AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

A route to convert CO2: synthesis of 3,4,5-trisubstituted oxazolones

 Uncategorized  Comments Off on A route to convert CO2: synthesis of 3,4,5-trisubstituted oxazolones
Nov 292014
 

 

 

Green Chem., 2015, Advance Article

DOI: 10.1039/C4GC02033B, Paper

Jiayin Hu, Jun Ma, Zhaofu Zhang, Qinggong Zhu, Huacong Zhou, Wenjing Lu, Buxing Han

CO2 can react with various propargylic amines to form 3,4,5-trisubstituted oxazolones catalyzed by the active, selective and stable ionic liquids.

 

 

Production of value-added chemicals using carbon dioxide (CO2) as a feedstock is favorable to the sustainable development of the chemical industry. In this work, we have discovered for the first time that CO2 can react with propargylic amines to produce 3,4,5-trisubstituted oxazolones, a class of very useful chemicals. It was found that the ionic liquid (IL) 1-butyl-3-methylimidazolium acetate ([Bmim][OAc]) can catalyze the reactions efficiently at atmospheric pressure under metal-free conditions. It was also found that [Bmim][OAc] and IL 1-butyl-3-methylimidazolium bis((trifluoromethyl)sulfonyl)imide ([Bmim][Tf2N]) have an excellent synergistic effect for promoting the reactions. The [Bmim][OAc]/[Bmim][Tf2N] catalytic system can be reused at least five times without loss in catalytic activity and selectivity. The reaction mechanism was proposed on the basis of density functional theory (DFT) calculation and the experimental results.

Share

QUININE

 Uncategorized  Comments Off on QUININE
Nov 282014
 

Quinine-3D-balls.pngQuinine.svg

Quinine

Molecular Formula: C20H24N2O2
Molecular Weight: 324.417

 
IUPAC Name: 6′-Methoxycinchonan-9-ol

CAS Number: 56-54-2
PubChem: 1065
NMRShiftDB: 10016314
Spectrometer: Bruker AV 400 MHz
Probe: 1mm MicroProbe
Solvent: CDCl3
Sample Concentration: ca. 200 µg

 

 

Quinine (US /ˈkwnn/UK /ˈkwɪnn/ or /kwɪˈnn/ kwin-een) is a natural white crystalline alkaloid having antipyretic (fever-reducing),antimalarialanalgesic (painkilling), and anti-inflammatory properties and a bitter taste. It is a stereoisomer of quinidine, which, unlike quinine, is an antiarrhythmic. Quinine contains two major fused-ring systems: the aromatic quinoline and the bicyclic quinuclidine.

Quinine occurs naturally in the bark of the cinchona tree, though it has also been synthesized in the laboratory. The medicinal properties of the cinchona tree were originally discovered by the Quechua, who are indigenous to Peru and Bolivia; later, the Jesuitswere the first to bring cinchona to Europe.

Quinine was the first effective Western treatment for malaria caused by Plasmodium falciparum, appearing in therapeutics in the 17th century. It is pre-dated as a malarial treatment by the Chinese herbalist’s use of Artemisia annua, described in a 4th-century text, a plant from which the antimalarial drug artemisinin was derived. It remained the antimalarial drug of choice until the 1940s, when other drugs such as chloroquine that have fewer unpleasant side effects replaced it. Since then, many effective antimalarials have been introduced, although quinine is still used to treat the disease in certain critical circumstances, such as severe malaria, and in impoverished regions due to its low cost. Quinine is available with a prescription in the United States and “over-the-counter” (in minute quantities) in tonic water. Quinine is also used to treat lupus and arthritis. Quinine was also frequently prescribed in the US as an off-label treatment for nocturnal leg cramps, but this has become less prevalent due to a Food and Drug Administration statement warning against the practice.[2]

Quinine is highly fluorescent (quantum yield ~0.58) in 0.1 M sulfuric acid solution and it is widely used as a standard for fluorescence quantum yield measurement.[3][4] It is on the World Health Organization’s List of Essential Medicines, a list of the most important medications needed in a basic health system.[5]

1D Proton Spectrum:

1D Proton Spectrum

 

Medical uses

As of 2006, quinine is no longer recommended by the WHO (World Health Organization), as first-line treatment for malaria, and should be used only when artemisinins are not available.[6][7][8][9]

Quinine is a basic amine and is usually presented as a salt. Various existing preparations include the hydrochloride, dihydrochloride,sulfate, bisulfate and gluconate. This makes quinine dosing complicated, since each of the salts has a different weight.

The following amounts of each salt form contain equal amounts of quinine:

  • quinine base 100 mg
  • quinine bisulfate 169 mg
  • quinine dihydrochloride 122 mg
  • quinine hydrochloride 111 mg
  • quinine sulfate (actually (quinine)2H2SO4∙2H2O) 121 mg
  • quinine gluconate 160 mg

All quinine salts may be given orally or intravenously (IV); quinine gluconate may also be given intramuscularly (IM) or rectally (PR).[10][11] The main problem with the rectal route is the dose can be expelled before it is completely absorbed; in practice, this is corrected by giving a half dose again.

In the United States, quinine sulfate is commercially available in 324-mg tablets under the brand name Qualaquin; the adult dose is two tablets every eight hours. No injectable preparation of quinine is licensed in the US; quinidine is used instead.[12][13]

13C NMR

 

 

 

 

Top 10 FindIt Molecular Structures Consistent With Molecular Formula and Proton Resonances:

The correct structure is at position 7.

FindIt Structures

 Best 10 structures in decreasing rating (structure ID shown in parentheses):
    1: 0.957622 (  102439)    2: 0.957340 ( 3499717)    3: 0.955644 (   65753)
    4: 0.955078 (  847715)    5: 0.953062 ( 6336167)    6: 0.953047 (  585971)
    7: 0.952892 (    1065)    8: 0.950814 (    8547)    9: 0.949068 (  934598)
   10: 0.948731 ( 2037943)
			

2D Multiplicity-Edited HSQC Spectrum:

2D Multiplicity-Edited HSQC Spectrum

Determined HSQC Correlations:

Determined HSQC Correlations

Top 10 FindIt Molecular Structures Consistent With Proton and Protonated Carbon (HSQC) Resonances:

The correct structure is at position 1.

FindIt Structures

 Best 10 structures in decreasing rating (structure ID shown in parentheses):
    1: 0.937466 (    1065)    2: 0.915501 ( 6336167)    3: 0.900599 ( 3499717)
    4: 0.897815 (  183259)    5: 0.895740 ( 3083557)    6: 0.894629 (  101764)
    7: 0.894536 (   84495)    8: 0.891677 ( 4835740)    9: 0.890939 ( 3809868)
   10: 0.889296 ( 5191849)
	
		

Automated VerifyIt Proton Assignments:

Proton Assignments

Automated VerifyIt Carbon Assignments:

Carbon Assignments

2D COSY

2D DQF-COSY Spectrum:

2D DQF-COSY Spectrum

AssembleIt HSQC & DQF-COSY Derived Carbon-Carbon Correlations:

AssembleIt HSQC & DQF-COSY Derived Correlations

2D N15_HMBC Spectrum:

2D N15_HMBC Spectrum

AssembleIt HSQC & N15_HMBC Derived Nitrogen-Carbon Correlations:

AssembleIt HSQC & N15_HMBC Derived Correlations

2D HMBC Spectrum:

2D HMBC Spectrum

AssembleIt HSQC, HMBC, DQF-COSY & N15_HMBC Derived Correlations:

AssembleIt Derived Correlations

AssembleIt Derived Structure With NMRgraph Added Likely Oxygen Atoms:

AssembleIt Derived Structure Most likely structure (out of 277 possible ones) by agreement with carbon chemical shift prediction

Comments:

Quinine is used as an antimalaria drug.

Before performing a full structure elucidation, consider running FindIt. Only the molecular formula, proton, and/or (potentially only protonated) carbon shift information are needed.

At natural abundance, a 1D carbon spectrum is 5,700 times less sensitive to acquire than a 1D proton spectrum. Acquiring the shown HSQC and HMBC spectra instead is still faster than acquiring one 1D carbon spectrum. The full quinine structure elucidation is demonstrated using about 200 micro grams of sample. Only the shown NMR data from a room-temperature 1 mm capillary probe are used. No molecular formula (MF) or information from other spectroscopic methods are needed.

 

 

History

Quinine[34] is an effective muscle relaxant, long used by the Quechua, who are indigenous to Peru, to halt shivering due to low temperatures. The Peruvians would mix the ground bark of cinchona trees with sweetened water to offset the bark’s bitter taste, thus producing tonic water.

