPL-813; Huposuan yipitini
1203902-67-3, 430.50, C24 H26 N6 O2
1-Piperazinecarboxamide, 4-ethyl-N-[4-[(3-ethynylphenyl)amino]-7-methoxy-6-quinazolinyl]-
4-Ethyl-N-[4-[(3-ethynylphenyl)amino]-7-methoxy-6-quinazolinyl]-1-piperazinecarboxamide
Cancer; Glioblastoma; Non-small-cell lung cancer
Epitinib is in phase I clinical trials by Hutchison MediPharma for the treatment of solid tumours.
Epitinib succinate is an oral EGFR tyrosine kinase inhibitor in early clinical development at Hutchison China MediTech (Chi-Med) for the treatment of solid tumors and the treatment of glioblastoma patients with EGFR gene amplification.
- Originator Hutchison MediPharma
- Class Antineoplastics; Small molecules
- Mechanism of Action Epidermal growth factor receptor antagonists
- Phase I/II Glioblastoma; Non-small cell lung cancer
- No development reported Oesophageal cancer; Solid tumours
- 28 May 2018 No recent reports of development identified for preclinical development in Oesophageal-cancer in China (PO)
- 06 Mar 2018 Hutchison Medipharma plans a phase III pivotal study for Non-small cell lung cancer (NSCLC) patients with brain metastasis in China in 2018
- 06 Mar 2018 Phase-I/II clinical trials in Glioblastoma (Second-line therapy or greater) in China (PO)

PATENT

////////////Epitinib , PHASE 1, PHASE 2, Epitinib succinate, HMPL-813, Huposuan yipitini, 1203902-67-3,
Eluxadoline, エルクサドリン ,элуксадолин ,إيلوكسادولين ,艾沙多林 ,
![]()
Eluxadoline
- Molecular FormulaC32H35N5O5
- Average mass569.651 Da
5-({[(2S)-2-amino-3-(4-carbamoyl-2,6-dimethylphenyl)propanoyl][(1S)-1-(4-phenyl-1H-imidazol-2-yl)ethyl]amino}methyl)-2-methoxybenzoic acid
5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid
864821-90-9 CAS
JNJ-27018966
Molecular Formula: C32H35N5O5
Molecular Weight: 569.6508
Agents for Irritable Bowel Syndrome, mu-Opioid Agonists, delta-Opioid Antagonists
Eluxadoline
Trade Name: Viberzi®
Research Code: JNJ-27018966, JNJ27018966, JNJ 27018966
Chemical Name: 5 – [[[(2S) -2-amino-3- [4- (aminocarbonyl) -2,6-dimethylphenyl ] -1- oxopropyl] [(1S) -1- (4-phenyl-1H-imidazol-2-yl) ethyl] amino] methyl] -2-methoxybenzoic acid
MOA: mu opioid receptor agonist, Indication: Irritable bowel syndrome with diarrhea (IBS-D)
Approval Date: May 27, 2015 (US)
Originator: Furiex Pharmaceuticals Inc ( Furiex acquired Eluxadoline from Janssen in 2011 )
Developer: Forest Laboratories Inc. (acquired by Actavis PLC in 2014 )
Eluxadoline, sold under the brand names Viberzi (/vaɪˈbɜːrzi/ vy-BUR-zee) in the US and Truberzi in Europe,[2] is a medication taken by mouth for the treatment of diarrhea and abdominal pain in individuals with diarrhea-predominant irritable bowel syndrome (IBS-D).[3]It was approved for use in the United States in 2015.[4] The drug originated from Janssen Pharmaceutica and was developed by Actavis.
Contraindications
This drug is contraindicated in case of having:
- Blockage of the gallbladder or a sphincter of Oddi problem
- Problems with alcohol abuse
- Pancreatitis
- Liver problems
- Chronic or severe constipation[5]
Adverse effects
Common adverse effects are constipation and nausea, but rates of discontinuation due to constipation were low for both eluxadoline and placebo. Rare adverse effects: fatigue, bronchitis, viral gastroenteritis. Rare serious adverse effects include pancreatitis with a general incidence of 0.3% – higher incidence with 100 mg dose (0.3%) than with 75 mg dose (0.2%).[6] The risk is even greater in those who do not have a gall bladder and the medication is not recommended in this group.[7]
In March 2017, the U.S. Food and Drug Administration issued a safety alert for eluxadoline concerning an increased risk of serious pancreatitis in patients without a gallbladder.[8] An FDA review found that in such patients, spasm of the sphincter of Oddi may lead to severe pancreatitis.[9] The FDA reported that in some cases symptoms have occurred with just one or two doses at the recommended dosage for patients without a gallbladder (75 mg).[9] Of two deaths associated with eluxadoline reported up to February 2017, both occurred in patients without a gallbladder.[8]
Interactions
Elevated concentrations of eluxadoline were observed with co-administration of inhibitors of the transporter protein OATP1B1, such as:
Also, concurrent use of other drugs that cause constipation is not preferred, such as:
- Opioids
- Alosetron
- Anticholinergics
- Bismuth subsalicylate, potentially dangerous synergism.[10]
Eluxadoline increases the concentrations of drugs which are OATP1B1 and BCRP substrates. Also, co-administration of eluxadoline with rosuvastatin may increase the risk of rhabdomyolysis.[1]
Pharmacology
Mechanism of action
Eluxadoline is a μ- and κ-opioid receptor agonist and δ-opioid receptor antagonist [11] that acts locally in the enteric nervous system, possibly decreasing adverse effects on the central nervous system.[12][13]
Pharmacokinetics
In the in vitro studies, eluxadoline was found to be transported by OAT3 (SLC22A8), OATP1B1 (SLCO1B1) and BSEP (ABCB11) at the highest concentrations tested (400 ng/ml which is 162-fold larger than the observed Cmax of the highest therapeutic dose of 100 mg). However, it was not to be transported by OCT1 POU2F1, OAT1 Organic anion transporter 1, OCT2, OATP1B3 (SLCO1B3), P-gp (P-glycoprotein), or BCRP (ABCG2).
Multidrug resistance-associated protein 2 (MRP2)-vesicular accumulation of eluxadoline was observed, indicating that the drug is a substrate of MRP2. Eluxadoline was not found to inhibit BCRP-, BSEP-, MRP2-, OCT1-, OCT2-, OAT1-, OAT3-, or OATP1B3-mediated transport of probe substrates but inhibited the transport of probe substrates of OATP1B1 and P-gp. Also in the in vitro studies, it was observed that eluxadoline is an in vivo substrate of OATP1B1, OAT3, and MRP2. Finally, no inhibition or induction of cytochrome P450enzymes was observed.[14]
Following a 100 mg dose of eluxadoline, the Cmax was about 2 to 4 ng/ml and AUC was 12-22 ng.h/ml. Eluxadoline has linear pharmacokinetics with no accumulation upon repeated twice daily dosing. Taking eluxadoline with high fat meal decreased the Cmax by 50% and AUC by 60%.[1]
Chemistry
Synthesis
The synthesis of eluxadoline was extensively discussed in the patent No. WO2006099060 A2, with the title : “Process for the preparation of opioid modulators” which was published in Sept. 2006[15]
A CLIP
5 JAN 2014
Furiex Pharmaceuticals Inc. more than doubled in its best day of trading after its experimental drug alleviated diarrhea and abdominal pain caused by irritable bowel syndrome in two studies.
The drug eluxadoline met targets for improvements in stool consistency and abdominal pain that were developed in conjunction with U.S. and European regulators, the company said today. Furiex will apply for approval in June, Chairman Fred Eshelman said in an investor call today. He estimated annual sales of $750 million to $1 billion.
“By our math, it looks like a pretty doggone good market,” Eshelman said on the call, noting that there is only one currently approved drug available in the U.S. for the condition.
Diarrhea-predominant irritable bowel syndrome is a chronic disorder that affects about 28 million patients in the U.S. and Europe, Furiex said in the statement.Furiex said it would apply by mid-year for U.S. approval of the drug, eluxadoline, to treat diarrhea-predominant irritable bowel syndrome (IBS-d), a debilitating bowel disorder that affects about 28 million people in the United States and major European markets.
Furiex said it expected to seek European approval in early 2015.
“We believe that there are a lot of patients out there who need this drug. There is a huge unmet need,” Furiex Chief Medical Officer June Almenoff said in a telephone interview.