19th-century illustration of Cinchona calisaya

Quinine has been used in unextracted form by Europeans since at least the early 17th century. It was first used to treat malaria in Rome in 1631. During the 17th century, malaria was endemic to the swamps and marshes surrounding the city of Rome. Malaria was responsible for the deaths of severalpopes, many cardinals and countless common Roman citizens. Most of the priests trained in Rome had seen malaria victims and were familiar with theshivering brought on by the febrile phase of the disease. The Jesuit brother Agostino Salumbrino (1561–1642), an apothecary by training who lived inLima, observed the Quechua using the bark of the cinchona tree for that purpose. While its effect in treating malaria (and hence malaria-induced shivering) was unrelated to its effect in controlling shivering from rigors, it was still a successful medicine for malaria. At the first opportunity, Salumbrino sent a small quantity to Rome to test as a malaria treatment. In the years that followed, cinchona bark, known as Jesuit’s bark or Peruvian bark, became one of the most valuable commodities shipped from Peru to Europe. When King Charles II was cured of malaria at the end of the 17th Century with quinine, it became popular in London.[35] It remained the antimalarial drug of choice until the 1940s, when other drugs took over.[36]

The form of quinine most effective in treating malaria was found by Charles Marie de La Condamine in 1737.[37][38] Quinine was isolated and named in 1820 by French researchers Pierre Joseph Pelletier and Joseph Bienaimé Caventou.[39] The name was derived from the original Quechua (Inca) word for the cinchona tree bark, quina or quina-quina, which means “bark of bark” or “holy bark”. Prior to 1820, the bark was first dried, ground to a fine powder, and then mixed into a liquid (commonly wine) which was then drunk. Large-scale use of quinine as a prophylaxis started around 1850.

Quinine also played a significant role in the colonization of Africa by Europeans. Quinine had been said to be the prime reason Africa ceased to be known as the “white man’s grave”. A historian has stated, “it was quinine’s efficacy that gave colonists fresh opportunities to swarm into the Gold CoastNigeria and other parts of west Africa”.[40]

To maintain their monopoly on cinchona bark, Peru and surrounding countries began outlawing the export of cinchona seeds and saplings beginning in the early 19th century. The Dutch government persisted in its attempt to smuggle the seeds, and by the 1930s Dutch plantations in Java were producing 22 million pounds of cinchona bark, or 97% of the world’s quinine production.[40] During World War II, Allied powers were cut off from their supply of quinine when the Germans conquered the Netherlands and the Japanese controlled the Philippines and Indonesia. The United States had managed to obtain four million cinchona seeds from the Philippines and began operating cinchona plantations inCosta Rica. Nonetheless, such supplies came too late; tens of thousands of US troops in Africa and the South Pacific died due to the lack of quinine.[40] Despite controlling the supply, the Japanese did not make effective use of quinine, and thousands of Japanese troops in the southwest Pacific died as a result.[41][42][43][44]

Synthetic quinine

Robert B. Woodward

Cinchona trees remain the only economically practical source of quinine. However, under wartime pressure, research towards its synthetic production was undertaken. A formal chemical synthesis was accomplished in 1944 by American chemists R.B. Woodward and W.E. Doering.[45] Since then, several more efficient quinine total syntheses have been achieved,[46] but none of them can compete in economic terms with isolation of the alkaloid from natural sources. The first synthetic organic dyemauveine, was discovered by William Henry Perkin in 1856 while he was attempting to synthesize quinine.

 

References

  1.  “Qualaquin (quinine) dosing, indications, interactions, adverse effects, and more”.Medscape Reference. WebMD. Retrieved 29 January 2014.
  2.  “FDA Drug Safety Communication: New risk management plan and patient Medication Guide for Qualaquin (quinine sulfate)”Food and Drug Administration. 2010-08-07. Retrieved 2011-02-21.
  3.  Joseph R. Lakowicz. Principles of Fluorescence Spectroscopy 3rd edition. Springer(2006). ISBN 978-0387-31278-1. Chapter 2. page 54.
  4.  Quinine sulfate ogi.edu. Retrieved 16 August 2013
  5.  “WHO Model List of Essential Medicines” (PDF). World Health Organization. October 2013. Retrieved 22 April 2014.
  6.  World Health Organization (2006). “Guidelines for the treatment of malaria”. World Health Organization. Retrieved 10 August 2009.
  7. Dorndorp A, Nosten F, Stepniewska K, et al. (2005). “Artesunate verus quinine for treatment of severe falciparum malaria: a randomised trial”. Lancet 366 (9487): 717–25.doi:10.1016/S0140-6736(05)67176-0PMID 16125588.
  8.  Reyburn, H; Mtove, G; Hendriksen, I; Von Seidlein, L (2009). “Oral quinine for the treatment of uncomplicated malaria”. Brit J Med 339: b2066. doi:10.1136/bmj.b2066.PMID 19622550.
  9.  Achan J, Tibenderana JK, Kyabayinze D, et al. (2009). “Effectiveness of quinine versus artemether-lumefantrine for treating uncomplicated falciparum malaria in Ugandan children: randomised trial”. Brit Med J 338: b2763. doi:10.1136/bmj.b2763.
  10.  Barennes H, et al. (1996). “Efficacy and pharmacokinetics of a new intrarectal quinine formulation in children with Plasmodium falciparum malaria”. Brit J Clin Pharmacol 41 (5): 389. doi:10.1046/j.1365-2125.1996.03246.x.
  11.  Barennes, H.; Balima-Koussoubé, T; Nagot, N; Charpentier, JC; Pussard, E (2006).“Safety and efficacy of rectal compared with intramuscular quinine for the early treatment of moderately severe malaria in children: randomised clinical trial”Brit Med J 332(7549): 1055–57. doi:10.1136/bmj.332.7549.1055PMC 1458599PMID 16675812.
  12.  Center for Disease Control (1991). “Treatment with Quinidine Gluconate of Persons with Severe Plasmodium falciparum Infection: Discontinuation of Parenteral Quinine”Morb Mort Weekly Rep 40 (RR–4): 21–23. Retrieved 2006-05-06.
  13.  Magill, A; Panosian, C (2005). “Making Antimalarial Agents Available in the United States”. New Engl J Med 353 (4): 335–337. doi:10.1056/NEJMp058167.PMID 16000347.
  14.  Jamaludin A, Mohamad M, Navaratnam V, et al. (1988). “Relative bioavailability of the hydrochloride, sulphate and ethyl carbonate salts of quinine”Br J Clin Pharmacol 25 (2): 261–3. doi:10.1111/j.1365-2125.1988.tb03299.xPMC 1386482PMID 3358888.
  15.  Optically active isomers of quinine and quinidine and their respective biological actionAccessed 26/1/2009
  16.  Sanders, L. “Poison Pill”The New York Times Magazine, 4/13/2008.
  17.  http://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?id=8546
  18.  Dannenberg AL; Behal, FJ; Johnson, J; Johnson, Jamie (1983). “Use of quinine for self-induced abortion”. The Southern Medical Journal 76 (7): 846–849. doi:10.1097/00007611-198307000-00007PMID 6867792.
  19.  Yeka A, Achan J, D’Alessandro U, Talisuna AO (2009). “Quinine monotherapy for treating uncomplicated malaria in the era of artemisinin-based combination therapy: an appropriate public health policy?”. Lancet Infect Dis 9 (7): 448–452. doi:10.1016/S1473-3099(09)70109-4PMID 19555904.
  20.  “NPS warns on quinine”. Auspharm e News, 6 January 2010.
  21. Jump up to:a b Roche, R. J.; Silamut, K.; Pukrittayakamee, S.; Looareesuwan, S.; Molunto, P.; Boonamrung, S.; White, N. J. (1990). “Quinine induces reversible high-tone hearing loss”.British Journal of Clinical Pharmacology 29 (6): 780. doi:10.1111/j.1365-2125.1990.tb03704.xedit
  22.  Paintaud, G.; Alván, G.; Berninger, E.; Gustafsson, L. L.; Idrizbegovic, E.; Karlsson, K. K.; Wakelkamp, M. (1994). “The concentration-effect relationship of quinine-induced hearing impairment”. Clinical Pharmacology and Therapeutics 55 (3): 317–23.doi:10.1038/clpt.1994.32PMID 8143397edit
  23. Tange, R. A.; Dreschler, W. A.; Claessen, F. A. P.; Perenboom, R. M. (1997). “Ototoxic reactions of quinine in healthy persons and patients with Plasmodium falciparum infection”.Auris Nasus Larynx 24 (2): 131. doi:10.1016/S0385-8146(96)00031-4edit
  24.  Department of Clinical Pharmacology, Huddinge University Hospital, Sweden (1994). “The concentration-effect relationship of quinine-induced hearing impairment”. Clin Pharmacol Ther 55 (3): 317–323. doi:10.1038/clpt.1994.32PMID 8143397.
  25.  “FDA Orders Stop to Marketing Of Quinine for Night Leg Cramps”FDA Consumer MagazineFood and Drug Administration. July–August 1995. Archived from the originalon 2008-01-15. Retrieved 2009-07-31.
  26.  “FDA Orders Unapproved Quinine Drugs from the Market and Cautions Consumers About Off-Label Use of Quinine to Treat Leg Cramps”. United States Food and Drug Administration. 2006-12-11. Retrieved 2009-07-31.
  27.  “Malaria Surveillance – United States, 2004”. Center for Disease Control. 2006-11-22. Retrieved 2009-11-22.
  28.  Ballestero, JA; Plazas, PV; Kracun, S; Gómez-Casati, ME; Taranda, J; Rothlin, CV; Katz, E; Millar, NS et al. (2005). “Effects of Quinine, Quinidine, and Chloroquine on α9α10 Nicotinic Cholinergic Receptors”. Molecular Pharmacology 68 (3): 822–829.doi:10.1124/mol.105.014431PMID 15955868.
  29.  Charming, Cheryl (2006). Miss Charming’s Guide for Hip Bartenders and Wayout Wannabes. USA: Sourcebooks, Inc. p. 189. ISBN 978-1-4022-0804-1.
  30.  “Basic Concepts in Fluorescence”.
  31.  Hobhouse, Henry (2004). Šest rostlin, které změnily svět (in Czech). Prague: Akademie věd České republiky. p. 59. ISBN 80-200-1179-X.
  32.  Microgram Bulletin, Volume 42, Number 10, October 2009, Page 79. Retrieved 22 September 2012.
  33.  Porritt, M., Cryptocaryon irritans, Reef Culture Magazine, 1. Retrieved 9th Jul 2009
  34.  History of quinine: Friedrich A. Flückiger and Daniel Hanbury, Pharmacographia: A history of the principal drugs of vegetable origin, met with in Great Britain and British India(London, England: Macmillan and Co., 1874), pages 302-331: Cortex Cinchonæ.
  35.  Rocco, Fiametta (2004). Quinine: malaria and the quest for a cure that changed the world. New York, NY: Perennial.
  36. Loren, Humphrey (2000). Quinine and Quarantine.
  37.  de la Condamine (1738) “Sur l’arbre du quinquina” (On the quinquina tree) Histoire de l’Académie royale des Sciences, pages 226-243.
  38. See also: Joseph de Jussieu, Description de l’arbre à quinquina: mémoire inédit de Joseph de Jussieu (1737) (Description of the quinquina tree: unpublished memoir of Joseph de Jussieu (1737)). De Jussieu accompanied de la Condamine on the latter’s expedition to Peru.
  39. Pelletier and Caventou (1820) “Suite: Des recherches chimiques sur les quinquinas”(Continuation: Chemical research on quinquinas), Annales de Chimie et de Physique, vol. 15, pages 337-365. The authors name quinine on page 348: “…, nous avons cru devoir la nommer quinine, pour la distinguer de la cinchonine par un nom qui indique également son origine.” (…, we thought that we should name it “quinine” in order to distinguish it from cinchonine by means of a name that also indicates its origin.)
  40.  Conner, Clifford D. (2005). A People’s History of Science: Miners, Midwives, and ‘Low Mechanicks’. New York: Nation Books. pp. 95–96. ISBN 1-56025-748-2. Also citesPorter, Roy (1998). The Greatest Benefit to Mankind: A Medical History of Humanity. New York: W. W. Norton. pp. 465–466. ISBN 0-393-04634-6.
  41.  Louis Morton (1953). “29”The Fall of the Philippines. Washington, D.C.: United States Army. p. 524.
  42.  Alan Hawk. “Remembering the war in New Guinea: Japanese Medical Corps — malaria”.
  43.  Lt. Gen. Leonard D. Heaton, ed. (1963). “8”Preventive Medicine in World War II: Volume VI, Communicable Diseases: Malaria. Washington, D.C.: Department of the Army. pp. 401 and 434.
  44.  “Notes on Japanese Medical Services”Tactical and Technical Trends (U.S. War Department) (36). 1943.
  45.  Woodward R, Doering W (1944). “The Total Synthesis of Quinine”. J Am Chem Soc 66(849).
  46.  Kaufman, Teodoro S.; Rúveda, Edmundo A. (2005). “Die Jagd auf Chinin: Etappenerfolge und Gesamtsiege”. Angewandte Chemie, Int. Ed. (in German) 117 (6): 876–907. doi:10.1002/ange.200400663.

Further reading

External links

In total synthesis, the Quinine total synthesis describes the efforts in synthesis of quinine over a 150 year period. The development of synthetic quinine is considered a milestone in organic chemistry although it has never been produced industrially as a substitute for natural occurring quinine. The subject has also been attended with some controversy: in 2001 Gilbert Stork published the first stereoselective quinine synthesis and he shed doubt (calling it a myth) on the earlier claim in 1944 by Bob Woodward and William Doering on account that they had obtained not quinine but a precursor molecule. In 2001, an editorial in Chemical & Engineering Newssupported Storks claim but according to a critical 30 page review in this matter published in 2007 in Angewandte Chemie the Woodward/Doering claim is valid.

 

Chemical structure

The aromatic component of the quinine molecule is a quinoline with a methoxy substituent. The amine component has a quinuclidine skeleton and the methylene bridge in between the two components has a hydroxyl group. The substituent at the carbon-3 position is a vinyl group. The molecule is optically active with five stereogenic centers (the N1 and C4 constituting a single asymmetric unit), making synthesis potentially difficult because it is one of 16 stereoisomers.

Quinine total synthesis timeline

Quinine degradation by Pasteur
  • 1856William Henry Perkin attempts quinine synthesis by oxidation of N-allyl toluidine based on the erroneous idea that 2 equivalents of this compound with chemical formulaC10H13N plus three equivalents of oxygen yield one equivalent of C20H24N2O2 (quinine’s chemical formula) and one equivalent of water [2] His oxidations with other toluidines sets him on the path of mauveine which eventually leads to the birth of chemical industry.
Attempt at quinine by William Perkin
  • 1907: the correct atom connectivity established by Paul Rabe [3]
  • 1918: Paul Rabe and Karl Kindler synthesize quinine from quinotoxine,[4] reversing the Pasteur chemistry. The lack of experimental details in this publication would become a major issue in the Stork/Woodward controversy almost a century later.
Quinine synthesis by Rabe & Kindler
The first step in this sequence is sodium hypobromite addition to quinotoxine to an N-bromo intermediate possibly with structure 2. The second step is organic oxidation withsodium ethoxide in ethanol. Because of the basic conditions the initial product quininone interconverts with quinidinone via a common enol intermediate and mutarotation is observed. In the third step the ketone group is reduced with aluminum powder and sodium ethoxide in ethanol and quinine can be identified. Quinotoxine is the first relay molecule in the Woodward/Doering claim.
Final step in Rabe Kindler synthesis: reduction
  • 1939: Rabe and Kindler re investigate a sample left over from their 1918 experiments and identify and isolate quinine (again) together with diastereomers quinidineepi-quinine and epi-quinidine [5]
  • 1940Robert Burns Woodward signs on as a consultant for the Polaroid Corporation at the request of Edwin H. Land. Quinine is of interest to Polaroid for its light polarizingproperties.
  • 1943Prelog and Proštenik interconvert an allyl piperidine called homomeroquinene and quinotoxine.[6] Homomeroquinene (the second relay molecule in the Woodward/Doering claim) is obtained in several steps from the biomolecule cinchonine (related to quinidine but without the methoxy group):
Homomeroquinene synthesis
The key step in the assembly of quinotoxine is a claisen condensation:
Claisen condensation in Prelog conversion of homomeroquinene to quinotoxine
  • 1944Bob Woodward and W.E. Doering report the synthesis of quinine [7] starting from 7-hydroxyisoquinoline. Although the title of their 1 page publication is The total synthesis of quinine it is oddly not the synthesis of quinine but that of the precursor homomeroquinene (racemic) and then with groundwork already provided by Prelog a year earlier to quinotoxine (enantiopure after chiral resolution) that is described.
homomeroquinene synthesis by Woodward / Doering
Woodward and Doering argue that Rabe in 1918 already proved that this compound will eventually give quinine but do not repeat Rabe’s work. In this project 27-year-old assistant-professor Woodward is the theorist and post doc Doering (age 26) the bench worker. According to William, Bob was able to boil water but an egg would be a challenge. As many natural quinine resources are tied up in the enemy-held Dutch East Indies synthetic quinine is a promising alternative for fighting malaria on the battlefield and both men become instant war heroes making headlines in the New York TimesNewsweek and Life magazine.
  • 1944: The then 22 year old Gilbert Stork writes to Woodward asking him if he did repeat Rabe’s work.
  • 1945: Woodward and Doering publish their second lengthy Quinine paper.[8] One of the two referees rejects the manuscript (too much historic material, too much experimental details and poor literary style with inclusion of words like adumbrated and apposite) but it is published without changes nonetheless.
  • 1974: Kondo & Mori synthesize racemic vinylic gamma-lactones, a key starting material in Stork’s 2001 quinine synthesis.:[9]

vinyl lactone synthesis from trans-2-butene-1,4-diol and ethyl orthoacetate

The starting materials are trans-2-butene-1,4-diol and ethyl orthoacetate and the key step is a Claisen rearrangement
Lactone Chiral resolution
In this process the racemic lactone reacts in aminolysis with (S)-methylbenzylamine assisted by triethylaluminum to a diastereomeric pair of amides which can be separated by column chromatography. The S-enantiomer is converted back to the S-lactone in two steps by hydrolysis with potassium hydroxide and ethylene glycol followed by azeotropic ring closure.
  • 2001: Gilbert Stork publishes his stereoselective quinine synthesis.[11] He questions the validity of the Woodward/Doering claim: “the basis of their characterization of Rabe’s claim as “established” is unclear”. The Chemical & Engineering News is equally critical.[12]
Quinine Stork synthesis overview
  • 2007: Researcher Jeffrey I Seeman in a 30 page review [13] concludes that the Woodward–Doering/ Rabe–Kindler total synthesis of quinine is a valid achievement. He notes that Paul Rabe was an extremely experienced alkaloid chemist, that he had ample opportunity to compare his quinine reaction product with authentic samples and that the described 1918 chemistry was repeated by Rabe although not with quinotoxine itself but still with closely related derivatives.
  • 2008: Smith and Williams revisit and confirm Rabe’s d-quinotoxine to quinine route [14]
2008 Rabe quinine revisited

Stork quinine total synthesis

The Stork quinine synthesis starts from chiral (S)-4-vinylbutyrolactone 1. The compound is obtained by chiral resolution and in fact, in the subsequent steps all stereogenic centers are put in place by chiral induction: the sequence does not contain asymmetric steps.