Currently approved drugs for IBS address constipation associated with the disorder, but there are few options for diarrhea predominant IBS.
Furiex founder and chairman Fred Eshelman said he believes the drug has the potential for blockbuster sales, which he defined as annual sales of between $750 million and $1 billion.
Eluxadoline was tested at two doses against a placebo over the course of 12 weeks to meet requirements by the U.S. Food and Drug Administration, and for 26 weeks for European health regulators, in Phase III studies involving 2,428 patients, Furiex said.
For the combined goal of improvement in abdominal pain and stool consistency for at least half the days in the study, eluxadoline achieved a statistically significant improvement at the 100 milligram and 75 mg doses through 12 weeks in both studies.
On the 26-week measure, the higher dose succeeded in both studies but the lower dose missed statistical significance in one of the two trials, according to initial results released by the company.
The success appeared to be driven by the percentage of patients reporting improvements in diarrhea, which ranged from 30 percent to 37 percent versus 22 percent and 20.9 percent for the placebo groups.
When the composite goal was broken into its two components, researchers found a numerical improvement in pain response rates that did not achieve statistical significance.
The drug appeared to be safe and well-tolerated in both studies, Furiex said. The most commonly reported side effects were constipation and nausea.
The company plans to present a far more detailed analysis of the late stage studies at an upcoming medical meeting.
“We’re very excited about the path ahead and about how this can transform patients’ lives,” Almenoff said.
Mu Delta is a locally active mu opioid receptor agonist and delta opioid receptor antagonist in phase III clinical evaluation at Furiex Pharmaceuticals for the oral treatment of diarrheal predominant irritable bowel syndrome (d-IBS).
The product candidate holds an advantage over currently marketed products for this indication because it acts locally on the enteric nervous system, possibly decreasing adverse effects on the central nervous system. In 2011, fast track designation was assigned in the U.S. for the treatment of d-IBS. In 2011, Mu Delta was licensed to Furiex Pharmaceuticals by Janssen for the treatment of d-IBS, granting an option to Furiex to continue development and commercialization following phase II proof of concept studies.
The opioid receptors were identified in the mid-1970’s, and were quickly categorized into three sub-sets of receptors (mu, delta and kappa). More recently the original three types of receptors have been further divided into sub-types. Also known is that the family of opioid receptors are members of the G-protein coupled receptor (GPCR) super-family. More physiologically pertinent are the well established facts that opioid receptors are found throughout the central and peripheral nervous system of many mammalian species, including humans, and that modulation of the respective receptors can elicit numerous, albeit different, biological effects, both desirable and undesirable (D. S. Fries, “Analgesics”, inPrinciples of Medicinal Chemistry, 4th ed.; W. O. Foye, T. L. Lemke, and D. A. Williams, Eds.; Williams and Wilkins: Baltimore, Md., 1995; pp. 247-269; J. V. Aldrich, “Analgesics”, Burger’s Medicinal Chemistry and Drug Discovery, 5thEdition, Volume 3: Therapeutic Agents, John Wiley & Sons, Inc., 1996, pp. 321-441). In the most current literature, the likelihood of heterodimerization of the sub-classes of opioid receptors has been reported, with respective physiological responses yet undetermined (Pierre J. M. Riviere and Jean-Louis Junien, “Opioid receptors: Targets for new gastrointestinal drug development”, Drug Development 2000, pp. 203-238).
A couple biological effects identified for opioid modulators have led to many useful medicinal agents. Most significant are the many centrally acting mu opioid agonist modulators marketed as analgesic agents to attenuate pain (e.g., morphine), as well as peripherally acting mu agonists to regulate motility (e.g., loperamide). Currently, clinical studies are continuing to evaluate medicinal utility of selective delta, mu, and kappa modulators, as well as compounds possessing combined sub-type modulation. It is envisioned such explorations may lead to agents with new utilities, or agents with minimized adverse side effects relative to currently available agents (examples of side effects for morphine includes constipation, respiratory depression, and addiction potential). Some new GI areas where selective or mixed opioid modulators are currently being evaluated includes potential treatment for various diarrheic syndromes, motility disorders (post-operative ileus, constipation), and visceral pain (post operative pain, irritable bowel syndrome, and inflammatory bowel disorders) (Pierre J. M. Riviere and Jean-Louis Junien, “Opioid receptors: Targets for new gastrointestinal drug development” Drug Development, 2000, pp. 203-238).
Around the same time the opioid receptors were identified, the enkephalins were identified as a set of endogenous opioid ligands (D. S. Fries, “Analgesics”, inPrinciples of Medicinal Chemistry, 4th ed.; W. O. Foye; T. L. Lemke, and D. A. Williams, Eds.; Williams and Wilkins: Baltimore, Md., 1995; pp. 247-269). Schiller discovered that truncating the original pentapeptide enkephalins to simplified dipeptides yielded a series of compounds that maintained opioid activity (Schiller, P. WO 96/06855). However one potential drawback cited for such compounds is the likelihood of their inherent instability (P. W. Schiller et al., Int. J. Pept. Protein Res. 1993, 41 (3), pp. 313-316).
More recently, a series of opioid pseudopeptides containing heteroaromatic or heteroaliphatic nuclei were disclosed, however this series is reported showing a different functional profile than that described in the Schiller works. (L. H. Lazarus et al., Peptides 2000, 21, pp. 1663-1671).
Most recently, works around morphine related structures were reported by Wentland, et al, where carboxamido morphine derivatives and it’s analogs were prepared (M. P. Wentland et al., Biorg. Med. Chem. Letters 2001, 11, pp. 1717-1721; M. P. Wentland et al., Biorg. Med. Chem. Letters 2001, 11, pp. 623-626). Wentland found that substitution for the phenol moiety of the morphine related structures with a primary carboxamide led anywhere from equal activities up to 40 fold reduced activities, depending on the opioid receptor and the carboxamide. It was also revealed that any additional N-substitutions on the carboxamide significantly diminished the desired binding activity.
Compounds of the present invention have not been previously disclosed and are believed to provide advantages over related compounds by providing improved pharmacological profiles.
Opioid receptor modulators, agonists or antagonists are useful in the treatment and prevention of various mammalian disease states, for example pain and gastrointestinal disorders such as diarrheic syndromes, motility disorders including post-operative ileus and constipation, and visceral pain including post-operative pain, irritable bowel syndrome and inflammatory bowel disorders.
It is an object of the present invention to provide opioid receptor modulators. It is a further object of the invention to provide opioid receptor agonists and opioid receptor antagonists. It is an object of the present invention to provide opioid receptor ligands that are selective for each type of opioid receptor, mu, delta and kappa. It is a further object of the present invention to provide opioid receptor ligands that modulate two or three opioid receptor types, mu, delta and kappa, simultaneously.
It is an object of the invention to provide certain instant compounds that are also useful as intermediates in preparing new opioid receptor modulators. It is also an object of the invention to provide a method of treating or ameliorating a condition mediated by an opioid receptor. And, it is an object of the invention to provide a useful pharmaceutical composition comprising a compound of the present invention useful as an opioid receptor modulator.
5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1 h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid is an opoid receptor modulator (mu receptor agonist and delta receptor antagonist) and may be useful for treating irritable bowel syndrome, pain or other opioid receptor disorders.
5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid and methods of making this molecule are disclosed in
US application 2005/02033143. Example 9 of US application 2005/02033143 makes the hydrochloride salt of 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid.
Applicants have discovered a process of making the zwitterion of 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid and two novel crystals of this zwitterion. In Applicant’s hands, these novel crystals provide improved properties and can be purified at higher purity. Applicant’s new process results in improved and less costly process manufacturing conditions than the procedure disclosed in US application 2005/02033143.
FIG. 6 is the molecular structure of the zwitterion 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1h-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid.