Stork Quinine synthesis Stork quinine synthesis II
Stork quinine synthesis Introducing C8 and nitrogen

The lactone is ring-opened with diethylamine to amide 2 and its hydroxyl group is protected as a tert-butyldimethyl (TBS) silyl ether in 3. The C5 and C6 atoms are added as tert-butyldiphenylsilyl (TBDPS) protected iodoethanol in a nucleophilic substitution of acidic C4 with LDA at -78°C to 4 with correct stereochemistry. Removal of the silyl protecting group with p-toluenesulfonic acid to alcohol 4b and ring-closure by azeotropic distillation returns the compound to lactone 5 (direct alkylation of 1 met with undisclosed problems).

The lactone is then reduced to the lactol 5b with diisobutylaluminum hydride and its liberated aldehyde reacts in a Wittig reaction with methoxymethylenetriphenylphosphine(delivering the C8 atom) to form enol ether 6. The hydroxyl group is replaced in a Mitsunobu reaction by an azide group with diphenylphosphoryl azide in 7 and acid hydrolysis yields the azido aldehyde 8.

Stork Quinine synthesis Stork quinine synthesis II
First ring closure Second ring closure

The methyl group in 6-methoxy-4-methylquinoline 9 is sufficiently acidic for nucleophilic addition of its anion (by reaction with LDA) to the aldehyde group in 8 to form 10 as a mixture of epimers. This is of no consequence for stereocontrol because in the next step the alcohol is oxidized in a Swern oxidation to ketone 11. A Staudinger reaction withtriphenylphosphine closes the ring between the ketone and the azide to the tetrahydropyridine 12. The imine group in this compound is reduced to the amine 13 with sodium borohydride with the correct stereospecificity. The silyl protecting group is removed with hydrogen fluoride to alcohol 14 and then activated as an mesyl leaving group by reaction with mesyl chloride in pyridine which enables the third ring closure to 15. In the final step the C9 hydroxyl group was introduced by oxidation with sodium hydridedmso and oxygen with quinine to epiquinine ratio of 14:1.

Woodward / Doering formal quinine total synthesis

The 1944 Woodward / Doering synthesis starts from 7-hydroxyisoquinoline 3 for the quinuclidine skeleton which is somewhat counter intuitive because one goes from a stable heterocyclic aromat to a completely saturated bicyclic ring. This compound (already known since 1895) is prepared in two steps.

Woodward / Doering Quinine synthesis Woodward / Doering Quinine synthesis
Woodward/Doering quinine synthesis part I Part II

The first reaction step is condensation reaction of 3-hydroxybenzaldehyde 1 with (formally) the diacetal of aminoacetaldehyde to the imine 2 and the second reaction step is cyclization in concentrated sulfuric acid. Isoquinoline 3 is then alkylated in another condensation by formaldehyde and piperidine and the product is isolated as the sodium salt of4.

Part III
Woodward/Doering quinine synthesis part III

Hydrogenation at 220°C for 10 hours in methanol with sodium methoxide liberates the piperidine group and leaving the methyl group in 5 with already all carbon and nitrogen atoms accounted for. A second hydrogenationtakes place with Adams catalyst in acetic acid to tetrahydroisoquinoline 6. Further hydrogenation does not take place until the amino group is acylated with acetic anhydride in methanol but by then 7 is again hydrogenated with Raney nickel in ethanol at 150°C under high pressure to decahydroisoquinoline 8. The mixture of cis and trans isomers is then oxidized by chromic acid in acetic acid to the ketone 9. Only the cis isomer crystallizes and used in the next reaction step, a ring opening with the alkyl nitrite ethyl nitrite with sodium ethoxide in ethanol to10 with a newly formed carboxylic ester group and an oxime group. The oxime group is hydrogenated to theamine 11 with platinum in acetic acid and alkylation with iodomethane gives the quaternary ammonium salt 12and subsequently the betaine 13 after reaction with silver oxide.

Quinine’s vinyl group is then constructed by Hofmann elimination with sodium hydroxide in water at 140°C. This process is accompanied by hydrolysis of both the ester and the amide group but it is not the free amine that is isolated but the urea 14 by reaction with potassium cyanate. In the next step the carboxylic acid group isesterified with ethanol and the urea group replaced with a benzoyl group. The final step is a claisen condensation of 15 with ethyl quininate 16, which after acidic workup yields racemic quinotoxine 17. The desired enantiomer is obtained by chiral resolution with the chiral dibenzoyl ester of Tartaric acid. The conversion of this compound to quinine is based on the Rabe/Kindler chemistry discussed in the timelime.

External links

References

  1.  Pasteur, L. Compt. rend. 1853, 37, 110.
  2.  Perkin, W. H. J. Chem. Soc. 1896, 69, 596
  3. Rabe, P.; Ackerman, E.; Schneider, W. Ber. 1907, 40, 3655
  4.  Rabe, P.; Kindler, K. Chem. Ber. 1918, 51, 466
  5.  P. Rabe, K. Kindler, Ber. Dtsch. Chem. Ges. B 1939, 72, 263–264.
  6.  Proštenik, M.; Prelog, V. HelV. Chim. Acta 1943, 26, 1965.
  7.  The Total Synthesis of Quinine R. B. Woodward and W. E. Doering J. Am. Chem. Soc.1944; 66(5) pp 849 – 849; doi:10.1021/ja01233a516
  8.  The Total Synthesis of Quinine R. B. Woodward and W. E. Doering J. Am. Chem. Soc.1945; 67(5) pp 860 – 874; doi:10.1021/ja01221a051
  9.  SYNTHESIS OF γ-LACTONES BY THE CONDENSATION OF 2-ALKENE-1,4-DIOLS WITH ORTHOCARBOXYLIC ESTERS Kiyosi Kondo and Fumio Mori Chemistry Letters Vol.3 (1974) , No.7 pp.741-742 doi:10.1246/cl.1974.741
  10.  Synthesis and Absolute Configuration of the Acetalic Lignan (+)-Phrymarolin Fumito Ishibashi and Eiji Taniguchi Bulletin of the Chemical Society of Japan Vol.61 (1988) , No.12 pp.4361-4366 doi:10.1246/bcsj.61.4361
  11.  The First Stereoselective Total Synthesis of Quinine Gilbert Stork, Deqiang Niu, A. Fujimoto, Emil R. Koft, James M. Balkovec, James R. Tata, and Gregory R. Dake J. Am. Chem. Soc.;2001; 123(14) pp 3239 – 3242; (Article) doi:10.1021/ja004325r.
  12.  M. Jacobs, Chemical & Engineering News 2001, 79 (May 7), 5.
  13.  Review: The Woodward-Doering/Rabe-Kindler Total Synthesis of Quinine: Setting the Record Straight Jeffrey I. Seeman Angew. Chem. Int. Ed. 2007, 46, 1378–1413doi:10.1002/anie.200601551 PMID 17294412
  14.  Communication Rabe Rest in Peace: Confirmation of the Rabe-Kindler Conversion of d-Quinotoxine to Quinine: Experimental Affirmation of the Woodward-Doering Formal Total Synthesis of Quinine Aaron C. Smith, Robert M. Williams Angewandte Chemie International Edition 2008, 47, 1736–1740 doi:10.1002/anie.200705421

 

Share

DEXAMETHASONE

 Uncategorized  Comments Off on DEXAMETHASONE
Nov 272014
 

 

Dexamethasone

-alpha-fluoro-11-beta,17-alpha,21-trihydroxy-16-alpha-methylpregna-1,4-diene-3,20-dione; 9-alpha-fluoro-16-alpha-methylprednisolone; 9alpha-Fluoro-11beta,17alpha,21-trihydroxy-16alpha-methylpregn-1,4-diene-3,20-dione

CAS 50-02-2

 

1H NMR

IR

 

 

13C NMR

 

MASS

 

Share

LIDOCAINE SPECTRAL VISIT

 SYNTHESIS, Uncategorized  Comments Off on LIDOCAINE SPECTRAL VISIT
Nov 272014
 

 

2-(Diethylamino)-N-(2,6-dimethylphenyl)acetamide

Lidocaine is used in drugs formulated for local anaesthetics.

MW= 234.34 g/mol; MP= 68-69 o C; bruttoformula: C14H22N2O;

CAS-number: 137-58-6;

 

Lidocaine is an antiarrhythmic medicine and also serves as a local anaesthetic drug. It is utilized in topical application to relieve pain, burning and itching sensation caused from skin inflammations. This drug is mainly used for minor surgeries. Figure 1 shows the 1H NMR spectrum of 200 mM lidocaine in CDCl3.

Figure 1. Proton NMR spectrum of 200 mM lidocaine in CDCl3.

Calculated H-NMR spectrum of lidocaine

1H NMR Relaxation

Figures 2, 3 and 4 show the relaxation time measurements. It can be seen that the relaxation times are shortest for the CH2 protons and longest for the CH protons. The first data point amplitude increases with the number of protons for the related peak.

Figure 2. Proton T1 relaxation time measurement of 200 mM lidocaine in CDCl3.

Figure 3. Proton T2 relaxation time measurement of 200 mM lidocaine in CDCl3.

Figure 4. COSY spectrum of 200 mM lidocaine in CDCl3. The cross-peaks and corresponding exchanging protons are labeled by colour-coded arrows and ellipses.