SYNTHESIS OF 5-formyl-2- methoxy-benzoic acid methyl ester
Example 8: 2-Methoxy-5-formylbenzoic acid
Lithium hydroxide (1.04g, 0.043mol, 3eq) in water (lOmL) was added to a stirred solution of methyl 2-methoxy-5-formylbenzoate (2.8g, 0.014mol, leq) in a mixture of tetrahydrofuran (30mL) and methanol (20mL). The solution was stirred overnight, acidified to pH 1 with 10% HCl and the organic solvents removed in vacuo. The aqueous solution was extracted with ethyl acetate (lOOmL) and the organic solution washed with brine (lOOmL), then extracted with saturated aqueous sodium bicarbonate (3 x lOOmL). The basic solution was washed with ethyl acetate (lOOmL), then acidified to pH 1 with 10% HCl and back extracted with dichloromethane (3 x lOOmL). The organic solution was dried over sodium sulfate and evaporated in vacuo to give a cream coloured powder (2.01g, 77%). 1H NMR (CDC13) δ 9.99 (s, IH, O=C- H), 4.14 (s, 3H, CH3).
ANALOGOUS METHOD TO PREPARE..2-methoxy-5-{[1 -(4-phenyl-1 H-imidazol-2-yl)- ethylamino]-methyl}-benzoic acid methyl ester

USE 5-formyl-2- methoxy-benzoic acid methyl ester for 3,4- dimethoxybenzaldehyde, TO GET 2-methoxy-5-{[1 -(4-phenyl-1 H-imidazol-2-yl)- ethylamino]-methyl}-benzoic acid methyl ester
Example 4
(3,4-Dimethoxy-benzyl)-[1-(4-phenyl-1 H-imidazol-2-yl)-ethyl]-amine
A solution of 1-(4-phenyl-1 W-imidazol-2-yl)-ethylamine (0.061 g, 0.33 mmol) of Example 3, and 0.55 g (0.33 mmol) of 3,4-dimethoxybenzaldehyde in 5 ml_ of anhydrous methanol was stirred at room temperature for 1 h and then cooled to about 0-100C in an ice bath for 1 h. The reaction was treated carefully with 0.019 g (0.49 mmol) of sodium borohydride in one portion and maintained at about 0-100C for 21 h. Cold 2M aqueous HCI was added dropwise (30 drops), the mixture was stirred for 5 min, and then partially concentrated in vacuo unheated. The residual material was taken up in EtOAc to yield a suspension that was treated with 5 ml_ of cold 3M aqueous NaOH and stirred vigorously until clear. The phases were separated and the aqueous layer was extracted three times additional with EtOAc. The combined extracts were dried over MgSO4, filtered, and concentrated to yield (3,4-dimethoxy- benzyl)-[1-(4-phenyl-1 H-imidazol-2-yl)-ethyl]-amine as a light yellow oil (HPLC: 87% @ 254nm and 66% @ 214 nm).
MS (ES+) (relative intensity): 338.1 (100) (M+1)
This sample was of sufficient quality to use in the next reaction without further purification.
SYNTHESIS
In an embodiment, the present invention is directed to processes for the preparation of the compound of formula (IV)
also known as, 5-({[2-amino-3-(4-carbamoyl-2,5-dimethyl-phenyl)- propionyl]-[1 -(4-phenyl-1 H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy- benzoic acid
Example 1
(S)-2-ferf-Butoxycarbonylamino-3-(4-carbamoyl-2.6-dimethyl-phenyl)- propionic acid
STEP A: Trifluoromethanesulfonic acid 4-bromo-3,5-dimethyl-phenyl ester
To a cooled (0°C) solution of 4-bromo-3,5-dimethylphenol (3.05 g, 15.2 mmol) in pyridine (8 ml_) was added trifluoromethanesulfonic anhydride (5.0 g, 17.7 mmol) dropwise. After completion of addition, the resulting mixture was stirred at 0°C for 15 min, and then at room temperature overnight. The reaction was quenched by addition of water, and then extracted with EtOAc. The organic extracts were washed sequentially with water, 2N HCI (2x ), brine, and then dried over MgSO4. Filtration and evaporation to dryness yielded compound 1 b as a colorless oil.
1H NMR (300 MHz, CDCI3): δ 2.45 (6H, s), 7.00 (2H, s).
Step B: 4-Bromo-3,5-dimethylbenzoic acid
Into a solution of compound 1 b (6.57 g, 19.7 mmol) in DMF (65 ml_) were added K2CO3 (13.1 g, 94.7 mmol), Pd(OAc)2 (0.44 g, 1.97 mmol) and 1 ,1′-bis(diphenylphosphino)ferrocene (2.29 g, 4.14 mmol). The resulting mixture was bubbled in gaseous CO for 10 min and was heated to 60°C for 7.5h with a CO(9) balloon. The cooled mixture was partitioned between aqueous NaHCO3 and EtOAc, and filtered. The aqueous phase was separated, acidified with aqueous 6N HCI, extracted with EtOAc, and then dried over Na2SO4. Filtration and concentration of the filtrate yielded crude compound 1c as a brown residue, which was used in the next step without further purification. STEP C: Method A: 4-Bromo-3,5-dimethyl-benzamide
Into a suspension of compound 1c in DCM (40 ml_) was added SOCI2 (3.1 rnL, 42 mmol) and the mixture was heated at reflux for 2 h. Upon removal of the solvent by evaporation, the residue was dissolved in DCM (40 ml_) and then ammonium hydroxide (28% NH3 in water, 2.8 ml_) was added. The reaction mixture was heated at 5O0C for 2 h and concentrated. The residue was diluted with H2O, extracted with EtOAc, and the organic portion was dried over Na2SO4. After filtration and evaporation, the residue was purified by flash column chramotagraphy (eluent: EtOAc) to yield compound 1 d as an off-white solid.
1H NMR (300 MHz, CD3CN): δ 2.45 (6H, s), 5.94 (1 H, br s), 6.71 (1 H, br s), 7.57 (2H, s)
MS(ES+)(relative intensity): 228.0 (100%) (M+1).
Step C: Method B: 4-Bromo-3,5-dimethyl-benzamide
A mixture of compound 1 b (3.33 g, 10 mmol), PdCI2 (0.053 g, 0.3 mmol), hexamethyldisilazane (HMDS, 8.4 ml_, 40 mmol), and DPPP (0.12 g, 0.3 mmol) was bubbled with a gaseous CO for 5 min and then stirred in a CO balloon at 80°C for 4 h. To the reaction mixture was added MeOH (5 ml_). The reaction mixture was stirred for 10 min, diluted with 2N H2SO4 (200 ml_), and then extracted with EtOAc. The EtOAc extract was washed with saturated aqueous NaHCO3, brine, and then dried over Na2SO4. Filtration and evaporation of the resultant filtrate yielded a residue, which was purified by flash column chromatography (eluent: EtOAc) to yield compound 1d as a white solid.
Step D: 2-terf-Butoxycarbonylaminoacrylic acid methyl ester
To a suspension of /V-Boc-serine methyl ester (Compound 1e, 2.19 g, 10 mmol) and EDCI (2.01 g, 10.5 mmol) in DCM (70 ml_) was added CuCI (1.04 g, 10.5 mmol). The reaction mixture was stirred at room temperature for 72 h. Upon removal of the solvent, the residue was diluted with EtOAc, washed sequentially with water and brine and then dried over MgSO4. The crude product was purified by flash column chromatography (eluent: EtOAc:hexane ~1 :4) to yield compound 1f as a colorless oil.
1H NMR (300 MHz, CDCI3): δ 1.49 (9H, s), 3.83 (3H, s), 5.73 (1 H, d, J = 1.5 Hz), 6.16 (1 H1 S), 7.02 (1 H, s).
STEP E: (2)-2-fert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)acrylic acid methyl ester
A flask charged with compound 1d (0.46 g, 2.0 mmol), compound 1f (0.80 g, 4.0 mmol), tri-o-tolylphosphine (0.098 g, 0.32 mmol) and DMF (8 ml_) was purged with N2(g) 3 times. After the addition of tris(dibenzylideneacetone)dipalladium (0) (0.074 g, 0.08 mmol) and TEA (0.31 ml_, 2.2 mol), the reaction mixture was heated at 110°C for 24 h. At that time, the reaction was quenched by addition of water, and then extracted with EtOAc. The organic phase was washed with 1 N HCI, saturated aqueous NaHCO3, brine, and dried over MgSO4. The mixture was concentrated to a residue, which was purified by flash column chromatography (eluent: EtOAc:hexane~1 :1 to EtOAc only) to yield compound 1g as a white solid.
1H NMR (300 MHz, CD3OD): δ 1.36 (9H, s), 2.26 (6H, s), 3.83 (3H, s), 7.10 (1 H, s), 7.56 (2H, s); 13C NMR (75 MHz, DMSO-d6): δ 17.6, 25.7, 50.2, 78.7, 124.9, 126.4,
128.3, 131.2, 135.2, 135.5, 152.8, 164.3, 169.6;
MS (ES+) (relative intensity): 349.1 (38%)(M+1).
STEP F: (S)-2-ferf-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)propionic acid methyl ester
Into a reactor charged with a solution of compound 1g (0.56 g, 1.6 mmol) in degassed MeOH (80 mL) was added [Rh(COd)(H1R-DIPAMP)J+BF4 “ under a stream of argon. The reactor was sealed and flushed with H2, stirred at 6O0C under 1000 psi of H2 for 14 days. The crude product was purified by flash column chromatography (eluent: EtOAc:hexane ~1 :1) to yield compound 1 h as a white solid. ee: >99%; 1H NMR (300 MHz, CDCI3): δ 1.36 (9H, s), 2.39 (6H, s), 3.11 (2H, J = 7.2 Hz), 3.65 (3H, s), 4.53-4.56 (1 H, m), 5.12 (1 H, d, J = 8.7 Hz), 5.65 (1 H, br s), 6.09 (1 H, br s), 7.46 (2H, s);
MS(ES+) (relative intensity): 250.9 (100) (M-BoC)+.
STEP G: (S)-2-tert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)propionic acid
Into an ice-cooled solution of compound “I h (0.22 g, 0.63 mmol) in THF (3.5 ml_) was added an aqueous LiOH solution (1 N, 3.5 ml_) and the reaction mixture stirred at 0°C. Upon completion of the reaction, the reaction mixture was concentrated and the aqueous phase was neutralized with cooled aqueous 1 N HCI at 0°C, and then extracted with EtOAc. The combined extracts were dried over Na2SO4 overnight. Filtration and evaporation of the filtrate to dryness yielded compound 1j as a white solid. 1H NMR (300 MHz, DMSO-cfe): δ 1.30 (9H, s), 2.32 (6H, s), 2.95(1 H, dd,
J= 8.8, 13.9 Hz), 3.10 (1 H, dd, J= 6.2, 14.0 Hz), 4.02-4.12 (1 H, m), 7.18-7.23 (2H, m), 7.48 (2H1 s), 7.80 (1 H, s);
MS(ES+) (relative intensity): 236.9 (6) (M-BoC)+.
Example 5
5-((r2-Amino-3-(4-carbamoyl-2.6-dimethyl-phenyl)-propionvn-n-(4-phenyl- 1 H-imidazol-2-yl)-ethvπ-aminol-methyl)-2-methoxy-benzoic acid
STEP A. 2-Methoxy-5-{[1-(4-phenyl-1 W-imidazol-2-yl)-ethylamino]-methyl}- benzoic acid methyl ester
Using the procedures described for Example 4, substituting 5-formyl-2- methoxy-benzoic acid methyl ester (WO 02/22612) for 3,4- dimethoxybenzaldehyde, 2-methoxy-5-{[1 -(4-phenyl-1 H-imidazol-2-yl)- ethylamino]-methyl}-benzoic acid methyl ester was prepared.
STEP B. 5-({[2-ferf-ButoxycarbonylmethyI-3-(4-carbamoyl-2,6-dimethyl- phenyl)-propionyl]-[1 -(4-phenyl-1 H-imidazoI-2-yl)-ethyl]-amino}-methyl)-2- methoxy-benzoic acid methyl ester
Using the procedure of Example 3 for the conversion of Cpd 3d to Cpd 3e, substituting 2-methoxy-5-{[1-(4-phenyl-1 /-/-imidazol-2-yl)-ethylamino]- methylj-benzoic acid methyl ester for Cpd 3d and substituting 2-tert- Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionic acid for 2- tø/t-Butoxycarbonylamino-3-(4-hydroxy-2,6-dimethyl-phenyl)-propionic acid, Cpd 5a was prepared.
STEP C. 5-({[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)-propionyl]-[1 -(4-phenyl-1 W-imidazol-2-yl)-ethyl]-amino}-methyl)-2- methoxy-benzoic acid
5-({[2-tørf-Butoxycarbonylmethyl-3-(4-carbamoyl-2,6-dimethyl-phenyl)- propionyl]-[1-(4-phenyl-1 H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy- benzoic acid methyl ester was dissolved in an ice-chilled (0-10°C), mixed solvent system of THF (10 ml_) and MeOH (5 ml_). A LiOH H2O/water suspension (2.48 M; 3.77 ml_) was added dropwise, then the reaction was allowed to warm to room temperature and stirred overnight. The resulting mixture was cooled in an ice bath and the basic solution was neutralized with 2N citric acid until slightly acidic. The mixture was concentrated under reduced pressure to remove the volatile materials, after which time the remaining aqueous phase was extracted with EtOAc (3 x 26 ml_). These combined organic phases were dried over MgSO4, filtered, and concentrated under reduced pressure to yield a pale yellowish white solid. This crude material was dissolved in a 10% MeOH/CH2CI2 solution and adsorbed onto 30 g of silica. The adsorbed material was divided and chromatographed on an ISCO normal phase column over two runs, using a 40 g Redi-Sep column for both runs. The solvent system was a gradient MeOHZCH2CI2 system as follows: Initial 100% CH2CI2, 98%-92% over 40 min; 90% over 12 min, and then 88% over 13 min. The desired product eluted cleanly between 44-61 min. The desired fractions were combined and concentrated under reduced pressure to yield 5-({[2-terf- butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4- phenyl-1 /-/-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid, Cpd 5b, as a white solid.
STEP D. 5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1 – (4-phenyl-1 W-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid
A portion of Cpd 5b (0.27g, 0.41 mmol) was dissolved in EtOAc (39 ml_)/THF (5 ml_), filtered, and subsequently treated with gaseous HCI for 15 min. After completion of the HCI addition, the reaction was slowly warmed to room temperature and a solid precipitate formed. After 5 h the reaction appeared >97% complete by LC (@214nm; 2.56 min.). The stirring was continued over 3 d, then the solid was collected and rinsed with a small amount of EtOAc. The resulting solid was dried under high vacuum under refluxing toluene for 2.5 h to yield Cpd 5c as a white solid di-HCI salt.
Example 2
Racemic 2-terf-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethvl- phenvD-propionic acid
STEP A: Racemic 2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6- dimethyl-phenyl)propionic acid methyl ester
To a reactor charged with a solution of compound 1g (0.68 g, 1.95 mmol) in MeOH (80 mL) was added 10% Pd-C (0.5 g). The reactor was connected to a hydrogenator and shaken under 51 psi of H2 overnight. The mixture was filtered through a pad of Celite and the filtrate was concentrated to dryness to yield compound 2a as a white solid.
The 1H NMR spectrum was identical to that of (S)-2-tert- butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)propionic acid methyl ester, compound 1 h.
STEP B: Racemic 2-terf-butoxycarbonylamino-3-(4-carbamoyl-2,6- dimethyl-phenyl)propionic acid
Following the procedure described for Example 1 , STEP G (preparation of (S)-2-teAt-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)propionic acid), compound 2b – racemic 2-te/?-butoxycarbonylamino-3- (4-carbamoyl-2,6-dimethyl-phenyl)propionic acid – was prepared.
POLYMORPHS
Example 1 Preparation of the zwitterion of 5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid
A 1 L three-necked round-bottomed flask equipped with a mechanical stirrer, addition funnel and a thermocouple was charged without agitation. 34.2 g of 5-({[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid (see Example 9 of US 2005/0203143), 340 ml of acetone, and 17 ml of 204 mmolar concentrated HCl were combined in the flask. The stirring was started and the resulting slurry formed a clear solution. This solution was heated to 45° C. under vigorous stirring and aged at this temperature for a period of two hours. After the completion, the reaction mass was cooled to ambient temperature and the supernatant was removed by suction. The vessel along with the residue was rinsed with 20 ml of acetone and then removed as previously. 170 ml of water was added and the reaction mass and was aged under stirring until a homogeneus solution resulted. This solution was then added over a period of ˜½ hr to a solution of 90 ml of 1N NaOH and water. The pH was adjusted to 6.5-7.0 accordingly. The resulting slurry was aged for about 2 hrs at ambient temperature, cooled to 10-15° C., aged at that temperature for about 1 hr, and then filtered. The solid was washed with 10 ml water, air-dried for a period of 4 to 5 hrs, and then placed in a vacuum oven at 50-55° C. until the water content was less than 3%.