2D COSY

Figure 4 shows the 2D COSY spectrum where two spin systems (6,7,8) to (10,11) can be clearly seen. For instance, the methyl groups at 10 and 11 positions bond to aromatic protons at 6 and 8 positions, while the methyl groups at 16 and 17 positions bond to the ethylene groups at 14 and 15 positions. No coupling occurs at positions (6,7,8) to (16,17) or (14,15).

2D Homonuclear J-Resolved Spectroscopy

The chemical shift in the 2D homonuclear j-resolved spectrum appears along the direct (f2) direction and the effects of coupling between protons appear along the indirect (f1) dimension. This enables the assignment of chemical shifts of multiplets and may help in measuring unresolved couplings. Also, a decoupled 1D proton spectrum is produced by the projection along the f1 dimension. The 2D homonuclear j-resolved spectrum of lidocaine, plus the 1D proton spectrum (blue line) are shown in Figure 5.

Figure 5. Homonuclear j-resolved spectrum of 200 mM lidocaine in CDCl3. The multiplet splitting frequencies for different couplings are colour- coded.

The projection which is vertical reveals how the multiplets disintegrate into a single peak, which makes the 1D spectrum more simplified. Peak multiplicities are produced by vertical traces from peaks in the 2D spectrum and help in determining the frequencies of proton-proton coupling. When coupling frequencies are compared between different peaks, information can be obtained regarding which peaks are bonded to each other. Also, Information regarding the coupling strength can be obtained from the size of the coupling frequencies. These couplings substantiate the results of the COSY experiment.

However, in this experiment, the effects of second order coupling appear in the f1 direction as additional peaks which are equidistant from the coupling partners detached from the zero frequency in the f1 dimension. These peaks provide proof of second order coupling partners, but are generally considered as artifacts. Figure 6 shows these coupling partners and additional peaks marked by colour-coded arrows and ellipses.

Figure 6. Homonuclear j-resolved spectrum of 200 mM lidocaine in CDCl3 showing the extra peaks due to strong couplings.

1D 13C Spectra

Figure 7 shows the 13C NMR spectra of 1 M lidocaine in CDCl3. Since the 1D Carbonexperiment is highly susceptible to the 13C nuclei in the specimen, it easily and clearly resolves 9 resonances. In this experiment, only carbons coupled to protons are seen.

Figure 7. Carbon spectra of 1 M lidocaine in CDCl3.

Given the fact that the DEPT spectra do not display the peaks at 170 and 135ppm, they must be part of quaternary carbons. The DEPT-135 and the DEPT-45 experiments provide signals ofCH3, CH2 and CH groups, while the DEPT-90 experiment provides only the signal of CH groups. However, in DEPT-135 the CH2 groups occur as negative peaks. It can thus be summed up that the peaks between 45 and 60ppm belong to ethylene groups; the peaks between 10 and 20ppm are part of the methyl groups; and the peaks between 125 and 130ppm belong to methyne groups. A similar study can be carried out on the C and CH peaks.

Heteronuclear Correlation

The Heteronuclear Correlation (HETCOR) experiment identifies the proton signal that appears along the indirect dimension and the carbon signal along the direct dimension. Figure 8 shows the HETCOR spectrum of 1 M lidocaine in CDCl3. in the 2D spectrum, the peaks reveal which proton is attached to which carbon. This experiment helps in resolving assignment uncertainty from the ID carbon spectra.

Figure 8. HETCOR spectrum of 1 M lidocaine in CDCl3.

Heteronuclear Multiple Quantum Coherence

Heteronuclear Multiple Quantum Coherence (HMQC) is similar to the HETCOR experiment and is utilized to associate proton resonances to the carbons that are coupled directly to those protons. But in the HMQC experiment, the proton signal appears along the direct dimension and the carbon signal along the indirect dimension. Figure 9 shows the HMQC spectrum of 1 Mlidocaine in CDCl3. In the 2D spectrum, the peaks show which proton is attached to which carbon. For conclusive peak assignment, a similar study with the HETCOR spectrum can be carried out.

Figure 9. HMQC spectrum of 1 M lidocaine in CDCl3.

Heteronuclear Multiple Bond Correlation

The Heteronuclear Multiple Bond Correlation (HMBC) experiment can be employed to achieve long-range correlations of proton and carbon via two or three bond couplings. Similar to the HMQC experiment, the proton signal appears along the direct dimension and the carbon signal along the indirect dimension. Figure 10 shows the HMBC spectrum of 1 M lidocaine in CDCl3.

Figure 10. HMBC spectrum of 1 M lidocaine in CDCl3, with some of the long-range couplings marked.

The couplings amid the molecular positions appear analogous to the couplings seen in the COSY spectrum; however, the HMBC also displays couplings to quaternary carbons, which are not seen either in HMQC or COSY experiments. In addition, there is a correlation between protons and carbons. This is attributed to three-bond bonding from 14 and 15 and vice versa, as shown in light green in Figure 1.

IR

 

Chemical reaction of lidocaine with singlet oxygen. Rate constants for the chemical reaction between lidocaine and O2(1D g) were determined in methanol, acetonitrile and N,N-dimethylformamide. Lidocaine consumption was followed during the reaction. Rate constants for the chemical reaction, kRLID, are (1.05 ± 0.061) x 10M-1 s-1, (1.42 ± 0.073) x 10M-1 s-1and (0.61 ± 0.046) x 10M-1 s-1 in acetonitrile, methanol and N,N-dimethylformamide, respectively.

By using the Mair method (4) for hydroperoxide determination, a concentration equivalent to 0.0153 M of hydroperoxide was found when 0.03 M lidocaine in acetonitrile was irradiated for 12 h in the presence of Rose Bengal. The amount of hydroperoxide produced agrees with the consumption of lidocaine. Although we cannot isolate reaction products in quantities adequate for spectroscopic characterization, a rough idea of the product distribution was obtained by GC-MS analysis of the main lidocaine derivatives produced in the photooxidations. When 0.03 M lidocaine was irradiated for 12 h in the presence of Rose Bengal the results shown in Fig. 2 a) are obtained with the mass spectrometer in the positive chemical ionization (CI+) mode. Only four peaks appear in the chromatogram, the main one, with a retention time of 15.59 min, is that of unreacted lidocaine. Fig. 2 b) shows that the mass spectrum is that of lidocaine. The CI+ and EI (not included) mass spectra corresponding to peaks at retention times of 14.57, 13.22 and 7.77 min, indicate that 2-(ethylvinylamino)-N-(2,6-dimethylphenyl)-acetamide, 2-(1-azapropily-den)-N-(2,6-dimethyl-phenyl)-acetamide and 2,6-dimethylaniline are the probable main products of photooxidation of lidocaine. Figs. 2 c), 2 d) and 2 e), show the CI+ mass spectra and corresponding structures.

 

Figure 2.a) GC-MS chromatogram of 30 mM lidocaine in acetonitrile after 12 h of irradiation in the presence of Rose Bengal. b) CI+ mass spectrum of compound with retention time 15.58 m. c) CI+ mass spectrum of compound with retention time 14.67 m. d) CI+ mass spectrum of compound with retention time 13.22 m. e) CI+ mass spectrum of compound with retention time 7.76 m.

Share

Ethyl-2-butenoate NMR

 Uncategorized  Comments Off on Ethyl-2-butenoate NMR
Nov 262014
 

 

Ethyl-2-butenoate

1H-NMR proton decoupled spectrum of Ethyl-2-butenoate in CDCl3.

 

1H-NMR proton coupled spectrum of Ethyl-2-butenoate in CDCl3.

 

13C-NMR proton decoupled spectrum of Ethyl-2-butenoate in CDCl3.

 

DEPT spectrum of Ethyl-2-butenoate

 

COSY spectra

  • The information on the H that are coupling with each other is obtained by looking at the peaks inside the grid.  These peaks are usually shown in a contour type format, like height intervals on a map.
  • In order to see where this information comes from, let’s consider an example shown below, the COSY of ethyl 2-butenoate 
  • First look at the peak marked A in the top left corner.  This peak indicates a coupling interaction between the H at 6.9 ppm and the H at 1.8 ppm.  This corresponds to the coupling of the CH3 group and the adjacent H on the alkene.
  • Similarly, the peak marked B indicates a coupling interaction between the H at 4.15 ppm and the H at 1.25 ppm.  This corresponds to the coupling of the CH2 and the CH3 in the ethyl group.
  • Notice that there are a second set of equivalent peaks, also marked A and Bon the other side of the diagonal.

COSY spectra of ethyl 2-butenoate
(COSY spectra recorded by D. Fox, Dept of Chemistry, University of Calgary on a Bruker Advance DRX-400 spectrometer)


HETCOR spectra

  • The information on how the H are C are matched is obtained by looking at the peaks inside the grid.  Again, these peaks are usually shown in a contour type format, like height intervals on a map.
  • In order to see where this information comes from, let’s consider an example shown below, the HETCOR of ethyl 2-butenoate.
  • First look at the peak marked A near the middle of the grid.  This peak indicates that the H at 4.1 ppm is attached to the C at 60 ppm.  This corresponds to the -OCH2– group.
  • Similarly, the peak marked B towards the top right in the grid indicates that the H at 1.85 ppm is attached to the C at17 ppm.  Since the H is a singlet, we know that this corresponds to the CH3– group attached to the carbonyl in the acid part of the ester and not the CH3– group attached to the -CH2– in the alcohol part of the ester.
  • Notice that the carbonyl group from the ester has no “match” since it has no H attached in this example.

HETCOR spectra of ethyl 2-butenoate
(HETCOR spectra recorded by D. Fox, Dept of Chemistry, University of Calgary on a Bruker Advance DRX-400 spectrometer)

Share

Drug Discovery: Researchers optimize syntheses by adapting coupling reactions in array format

 drugs  Comments Off on Drug Discovery: Researchers optimize syntheses by adapting coupling reactions in array format
Nov 242014
 
09247-notw8-amine

The Merck group used arrays of coupling reactions to optimize the microgram-scale C–N coupling of an aryl halide (left) with an amine (over arrow) to give an arylamine. They then scaled up the process to gram level.
READ AT
Share

Zingerone

 Ayurveda, Uncategorized  Comments Off on Zingerone
Nov 222014
 

 

 

 

 

 

 

 

 

 

Zingerone Flower

 

Spice

 

In everyday life, ginger is used in cooking for its hot taste as well as its pungent smell.

Ecologists have studied the relationships of food and culture for many years. During this time, they have found surprising ties between spices that taste good and health-promoting side effects. An example of one of these spices is ginger.

The sensory perception of ginger in the mouth and the nose arises from two distinct groups of chemicals:

  • The volatile oils, a mixture of terpenoids that give ginger its characteristic aroma and modifies its taste.   

  • The non-volatile pungent chemicals like gingerolsshogaolsparadols and zingerone produce the “hot” taste in the mouth. It is these non-volatile pungent chemicals of ginger that give it, its real value.

 

 

Medicine

Ginger for many years has been the traditional remedy for colds. In modern medicine today, zingerone is used to treat a variety of medical problems. Zingerone reacts with free radicals that can cause tissue damage and inflammation. At Case Western University, research has been done showing that a topically applied extract containing zingerone may help prevent some skin cancers. In capsule form, ginger can also be used to replace anti-inflammatory drugs. In a recent study, ginger was found to be more effective than drugs in the treatment of nausea and motion sickness. Zingerone also has a major role in lipid oxidation since it is an anti-oxidant. It weakly inhibits oxidation of phospholipid liposomes in the presence of iron (III) and ascorbate to prevent heart-attacks.

 

Zingerone used in pharmaceuticals

It is these properties that have made zingerone a molecule of great importance and one that has been produced and synthesized for pharmaceutical use.

 

 

Structure

4-(4-hydroxy-3-methoxy-phenyl)-butan-2-one
Chemical Name Structure of Zingerone 3D Structure of Zingerone

 

Physical Properties

Synonyms Zingerone; Vanillyl Acetone
Odour Description Sweet, Spicy, Ginger, Vanilla, Woody
Appearance Yellow To Yellow-Brown Crystals
Mol./Wt. 194.2
Formula
Cas. # 122-48-5
Refractive Index 1.54400 – 1.54500 @ 20.00 �C.
Melting Point 40 – 41�C (Solvent – Aq. Ethanol)
Boiling Point 141 �C. @ 0.5 Torr
Boiling Point 290�C. @ 760.00 mmHg
Soluble in Ethyl Alcohol, 1:1 In 50% Alcohol
Natural Occurrence Ginger Root; Raspberry; Zingiber Officinale

 

 

 

Nuclear Magnetic Resonance (NMR) Spectrum of Zingerone
Table 1 : H NMR spectral data on Zingerone

 

 

Mass Spectrum of Zingerone

Pattern for Mass Fragmentation in the Spectrum

 

 

 

Infrared Spectrum of Zingerone

Table 3 : Infra Red spectral data on Zingerone

 

NOMURA METHOD

 

The Bunce & Reeves Method

 

Historical

Zingerone has been extracted from ginger for the past two thousand years.

A Brief History

Common Name:
Ginger
Latin Name: Zingiber Officinale
Family: zingiberaceae
Other Names: Based on its origin:

African ginger
Black ginger
Chochin (Asian ginger).
Gan Jiang (Jamaican ginger).

The word ginger comes from the ancient Sanskrit ‘singabera’, meaning ‘shaped like a horn’.  It first appeared in the writings of Confucius in the 5th century BC. and it has been used medicinally in the West for the past 2000 years. Various virtues have been ascribed to the spice; e.g. Henry VIII recommended it as a pro-phylactic against the plague. It was introduced by the Spaniards to the Americas and is now cultivated extensively in the West Indies. The Portuguese introduced it to West Africa. It is now used all over the world.

Only in the past century has zingerone been produced synthetically. A few interesting and unique syntheses have been chosen. Down the years, the technique of synthesis evolved. Though Lapworth & Wykes were the pioneers in the synthesis of zingerone, their method and reagents were not repeated. Below is a diagram of the apparatus used in their experiment.

Nomura, then Mannich and Merz founded the method used today. This is shown by Kim and Kim’s work. The other interesting method was formed by Bunce and Reeves. This method and the common one used today are elaborated on in the preparation section.

Year

Method Reagents
1917 Reduction and Decarboxylation of ethyl vanillylideneacetoacetateLapworth & Wykes The Pioneers in the synthesis.Na Amalgam and NaOH solution

 

1925 Reduction of 4-(4-hydroxy-3-methoxyphenyl)buten-2-one to 4-(4-hydroxy-3-methoxyphenyl)butan-2-one.Nomura Na Amalgam and Water 
1927 Reduction of 4-(4-hydroxy-3-methoxyphenyl)buten-2-one to 4-(4-hydroxy-3-methoxyphenyl)butan-2-one.Mannich & Merz Pd Catalyst, 
1979 Reaction of 4-benzyloxy-3-methoxybromomethylbenzene with the anion of acetone dimethylhydrazone, followed by oxidative hydrolysis, then hydrogenolysis to remove the benzyl groupEnders D. et Al Pd/C, Hydrogen, Methanol 
1989 Amberlyst-15-catalyzed addition of 3-buten-2-one to the phenolBunce & Reeves Amberlyst-15, Toluene, 20-50�C 
2004 Reduction of 4-(4-hydroxy-3-methoxyphenyl)buten-2-one to 4-(4-hydroxy-3-methoxyphenyl)butan-2-one.Kim D. & Kim J. Y. Pd/C Catalyst, 

 

 

Spice

ginger root
Ginger

 

As commented upon in the introduction, zingerone puts the ‘zing’ in ginger. In mustard oil, zingerone uses its properties to give it its flavour.

Zingerone has properties that give its strength of flavour. The higher molecular weight of zingerone in combination with the polar side-chain carbonyl group make zingerone molecules attract each other strongly. As a result, zingerone is not that volatile. The odour of ginger isn’t strong, but the hydrocarbon tail gives it a more intense flavour when it does come into contact with its receptor. Zingerone is also used in artificial flavouring and in fragrances.

A company called “Aroma Fragrance Fine Chemicals” (AFFC) have formulations are used globally for imparting attractive taste and aroma to processed foods and beverages. These chemicals can also add pleasing scents to perfumes, toiletries and detergents. Zingerone is one of their formulations. It is both a flavour and fragrance ingredient. This therefore means that zingerone is important in both processed foods and perfumes.

In industry, zingerone is therefore synthesised using the Kim & Kim’s method for this purpose.

 

Zingerone contains vanilloid (3-methoxy-4-hydroxy benzene) group in its structure. These phenolic hydroxyl groups provide the possibility to introduce a 4-ether-linked propanolamine side chain. Propanolamine derivatives were obtained by reacting zingerone with epichlorohydrin, and the obtained epoxide compounds were then reacted with isopropylamine, tert-butylamine or guaiacoxyethylamine, respectively, to yield 3 new derivatives shown below.

The syntheses above create third-generation β-adrenoceptor blockers, which possess ancillary cardiovascular actions other than β-adrenoceptor blockade, or cardioselective β-adrenoceptor blockers, have been shown to improve left ventricular function and decrease the risk of chronic heart failure. These β-adrenoceptor blockers are a break through in science and make the synthesis of zingerone very important in industry.

  • Zingerone
    • Ginger : www.wrc.net/wrcnet_content/herbalresources/materiamedica/materiamedica.aspx?mmid=13
  • Introduction
    • Picture of Bulbophyllum patens : Journal; Tan, Keng-Hong; Nishida, Ritsuo; JCECD8; J. Chem. Ecol.; EN; 26; 2; 2000; 533 – 546.
    • Ginger Flower:http://www-ang.kfunigraz.ac.at/~katzer/engl/generic_frame.html?Zing_off.html
    • Ecological Formulas Bottle : www.amazon.com
  • Historical
    • Cotton Diagram
      Patent Number: US2381210
      Publication date: 1945-08-07
      Inventor(s): COTTON WILLIAM J
      Applicant(s): PENNSYLVANIA COAL PRODUCTS COM
      Requested Patent: US2381210
      Application Number: US19430499408 19430820
      Priority Number(s): US19430499408 19430820
      IPC Classification:
      EC Classification: C07C45/62
      Equivalents:
  • Characterisation
    • NMR: Journal; Tan, Keng-Hong; Nishida, Ritsuo; JCECD8; J. Chem. Ecol.; EN; 26; 2; 2000; 533 – 546.
    • IR Spectrum and Mass Spectrum: NIST Mass Spec Data Center, S.E. Stein, director (http://webbook.nist.gov)
    • Mass Fragmentation: Journal; Locksley,H.D. et al.; JCPRB4; J. Chem. Soc. Perkin Trans. 1; EN; 1972; 3001-3006
    • IR Bands: Journal; Das, B.; Takhi, M.; Kumar, H. M. Sampath; Srinivas, K. V. N. S.; Yadav, J. S.; PYTCAS; Phytochemistry; EN; 33; 3; 1993; 697-700.
    • Physical Properties: http://www.thegoodscentscompany.com/data/rw1006231.html
  • Spice
    •  http://www.ulg.ac.be/lcfi/licence/interros/interro10.html
  • Medicine
    • Molecule: http://mcb.berkeley.edu/labs/berger/Structure%20images/Collaboration/lr5lab.jpeg
    • Reactions: Journal; Yeun-Chih Huang et al., Bioorganic & Medicinal Chemistry 9 (2001) 1739�1746
    • Graph and Table: Journal; Pearson, D. A. et al.; J. Agric. Food Chem. 1997, 45, 578-582
    • Diagram: http://images.medscape.com/pi/editorial/clinupdates/2002/2009/art-2009-1.fig16.gif           
  • Phytochemistry
    • GC Diagram: Journal; Tan, Keng-Hong; Nishida, Ritsuo; JCECD8; J. Chem. Ecol.; EN; 26; 2; 2000; 533 – 546.
    • Bulbophyllum patens (Phytochemistry): http://www.ulg.ac.be/lcfi/licence/interros/interro10.html
Share