Example 2 Preparation of the Form α Crystal
The Form α crystal can be prepared by storing the zwitterion of 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid at 0-25% relative humidity for 3 days. Representative PXRD, TGA, and DSC data are shown in FIGS. 1-3 respectively.
Example 3 Preparation of the Form β crystal
The Form β crystal can be prepared by storing the zwitterion of 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid at greater than 60% relative humidity for 3 days. Representative PXRD, TGA, and DSC data are shown in FIGS. 1, 4, and 5 respectively.
SYNTHESIS
Example 9 5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid
A. 2-Methoxy-5{[1-(4-phenyl-1 H-imidazol-2-yl)-ethylamino]-methyl}-benzoic acid methyl ester.
Using the procedures described for Example 3, substituting 5-formyl-2-methoxy-benzoic acid methyl ester (WO 02/22612) for 3,4-dimethoxybenzaldehyde, 2-methoxy-5-{[1-(4-phenyl-1H-imidazol-2-yl)-ethylamino]-methyl}-benzoic acid methyl ester was prepared.
B. 5-({[2-tert-Butoxycarbonyl methyl-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid methyl ester.
Using the procedure of Example 1 for the conversion of Cpd 1d to Cpd 1e, substituting 2-methoxy-5-{[1-(4-phenyl-1H-imidazol-2-yl)-ethylamino]-methyl}-benzoic acid methyl ester for Cpd 1 d and substituting 2-tert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl-propionic acid of Example 8 for 2-tert-Butoxycarbonylamino-3-(4-hydroxy-2,6-dimethyl-phenyl)-propionic acid, Cpd 9a was prepared.
C. 5-({[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[11-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid.
5-({[2-tert-Butoxycarbonyl methyl-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid methyl ester was dissolved in an ice-chilled (0-10° C.), mixed solvent system of THF (10 mL) and MeOH (5 mL). A LiOH.H2O/water suspension (2.48 M; 3.77 mL) was added dropwise, then the reaction was allowed to warm to room temperature and stirred overnight. The resulting mixture was cooled in an ice bath and the basic solution was neutralized with 2N citric acid until slightly acidic. The mixture was concentrated under reduced pressure to remove the volatile materials, after which time the remaining aqueous phase was extracted with EtOAc (3×26 mL). These combined organic phases were dried over MgSO4, filtered, and concentrated under reduced pressure to give 2.26 g (146% of theory) of pale yellowish white solid. This crude material was dissolved in a 10% MeOH/CH2Cl2 solution and adsorbed onto 30 g of silica. The adsorbed material was divided and chromatographed on an ISCO normal phase column over two runs, using a 40 g Redi-Sep column for both runs. The solvent system was a gradient MeOH/CH2Cl2 system as follows: Initial 100% CH2Cl2, 98%-92% over 40 min; 90% over 12 min, and then 88% over 13 min. The desired product eluted cleanly between 44-61 min. The desired fractions were combined and concentrated under reduced pressure to yield 1.74 g (113% of theory) of 5-({[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid, Cpd 9b, as a white solid.
D. 5-({[2-Amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid.
A portion of Cpd 9b (0.27g, 0.41 mmol) was dissolved in EtOAc (39 mL)/THF (5 mL), filtered, and subsequently treated with gaseous HCl for 15 min. After completion of the HCl addition, the reaction was slowly warmed to room temperature and a solid precipitate formed. After 5 h the reaction appeared >97% complete by LC (@214 nm; 2.56 min.). The stirring was continued over 3 d, then the solid was collected and rinsed with a small amount of EtOAc. The resulting solid was dried under high vacuum under refluxing toluene for 2.5 h to yield 0.19 g (71%) of desired Cpd 9c as a white solid di-HCl salt.
Example 8 (S)-2-tert-Butoxycarbonylamino-3-(2,6-dimethyl-4-trifluoromethanesulfonylphenyl)-propionic acid methyl ester
A. (S)-2-tert-Butoxycarbonylamino-3-(2,6-dimethyl-4-trifluoromethanesulfonylphenyl)-propionic acid methyl ester. Into a cool solution of Boc-L-(2,6-diMe)Tyr-OMe (7.0 g, 21.6 mmol; Sources: Chiramer or RSP AminoAcidAnalogues) and N-phenyltrifluoromethanesulfonimide (7.9 g, 22.0 mmol) in dichloromethane (60 mL) was added triethylamine (3.25 mL, 23.3 mmol). The resulting solution was stirred at 0° C. for 1 h and slowly warmed to rt. Upon completion, the reaction was quenched by addition of water. The separated organic phase was washed with 1 N NaOH aqueous solution, water and dried over Na2SO4 overnight. After filtration and evaporation, the residue was purified by flash column chromatography (eluent: EtOAc-hexane: 3:7) to give the desired product (9.74 g, 99%) as a clear oil; 1H NMR (300 MHz, CDCl3): δ 1.36 (9H, s), 2.39 (6H, s), 3.06 (2H, d, J=7.7 Hz), 3.64 (3H, s), 4.51-4.59 (1H, m), 5.12 (1H, d, J=8.5 Hz), 6.92 (2H, s); MS (ES+) (relative intensity): 355.8 (100) (M−Boc)+.
B. (S)4-(2-tert-Butoxycarbonylamino-2-methoxycarbonylethyl)-3,5-dimethylbenzoic acid. To a suspension of (S)-2-tert-butoxycarbonylamino-3-(2,6-dimethyl-4-trifluoromethanesulfonylphenyl)-propionic acid methyl ester (9.68 g, 21.3 mmol), K2CO3 (14.1 g, 0.102 mol), Pd(OAc)2 (0.48 g, 2.13 mmol) and 1,1′-bis(diphenylphosphino)ferrocene (2.56 g, 4.47 mmol) in DMF (48 mL) was bubbled in gaseous CO for 15 min. The mixture was heated to 60° C. for 8 h with a CO balloon. The cool mixture was partitioned between NaHCO3 and EtOAc, and filtered. The aqueous layer was separated, acidified with 10% citric acid aqueous solution, extracted with EtOAc, and finally dried over Na2SO4. Filtration and concentration of the filtrate resulted in a residue. The residue was recrystallized from EtOAc-hexanes to afford the desired product (7.05 g, 94%); 1H NMR (300 MHz, CDCl3): δ 1.36 (9H, s), 2.42 (6H, s), 3.14 (2H, J=7.4 Hz), 3.65 (3H, s), 4.57-4.59 (1H, m), 5.14 (1H, d, J=8.6 Hz), 7.75 (2H, s); MS(ES+) (relative intensity): 251.9 (100) (M−Boc)+.
C. (S)-2-tert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethylphenyl)propionic acid methyl ester. Into a stirring solution of (S)-4-(2-tert-butoxycarbonylamino-2-methoxycarbonylethyl)-3,5-dimethyl benzoic acid (3.00 g, 8.54 mmol), PyBOP (6.68 g, 12.8 mmol) and HOBt (1.74 g, 12.8 mmol) in DMF (36 mL) was added DIPEA (5.96 mL, 34.2 mmol) and NH4Cl (0.92 g, 17.1 mmol). The resulting mixture was stirred at rt for 40 min before being partitioned between aqueous NH4Cl solution and EtOAc. The separated organic phase was washed sequentially with 2N citric acid aqueous solution, saturated aqueous NaHCO3 solution, and brine, then dried over Na2SO4 overnight. After filtration and concentration, the residue was purified by flash column chromatography (eluent: EtOAc) to give the product. (3.00 g, 100%); 1H NMR (300 MHz, CDCl3): δ 1.36 (9H, s), 2.39 (6H, s), 3.11 (2H, J=7.2 Hz), 3.65 (3H, s), 4.53-4.56 (1H, m), 5.12 (1H, d, J=8.7 Hz), 5.65 (1H, brs), 6.09 (1H, br s), 7.46 (2H, s); MS(ES+) (relative intensity): 250.9 (100) (M−Boc)+.