Ragaglitazar ……..Dr. Reddy’s Research Foundation

 diabetes, Phase 3 drug  Comments Off on Ragaglitazar ……..Dr. Reddy’s Research Foundation
Nov 202014
 

Ragaglitazar

NNC-61-0029, (-) – DRF-2725, NN-622,

(−)DRF 2725

cas   222834-30-2

222834-21-1 (racemate)

Hyperlipidemia; Hypertriglyceridemia; Lipid metabolism disorder; Non-insulin dependent diabetes

PPAR alpha agonist; PPAR gamma agonist

(2S)-2-ETHOXY-3-{4-[2-(10H-PHENOXAZIN-10-YL)ETHOXY]PHENYL}PROPANOIC ACID,

(2S)-2-ethoxy-3-[4-(2-phenoxazin-10-ylethoxy)phenyl]propanoic acid, DRF, 1nyx

Molecular Formula: C25H25NO5
Molecular Weight: 419.4697 g/mol
Dr. Reddy’s Research Foundation (Originator), Novo Nordisk (Licensee)
Antidiabetic Drugs, ENDOCRINE DRUGS, Type 2 Diabetes Mellitus, Agents for, Insulin Sensitizers, PPARalpha Agonists, PPARgamma Agonists
Phase III
…………………..
EP 1049684; JP 2001519422; WO 9919313
Several related procedures have been described for the synthesis of the title compound. The Horner-Emmons reaction of 4-benzyloxybenzaldehyde (I) with triethyl 2-ethoxyphosphonoacetate (II) afforded the unsaturated ester (IIIa-b) as a mixture of E/Z isomers. Simultaneous double-bond hydrogenation and benzyl group hydrogenolysis in the presence of Pd/C furnished phenol (IV). Alternatively, double-bond reduction by means of magnesium in MeOH was accompanied by transesterification, yielding the saturated methyl ester (V). Further benzyl group hydrogenolysis of (V) over Pd/C gave phenol (VI). The alkylation of phenols (IV) and (VI) with the phenoxazinylethyl mesylate (VII) provided the corresponding ethers (VIII) and (IX), respectively. The racemic carboxylic acid (X) was then obtained by hydrolysis of either ethyl- (VIII) or methyl- (IX) esters under basic conditions.
…………………………………………
……………………………………………………..
The synthesis of ragaglitazar (Scheme 1) was commenced by treating substrate 2 under optimized phase-transfer catalyzed conditions, using solid cesium hydroxide monohydrate as the base, a pivalate protected benzyl bromide and the Park and Jew triflurobenzyl-hydrocinchonidinium bromide salt 1. We were delighted to find that this reaction produced 3 in good yield with good selectivity. Subsequent removal of the diphenylmethyl (DPM) group under Lewis acidic conditions followed by a Baeyer-Villager like oxidation yielded the - hydroxy aryl ester 4. At this point, we were again pleased to find that this ester could be recrystalized from warm ether to give essentially enantiomerically pure products (~95% ee). The free hydroxyl was then alkylated using triethyloxonium tetrafluoroborate, and then transesterification under catalytic basic conditions produced 5. A mesylated phenoxazine alcohol reacted with 5 to yield the methyl ester of 6, which was obtained by treatment with sodium hydroxide in methanol. The overall synthesis proceeds with 47% overall yield (41% from commercially available reagents) and is eight linear steps from the alkoxyacetophenone substrate 2, including a recrystalization.

……………………………………………………………..

J Med Chem 2001,44(16),2675

http://pubs.acs.org/doi/abs/10.1021/jm010143b

 

Abstract Image

(−)DRF 2725 (6) is a phenoxazine analogue of phenyl propanoic acid. Compound 6 showed interesting dual activation of PPARα and PPARγ. In insulin resistant db/db mice, 6 showed better reduction of plasma glucose and triglyceride levels as compared to rosiglitazone. Compound has also shown good oral bioavailability and impressive pharmacokinetic characteristics. Our study indicates that 6 has great potential as a drug for diabetes and dyslipidemia.

Figure

Scheme 1 a

 

a (a) NaH, DMF, 0−25 °C, 12 h; (b) triethyl 2-ethoxy phosphosphonoacetate, NaH, THF, 0−25 °C, 12 h; (c) Mg/CH3OH, 25 °C, 12 h; (d) 10% aq NaOH, CH3OH, 25 °C, 6 h; (e) (1) pivaloyl chloride, Et3N, DCM, 0 °C, (2) (S)-2-phenyl glycinol/Et3N; (f) 1 M H2SO4, dioxane/water, 90−100 °C, 80 h.

Compound 6 is prepared from phenoxazine using a synthetic route shown in Scheme 1. Phenoxazine upon reaction with p-bromoethoxy benzaldehyde 89 gave benzaldehyde derivative 9. Reacting 9 with triethyl 2-ethoxy phosphonoacetate afforded propenoate 10 as a mixture of geometric isomers. Reduction of 10 using magnesium methanol gave propanoate 11, which on hydrolysis using aqueous sodium hydroxide gave propanoic acid 12 in racemic form. Resolution of 12 using (S)(+)-2-phenyl glycinol followed by hydrolysis using sulfuric acid afforded the propanoic acid 6 in (−) form.

Nate, H.; Matsuki, K.; Tsunashima, A.; Ohtsuka, H.; Sekine, Y. Synthesis of 2-phenylthiazolidine derivatives as cardiotonic agents. II. 2-(phenylpiperazinoalkoxyphenyl)thiazolidine-3-thiocarboxyamides and corresponding carboxamides. Chem. Pharm. Bull198735, 2394−2411

(S)-3-[4-[2-(Phenoxazin-10-yl)ethoxy]phenyl]-2-eth-oxypropanoic Acid (6).  as a white solid, mp: 89−90 °C.

[α]D 25 = − 12.6 (c = 1.0%, CHCl3).

1H NMR (CDCl3, 200 MHz): δ 1.16 (t, J = 7.0 Hz, 3H), 1.42−1.91 (bs, 1H, D2O exchangeable), 2.94−3.15 (m, 2H), 3.40−3.65 (m, 2H), 3.86−4.06 (m, 3H), 4.15 (t, J = 6.6 Hz, 2H), 6.63−6.83 (m, 10H), 7.13 (d, J = 8.5 Hz, 2H). Mass m/z (relative intensity):  419 (M+, 41), 197 (15), 196 (100), 182 (35), 167 (7), 127 (6), 107 (19).

Purity by HPLC: chemical purity: 99.5%; chiral purity: 94.6% (RT 27.5).

…………………………………

http://www.google.com/patents/US6608194?cl=zh

EXAMPLE 23 (−) 3-[4-[2-(phenoxazin-10-yl)ethoxy]phenyl]-2-ethoxypropanoic acid:

 

Figure US06608194-20030819-C00052

 

The title compound (0.19 g, 54%) was prepared as a white solid from diastereomer [(2S-N(1S)]-3-[4-[2-(phenoxazin-10-yl)ethoxy]phenyl]-2-ethoxy-N-(2-hydroxy-1-phenyl)ethylpropanamide (0.45 g, 0.84 mmol) obtained in example 21by an analogous procedure to that described in example 22. mp: 89-90° C.

[α]D 25=−12.6 (c=1.0% CHCl3)

1H NMR (CDCl3, 200 MHz): δ 1.16 (t, J=7.02 Hz, 3H), 1.42-1.91 (bs, 1H, D2O exchangeable), 2.94-3.15 (complex, 2H), 3.40-3.65 (complex, 2H), 3.86-4.06 (complex, 3H), 4.15 (t, J=6.65 Hz, 2H), 6.63-6.83 (complex, 10H), 7.13 (d, J=8.54 Hz, 2H).