D. (S)-2-tert-Butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethylphenyl)propionic acid. Into an ice-cooled solution of methyl ester from Step C (2.99 g, 8.54 mmol) in THF (50 mL) was added an aqueous LiOH solution (1N, 50 mL) and stirred at 0° C. Upon consumption of the starting materials, the organic solvents were removed and the aqueous phase was neutralized with cooled 1N HCl at 0° C., and extracted with EtOAc, and dried over Na2SO4 overnight. Filtration and evaporation to dryness led to the title acid (S)-2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethylphenyl)propionic acid (2.51 g, 87%); 1H NMR (300 MHz, DMSO-d6): δ 1.30 (9H, s), 2.32 (6H, s), 2.95 (1H, dd, J=8.8, 13.9 Hz), 3.10 (1H, dd, J=6.2, 14.0 Hz), 4.02-4.12 (1H, m), 7.18-7.23 (2H, m), 7.48 (2H, s), 7.80 (1H, s); MS(ES+) (relative intensity): 236.9 (6) (M−Boc)+.
PAPER
Bioorg Med Chem Lett. 2012 Jul 15;22(14):4869-72.
PATENTS
1.WO 2005090315
2..WO 2006099060
3.WO 2009009480
4. WO 2010062590
5.US 2011263868 *
Patent
Eluxadoline chemically is 5-[[[(25)-2-amino-3-[4-(aminocarbonyl)-2, 6-dimethylphenyl] – 1 -oxopropyl] [( 15)- 1 -(4-phenyl- lH-imidazol-2-yl)ethyl] amino] methyl] -2-methoxybenzoic acid, represented by Formula I.
Formula I
Eluxadoline is a mu-opioid receptor agonist, indicated in adults for the treatment of irritable bowel syndrome with diarrhea (IBS-D).
U.S. Patent No. 7,741 ,356 describes a process for the preparation of eluxadoline. U.S. Patent Nos. 7,629,488 and 8,710,256 describe processes for the preparation of intermediates of eluxadoline.
PCT Publication No. WO2009/009480 purportedly discloses forms alpha and beta crystals of eluxadoline and processes thereof. PCT Publication No. WO2009/009480 discloses that form alpha crystals can be prepared by storing the zwitterion of eluxadoline at 0-25% relative humidity (RH) for 3 days and form beta crystals can be prepared by storing the zwitterion of eluxadoline at greater than 60% RH for 3 days.
PCT Publication No. WO2017/015606 purportedly discloses amorphous form, crystalline forms I, II, III and IV, and processes for their preparation and a process for the preparation of form alpha crystal of eluxadoline
PATENT
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018138274
Eluxadoline is the INN denomination assigned to the compound having lUPAC name 5-({[(2S)-2-amino-3-(4-carbamoyl-2,6-dimethylphenyl)propanoyl][(1 S)-1 -(4-phenyl-1 /-/-imidazol-2-yl)ethyl]amino}methyl)-2-methoxybenzoic acid and the formula reported below:
Eluxadoline is a μ- and κ-opioid receptor agonist and δ-opioid receptor antagonist that acts locally in the enteric nervous system. The drug, administered orally, is active locally in the intestine and is able to control gastrointestinal function (Gl) and at the same time to reduce the pain and mitigate the effect of constipation. Its use has been approved for the treatment of diarrhea and abdominal pain in individuals with diarrhea-predominant irritable bowel syndrome (IBS-D).
The family of compounds to which eluxadoline belongs is disclosed in patent application WO 2005/090315 A1 , while patent application WO 2006/099060 A2 is directed to processes for the preparation of these compounds.
As generally known, any active principle may exist under amorphous or different crystalline forms (polymorphs), either as pure compound or in forms in which, in the structure of the crystal, are present molecules of water (hydrates) or of another solvent (solvates); besides, in case of hydrates and solvates, the ratio between the number of molecules of active principle and molecules of water or solvent may vary, giving rise to different solid forms of the compound.
Different salts and solid-state forms of an active pharmaceutical ingredient may possess different properties. Such variations in the properties of different salts and solid-state forms may provide a basis for improving formulation, for example, by facilitating better processing or handling characteristics, changing the dissolution profile in a favourable direction, or improving stability (polymorphic and/or chemical) and shelf-life. These variations in the properties of different salts and solid-state forms may also offer improvements to the final dosage form, for instance, if they serve to improve bioavailability. Different salts, solid-state forms and solvates of an active pharmaceutical ingredient may also give rise to a variety of polymorphs or crystalline forms, which, in turn, may provide additional opportunities to assess variations in the properties and characteristics of a solid active pharmaceutical ingredient.
While not intending to be bound by any theory, certain solid forms are characterized by physical properties, e.g., stability, solubility and dissolution rate, appropriate for pharmaceutical and therapeutic dosage forms. Moreover, while not wishing to be bound by any theory, certain solid forms are characterized by physical properties (e.g., density, compressibility, hardness, morphology, cleavage, stickiness, solubility, water uptake, electrical properties, thermal behaviour, solid-state reactivity, physical stability, and chemical stability) affecting particular processes (e.g., yield, filtration, washing, drying, milling, mixing, tableting, flowability, dissolution, formulation, and lyophilization) which make certain solid forms suitable for the manufacture of a solid dosage form. Such properties can be determined using analytical chemical techniques, including solid-state analytical techniques (e.g., X-ray diffraction, microscopy, spectroscopy and thermal analysis), as described herein and known in the art.
For these reasons, chemical compounds useful in the pharmaceutical field are systematically screened looking for the physical form(s) that present an improved set of production, storage and handling properties, and which result in an improved administration to the patients.
Patent application WO 2009/009480 A2 discloses two crystalline forms of eluxadoline, referred to in the document respectively as Form a and Form β. Form a is characterized by an X-ray powder diffraction pattern having the main peaks at about 10.2°, 1 1.3°, 1 1.8°, 14.0°, 14.3°, 14.7°, 16.1 ° and 18.3° 2Θ, while Form β is characterized by an X-ray powder diffraction pattern having the main peaks at about 1 1.0°, 12.4°, 14.9°, 15.2°, 22.1 °, 25.6°, 27.4°, and 30.4° 2Θ.
Patent application WO 2017/015606 A1 discloses several crystalline forms of eluxadoline, referred to therein as Form I, Form II, Form III, and Form IV. Form I is characterized by an X-ray powder diffraction pattern having peaks at about 6.4°, 7.5°, 9.1 °, 10.0°, and 13.0° 2Θ. Form II is characterized by an X-ray powder diffraction profile having peaks at about 7.2°, 1 1 .6°, 12.1 °, 12.7° and 16.9° 2Θ. Form III is characterized by an X-ray powder diffraction pattern having peaks at about 9.3°, 10.2°, 1 1 .5°, 13.3° and 21.8° 2Θ. Form IV is characterized by an X-ray powder diffraction profile having peaks at about 9.3°, 10.2°, 1 1.5°, 13.3° and 21 .8° 2Θ.
However, no information is provided in any of these documents about any useful
properties from the standpoint of the pharmaceutical industry, neither regarding ease of handling of the forms in the production of formulations nor regarding the storage stability (polymorphic and/or chemical) of eluxadoline when prepared in one of these crystalline forms.
An object of the present invention is the provision of a novel process for the preparation of a polymorphic form a’ of eluxadoline (as defined hereinbelow) which, surprisingly, is polymorphically and chemically stable. Since this polymorphic form represents a valuable product, it is an object that upscaling of this process, in order to meet the needs of industrial-scale production, should be easily accomplishable. It is a further object of the present invention that said novel process should produce high-purity products which must contain as low an amount of possibly harmful compounds as possible.
Surprisingly, it was found that new solvate forms ε of eluxadoline allow for the realization of this process and, thus, of the new polymorphically and chemically stable crystalline form α’. It was found that in terms of the starting material from which the solvate forms ε of eluxadoline can be produced, they are extremely flexible.
Further, it was found that the reaction conditions necessary to produce these solvate forms are highly advantageous in terms of energy consumption in combination with the chemical nature of the solvents used
High Performance Liquid Chromatography-Ultraviolet Detection (HPLC-UV):
Chemical stability tests were performed using the following HPLC method
Column: XBridge C8 150 X 4.6 mm, 3.5 μηι
Mobile Phase A: 0.1 % (V/V) phosphoric acid aqueous solution
Mobile Phase B: Acetonitrile
Diluent: 1 :1 (V/V) Mixture of Mobile Phases A and B
Flow Rate: 1.3 mL/min
Runtime: 35 min
Column Temperature 30 °C
Autosampler Temperature: Ambient
Injection Volume: 5 μΙ_
Detection: 210 nm
Sample concentration: 0.4 mg/mL
Gradient Program:
PATENT
WO 2018020450
https://patents.google.com/patent/WO2018020450A2/en
Example 1
Preparation of Eluxadoline
Step 1- Preparation of 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[l- (4-phenyl- lH-imidazol-2-yl)-ethyl]-amino} -methyl)-2-methoxy-benzoic acid
To a stirred solution of 1 -(4-phenyl- lH-imidazole-2-yl)-ethyl amine (20 gm) and 5-formyl-2-methoxy-benzoic acid methyl ester (20 gm) in methanol was added catalytic amount of acetic acid (3 ml). The reaction mixture was cooled at 5°C-10°C and sodium borohydride (4 gm) was added. The reaction mixture was further stirred for 2-3 hours at room temperature. The resultant mixture was diluted with water and then partially concentrated. To this mixture was added 2N HCl solution followed by addition of dichloromethane. The phases were separated and the pH (9-1 1) of aqueous layer was adjusted using 2N NaOH solution; which was further extracted with dichloromethane. The combined organic layers were concentrated under vacuum to afford titled compound as oil (yield: 40 gm).
Step 2- Preparation of 5-({[2-tert-butoxycarbonylmethyl-3-(4-carbamoyl-2,6-dimethyl- phenyl)-propionyl] – [ 1 -(4-phenyl- 1 H-imidazol-2-yl)-ethyl] -amino} -methyl)-2-methoxy- benzoic acid methyl ester
To a stirring mixture of 2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6- dimethyl-phenyl-propionic acid (100 gm), l-ethyl-3-(3- dimethylaminopropyl)carbodiimide hydrochloride (159.4 gm) and 1- hydroxybenzotriazole (45.4 gm) in dimethylformamide (80 ml) & dichloromethane (1920 ml) was added 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[l-(4- phenyl-lH-imidazol-2-yl)-ethyl] -amino }-methyl)-2-methoxy-benzoic acid (step 1 product, 146.6 gm). The resulting mixture was stirred at room temperature for overnight and further diluted with water. The separated organic phase was washed sequentially with aqueous Na2C03 solution, IN HCl solution, water and brine. After concentration, the residue was further dissolved in DCM. The resultant solution was washed sequentially with water & IN HCl solution and then concentrated under vacuum to afford titled compound (yield: 145 gm).
Step 3- Preparation of methyl 5-((2-amino-3-(4-carbamoyl-2,6-dimethylphenyl)-N-(l-(4- phenyl- 1 H-imidazol-2-yl)ethyl) enzoate
To a stirred solution of 5-({[2-tert-butoxycarbonylmethyl-3-(4-carbamoyl-2,6- dimethyl-phenyl)-propionyl] – [ 1 -(4-phenyl- 1 H-imidazol-2-yl)-ethyl] -amino } -methyl)-2- methoxy-benzoic acid methyl ester (step -2 product, 20 gm) in THF (80 ml) was added Cone. HCl solution (30 ml). The reaction mixture was heated at 40°C-45°C. After completion of reaction, the mixture was concentrated and resultant residue was diluted with water. The pH (9-10) was adjusted using 3N NaOH solution; and resultant stick mass was dissolved in methanol. The resultant solution was concentrated under vacuum to afford titled compound (yield: 18.1 gm).
Step 4- Preparation of Eluxadoline
Into an ice cooled solution of methyl 5-((2-amino-3-(4-carbamoyl-2,6- dimethylphenyl)-N-( 1 -(4-phenyl- 1 H-imidazol-2-yl)ethyl)propanamido)methyl)-2- methoxybenzoate (step 3 product, 15 gm) in methanol was added an aqueous lithium hydroxide (3.23 gm in 30 ml water) and resultant mixture was heated at 40°C-45°C. After completion of reaction, mixture was concentrated and further diluted with water. The pH (6-7) was adjusted using 2N citric acid and resultant residue was dissolved in methanol. The resultant solution was added slowly to the acetone and stirring was continued for overnight. The solid precipitated was filtered, washed with acetone and dried to obtain an amorphous form of titled compound (Yield: 3.50 gm).
Example 2
Preparation of Eluxadoline
Step 1 : Preparation of 5-( {[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6-dimethyl- phenyl)-propionyl] – [ 1 -(4-phenyl- 1 W-imidazol-2-yl)-ethyl] -amino } -methyl)-2-methoxy- benzoic acid
Into an ice cooled solution of 5-({[2-tert-butoxycarbonylmethyl-3-(4-carbamoyl- 2,6-dimethyl-phenyl)-propionyl] – [ 1 -(4-phenyl- 1 H-imidazol-2-yl)-ethyl] -amino } -methyl)- 2-methoxy-benzoic acid methyl ester (160 gm) in methanol (800 ml) was added an aqueous solution of lithium hydroxide (29.46 gm in 350 ml water) and resultant mixture was stirred at room temperature for overnight. After completion of reaction, mixture was partially concentrated and further diluted with water. The pH (4-5) was adjusted using 2N citric acid and further stirred for 60 min. The solid precipitated was filtered, washed with water and dried to obtain titled compound (yield: 140 gm). Step 2: Preparation of Eluxadoline
To a stirred solution of 5-( {[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6- dimethyl-phenyl)-propionyl] – [ 1 -(4-phenyl- 1 H-imidazol-2-yl)-ethyl] -amino } -methyl)-2- methoxy-benzoic acid (step-1 product, 100 gm) in acetone (1200 ml) was added Cone. HC1 solution (50 ml). The reaction mixture was heated at 40°C-45°C. After completion of reaction, the supernatant solution was decanted; resultant residue was rinsed with acetone and further dissolved in water. The pH (6-7) was adjusted using IN NaOH solution and the precipitated was filtered, washed with water and dried to obtain an amorphous form of eluxadoline (yield: 72 gm).
Example 3
Preparation of Eluxadoline
To a stirred solution of 5-( {[2-tert-butoxycarbonylamino-3-(4-carbamoyl-2,6- dimethyl-phenyl)-propionyl] – [ 1 -(4-phenyl- 1 -imidazol-2-yl)-ethyl] -amino } -methyl)-2- methoxy-benzoic acid (50 gm) in dichloromethane (250 ml) were added solution of Cone. HC1 (50 ml) and water (50 ml). The reaction mixture was heated at 35°C-40°C and further stirred for 10-20 minutes. Tetrahydrofuran (50 ml) & Cone. HC1 (20 ml) were added to the sticky mass and reaction mixture was heated at 40°C for 2 hours. After completion of reaction, the mixture was diluted with water. The pH (6-7) was adjusted using 4N NaOH solution and the obtained sticky mass was dissolved in methanol. The resultant solution was concentrated under vacuum to afford eluxadoline (yield: 16 gm).
Example 4
Preparation of amorphous form of eluxadoline Eluxadoline (1 gm) was dissolved in methanol (20 ml) at 25-30°C. Water (60 ml) was added to the resultant solution and stirred for 15-20 minutes. The resultant slurry was filtered, washed with water and further dried to obtain amorphous form of eluxadoline (Yield: 0.80 gm).
Example 5
Preparation of amorphous form of eluxadoline
Eluxadoline (1 gm) was dissolved in methanol (20 ml) at 25-30°C. Acetone (80 ml) was added to the resultant solution and stirred for 15-20 minutes. The resultant slurry was filtered, washed with acetone and further dried to obtain amorphous form of eluxadoline (Yield: 0.80 gm).
Example 6
Preparation of Form L of eluxadoline
Eluxadoline (1 gm) was charged into flask containing acetonitrile (60 ml) and slurried for 24 hours to 25 hours at 50°C. The resultant solid was filtered, and dried to obtain titled compound (Yield: 0.80 gm).
clip
Eluxadoline (Viberzi)


-
60.Garnock-Jones, K. P. Eluxadoline: First Global Approval Drugs 2015, 75, 1305– 1310 DOI: 10.1007/s40265-015-0436-4
-
61.Davenport, J. M.; Covington, P.; Bonifacio, L.; McIntyre, G.; Venitz, J. Effect of Uptake Transporters OAT3 and OATP1B1 and Efflux Transporter MRP2 on the Pharmacokinetics of Eluxadoline J. Clin. Pharmacol.2015, 55, 534– 542 DOI: 10.1002/jcph.442
-
62.Wade, P. R.; Palmer, J. M.; McKenney, S.; Kenigs, V.; Chevalier, K.; Moore, B. A.; Mabus, J. R.;Saunders, P. R.; Wallace, N. H.; Schneider, C. R.; Kimball, E. S.; Breslin, H. J.; He, W.; Hornby, P. J.Modulation of Gastrointestinal Function by MuDelta, a Mixed μ Opioid Receptor Agonist/δ Opioid Receptor Antagonist Br. J. Pharmacol. 2012, 167, 1111– 1125 DOI: 10.1111/j.1476-5381.2012.02068.x
-
63.Lembo, A. J.; Lacy, B. E.; Zuckerman, M. J.; Schey, R.; Dove, L. S.; Andrae, D. A.; Davenport, J. M.;McIntyre, G.; Lopez, R.; Turner, L.; Covington, P. S. Eluxadoline for Irritable Bowel Syndrome with DiarrheaN. Engl. J. Med. 2016, 374, 242– 253 DOI: 10.1056/NEJMoa1505180
-
64.Breslin, H. J.; Cai, C.; He, W.; Kavash, R. W. Preparation of Imidazole Derivatives as Opioid Receptor Modulators. WO 20050203143A1, 2005.
-
65.cai, C.; He, W. Process for the Preparation of Amino Acid Derivatives as Opioid Modulators. WO 2006099060A1, 2006.
|
12-24-2010
|
NOVEL COMPOUNDS AS OPIOID RECEPTOR MODULATORS
|
|
|
8-32-2010
|
Compounds as opioid receptor modulators
|
|
|
6-23-2010
|
Compounds as opioid receptor modulators
|
|
|
2-12-2010
|
PROCESS FOR THE PREPARATION OF OPIOD MODULATORS
|
|
|
12-9-2009
|
Process for the preparation of opioid modulators
|
| US7629488 * | Mar 6, 2006 | Dec 8, 2009 | Janssen Pharmaceutica N.V. | Process for the preparation of opioid modulators |
| US7741356 * | Mar 14, 2005 | Jun 22, 2010 | Janssen Pharmaceutica N.V. | Compounds as opioid receptor modulators |
| US7786158 * | Oct 24, 2007 | Aug 31, 2010 | Janssen Pharmaceutica N.V. | Compounds as opioid receptor modulators |
| US7994206 | Jul 7, 2008 | Aug 9, 2011 | Janssen Pharmaceutica, N.V. | Crystals and process of making 5-({[2-amino-3-(4-carbamoyl-2,6-dimethyl-phenyl)-propionyl]-[1-(4-phenyl-1H-imidazol-2-yl)-ethyl]-amino}-methyl)-2-methoxy-benzoic acid |
| CN1950342A | Mar 14, 2005 | Apr 18, 2007 | 詹森药业有限公司 | Novel compounds as opioid receptor modulators |
References
- ^ Jump up to:a b c “Viberzi (eluxadoline) Tablets, for Oral Use, CIV. Full Prescribing Information”. Actavis Pharma, Inc. Parsippany, NJ 07054 USA. Retrieved 26 December 2015.
- ^ “Truberzi”. European Medicines Agency. 29 September 2016.
- ^ Fragkos, Konstantinos C (2017-09-25). “Spotlight on eluxadoline for the treatment of patients with irritable bowel syndrome with diarrhea”. Clinical and Experimental Gastroenterology. 10: 229–240. doi:10.2147/ceg.s123621.
- ^ “FDA approves two therapies to treat IBS-D”. http://www.fda.gov. Retrieved 2015-06-01.
- ^ “Viberzi Information from Drugs.com”. http://www.drugs.com. Retrieved 2015-06-01.
- ^ Limbo AJ, et al. Eluxadoline in Irritable Bowel Syndrome with Diarrhea. NEJM 2016;374:242-53
- ^ Commissioner, Office of the (15 March 2017). “Safety Alerts for Human Medical Products – Viberzi (eluxadoline): Drug Safety Communication – Increased Risk of Serious Pancreatitis In Patients Without A Gallbladder”. http://www.fda.gov. Retrieved 19 March 2017.
- ^ Jump up to:a b Brooks, Megan (March 2017). “FDA: Avoid IBS Drug Viberzi in Patients With No Gallbladder”. http://www.medscape.com. Retrieved 2017-09-18.
- ^ Jump up to:a b Commissioner, Office of the. “Safety Alerts for Human Medical Products – Viberzi (eluxadoline): Drug Safety Communication – Increased Risk of Serious Pancreatitis In Patients Without A Gallbladder”. http://www.fda.gov. Retrieved 2017-09-18.
- ^ “bismuth subsalicylate”. reference.medscape.com. Retrieved 2016-05-10.
- ^ Levy-Cooperman, N; McIntyre, G; Bonifacio, L; McDonnell, M; Davenport, JM; Covington, PS; Dove, LS; Sellers, EM (December 2016). “Abuse Potential and Pharmacodynamic Characteristics of Oral and Intranasal Eluxadoline, a Mixed μ- and κ-Opioid Receptor Agonist and δ-Opioid Receptor Antagonist”. The Journal of Pharmacology and Experimental Therapeutics. 359 (3): 471–481. doi:10.1124/jpet.116.236547. PMC 5118645. PMID 27647873.
- ^ “Actavis Announces FDA Acceptance for Filing of NDA for Eluxadoline”. http://www.drugs.com. Retrieved 2015-06-01.
- ^ “FDA Approves Viberzi (eluxadoline) for Irritable Bowel Syndrome with Diarrhea (IBS-D) in Adults”. http://www.drugs.com. Retrieved 2015-06-01.
- ^ Davenport, J. Michael; Covington, Paul; Bonifacio, Laura; McIntyre, Gail; Venitz, Jürgen (2015). “Effect of uptake transporters OAT3 and OATP1B1 and efflux transporter MRP2 on the pharmacokinetics of eluxadoline”. The Journal of Clinical Pharmacology. 55 (5): 534–542. doi:10.1002/jcph.442. ISSN 0091-2700. PMC 4402028.
- ^ [1], Process of the Preparation of Opioid modulators.
The active ingredient in VIBERZI is eluxadoline, a mu-opioid receptor agonist.
The full chemical name is 5-[[[(2S)-2-amino-3-[4-(aminocarbonyl)-2,6-dimethylphenyl]-1- oxopropyl][(1S)-1-(4-phenyl-1H-imidazol-2-yl)ethyl]amino]methyl]-2-methoxybenzoic acid.
Eluxadoline has a molecular weight of 569.65 and a molecular formula of C32H35N5O5. The chemical structure of eluxadoline is:
![]() |
VIBERZI is available as 75 mg and 100 mg tablets for oral administration. In addition to the active ingredient, eluxadoline, each tablet contains the following inactive ingredients: silicified microcrystalline cellulose, colloidal silica, crospovidone, mannitol, magnesium stearate, and Opadry II (partially hydrolyzed polyvinyl alcohol, titanium dioxide, polyethylene glycol, talc, iron oxide yellow, and iron oxide red).
| Clinical data | |
|---|---|
| Trade names | Viberzi (US), Truberzi (Europe) |
| Synonyms | JNJ-27018966 |
| License data | |
| Routes of administration |
Oral |
| ATC code | |
| Legal status | |
| Legal status |
|
| Pharmacokinetic data | |
| Protein binding | 81% |
| Elimination half-life | 3.7–6 hours |
| Excretion | 82.2% (feces), <1% (urine)[1] |
| Identifiers | |
| CAS Number | |
| PubChem CID | |
| IUPHAR/BPS | |
| ChemSpider | |
| UNII | |
| KEGG | |
| Chemical and physical data | |
| Formula | C32H35N5O5 |
| Molar mass | 569.6508 g/mol |
| 3D model (JSmol) | |
//////////////////JNJ-27018966, iberzi, элуксадолин ,إيلوكسادولين ,艾沙多林 ,ELUXADOLINE, FDA 2015, エルクサドリン,
CC1=CC(=CC(=C1CC(C(=O)N(CC2=CC(=C(C=C2)OC)C(=O)O)C(C)C3=NC=C(N3)C4=CC=CC=C4)N)C)C(=O)N























