………………………..

http://www.google.com/patents/EP1049684A1?cl=en

Example 23

(S)-3-[4-[2-(phenoxazin-10-yl)ethoxy]phenyl]-2-ethoxypropanoic acid :

 

Figure imgf000051_0002

The title compound (0.19 g, 54 %) was prepared as a white solid from diastereomer [(2S- N(lS)]-3-[4-[2-(phenoxazin-10-yl)ethoxy]phenyl]-2-ethoxy-N-(2-hydroxy-l- phenyl)propanamide (0.45 g, 0.84 mmol) obtained in example 21b by an analogous procedure to that described in example 22. mp : 89 – 90 °C. [α]D 25 = – 12.6 (c = 1.0 %, CHC13)

*H NMR (CDC13, 200 MHz) : δ 1.16 (t, J = 7.02 Hz, 3H), 1.42 – 1.91 (bs, IH, D20 exchangeable), 2.94 – 3.15 (complex, 2H), 3.40 – 3.65 (complex, 2H), 3.86 – 4.06 (complex, 3H), 4.15 (t, J = 6.65 Hz, 2H), 6.63 – 6.83 (complex, 10H), 7.13 (d, J = 8.54 Hz, 2H).

Patent Submitted Granted
Benzamides as ppar modulators [US2006160894] 2006-07-20
Novel tricyclic compounds and their use in medicine process for their preparation and pharmaceutical compositions containing them [US2002077320] 2002-06-20
Tricyclic compounds and their use in medicine process for their preparation and pharmaceutical compositions containing them [US7119198] 2006-07-06 2006-10-10
Tricyclic compounds and their use in medicine: process for their preparation and pharmaceutical compositions containing them [US6440961] 2002-08-27
Tricyclic compounds and their use in medicine process for their preparation and pharmaceutical compositions containing them [US6548666] 2003-04-15
Tricyclic compounds and their use in medicine process for their preparation and pharmaceutical compositions containing them [US6608194] 2003-08-19
CRYSTALLINE R- GUANIDINES, ARGININE OR (L) -ARGININE (2S) -2- ETHOXY -3-{4- [2-(10H -PHENOXAZIN -10-YL)ETHOXY]PHENYL}PROPANOATE [WO0063189] 2000-10-26
Pharmaceutically acceptable salts of phenoxazine and phenothiazine compounds [US6897199] 2002-11-14 2005-05-24
Tricyclic compounds and their use in medicine process for their preparation and pharmaceutical compositions containing them [US6939988] 2005-09-06

WO-2014181362

  1. wo/2014/181362 a process for the preparation of 3 … – WIPO

    patentscope.wipo.int/search/en/WO2014181362

    Nov 13, 2014 – (WO2014181362) A PROCESS FOR THE PREPARATION OF 3-ARYL-2-HYDROXY PROPANOIC ACID COMPOUNDS …

A process for the preparation of 3-aryl-2-hydroxy propanoic acid compounds

ragaglitazar; saroglitazar

Council of Scientific and Industrial Research (India)

Process for preparing enantiomerically pure 3-aryl-2-hydroxy propanoic acid derivatives (eg ethyl-(S)-2-ethoxy-3-(4-hydroxyphenyl)propanoate), using S-benzyl glycidyl ether as a starting material. Useful as intermediates in the synthesis of peroxisome proliferator activated receptor agonist such as glitazars (eg ragaglitazar or saroglitazar). Appears to be the first filing on these derivatives by the inventors; however see WO2014181359 (for a concurrently published filing) and US8748660 (for a prior filing), claiming synthesis of enantiomerically pure compounds.

  1. Dolling, U. H.; Davis, P.; Grabowski, E. J. J. Efficient Catalytic Asymmetric Alkylations. 1. Enantioselective Synthesis of (+)-Indacrinone via Chiral Phase-Transfer Catalysis. J. Am. Chem. Soc. 1984, 106, 446–447.
  2. Andrus, M. B.; Hicken, E. J.; Stephens, J. C. Phase-Transfer Catalyzed Asymmetric Glycolate Alkylation. Org. Lett. 2004, 6, 2289–2292.
  3. Andrus, M. B.; Hicken, E. J.; Stephens, J. C.; Bedke, D. K. Asymmetric Phase-Transfer Catalyzed Glycolate Alkylation, Investigation of the Scope, and Application to the Synthesis of (-)-Ragaglitazar. J. Org. Chem. 2005, ASAP.
  4. Henke, B. R. Peroxisome Proliferator-Activated Receptor  Dual Agonists for the Treatment of Type 2 Diabetes. J. Med. Chem. 2004, 47, 4118–4127.
  5. Wilson, T. M.; Brown, P. J.; Sternbach, D. D.; Henke, B. R. The PPARs: from orphan receptors to drug discovery. J. Med. Chem. 2000, 46, 1306–1317.
  6. Uchida, R.; Shiomi, K.; Inokoshi, J.; Masuma, R.; Kawakubo, T.; Tanaka, H.; Iwai, Y.; Omura, A. Kurasoins A and B, New Protein Farnesyltrasferase Inhibitors Produced by Paecilomyces sp. FO-3684. J. Antibio. 1996, 49, 932–934.
Share

Esoxybutynin, (S)-Oxybutynin

 Uncategorized  Comments Off on Esoxybutynin, (S)-Oxybutynin
Nov 172014
 

Chemical structure for Esoxybutynin [INN]

(S)-2-Cyclohexyl-2-phenylglycolic acid 4-diethylaminobut-2-ynyl ester

Drug name, 药物名称….. Esoxybutynin, (S)-Oxybutynin

 

(S)-Oxybutynin Structure

Sepracor (Originator)
RENAL-UROLOGIC DRUGS, Urinary Incontinence Therapy, Anticholinergics
Phase III

 

CAS No. 119618-22-3
Chemical Name: (S)-Oxybutynin
Synonyms: Esoxybutynin;(S)-Oxybutynin;(S)-OXYBUTYNIN HCL;(S)-OXYBUTYNIN CHLORIDE;(S)-OXYBUTYNIN HYDROCHLORIDE;(S)-Hydroxycyclohexylphenylacetic acid 4-(diethylamino)-2-butynyl ester;(S)-CYCLOHEXYL-HYDROXY-PHENYL-ACETIC ACID 4-DIETHYLAMINO-BUT-2-YNYL ESTER;(αS)-α-Cyclohexyl-α-hydroxybenzeneacetic acid 4-(diethylamino)-2-butin-1-yl ester;Benzeneacetic acid, a-cyclohexyl-a-hydroxy-, 4-(diethylamino)-2-butynyl ester, (S)-;(S)-α-Phenylcyclohexaneglycolic Acid 4-(Diethylamino)-2-butynyl Ester, Hydrochloride
CBNumber: CB1746039
Molecular Formula: C22H31NO3
Formula Weight: 357.49

Oxybutynin and its derivatives are applicable as a bronchodilator or a remedy for pollakisuria. Also, oxybutynin exerts a direct antispasmodic effect on various forms of smooth muscle, mainly by inhibiting the action of acetylcholine on smooth muscle as an anti-cholinergic drug and the like. Oxybutynin is marketed in hydrochloride form. Oxybutynin known as [α-cyclohexyl-hydroxy-benzeneaceticacid- 4-(diethyl amino)-2-butynyl ester] he US Patent No. 3,176,019 (‘019) discloses about 4-amino-2-butynol esters and their derivatives, particularly about oxybutynin hydrochloride. It also reveals about the synthesis of oxybutynin, wherein, the methyl phenyl cyclohexyl glycolate is reacted with 4-diethylamino-2-butynylacetate in presence of base to yield oxybutynin followed by further workup. Further, it is treated with 2N HCl solution to form hydrochloride salt. It is recrystallised by employing ethyl acetate or water to obtain pure oxybutynin hydrochloride. Further, the US Patent ‘019 unveils about the reaction of propargyl-2-cyclohexyl-2-hydroxy-2-phenyl acetate, /^-formaldehyde and diethyl amine in dry dioxane to obtain crude product of oxybutynin. The dry hydrogen chloride gas is passed through the ether solution of oxybutynin to yield the oxybutynin chloride as precipitate.

According to the prior art process oxybutynin is obtained as oil, which contains lot of impurities, therefore, it needs to purify high vacuum distillation. Also, the resultant oxybutynin base is having a low melting point, which may decompose during high vacuum distillation. Further, the existence of any polymorphism in oxybutynin is not disclosed in prior arts. In light of the foregoing, a need exists in the art for inventing a new form and the process thereof. Objects and Summary of the Invention

It is a principal object of the present invention is to provide a novel crystalline oxybutynin base in a solid state having improved quality.

Another object of the present invention is to provide a process for the preparation of novel crystalline oxybutynin base as a solid state. Further, object of the present invention is to provide a process for preparing an acid addition salt of oxybutynin employing crystalline oxybutynin base

In accordance with one preferred embodiment of the present invention, there is provided a crystalline oxybutynin base characterized by using different analytical tools including X-ray powder diffraction pattern, Thermo Gravimetric Analysis (TGA), and Differential Scanning Calorimetry (DSC).

Oxybutynin is used therapeutically in the treatment of intestinal hypermotility and in the treatment of urinary incontinence due to detrusor instability. Oxybutynin is sold for this purpose under the trade name of Ditropan®. Chemical names for oxybutynin are 4- (diethylamino)-2-butynyl-α-cyclohexyl-α-hydroxy benzeneacetate, and 4-(diethylamino)-2- butynylphenylcyclohexyl-glycolate. It is a racemic mixture of the R-enantiomer, R- oxybutynin, and the S-enantiomer, S-oxybutynin.

Use of the S-enantiomer of oxybutynin, S-oxybutynin, for the treatment of urinary incontinence has been described in U.S. Patent Numbers 5,532,278, and 5,736,577. The structure of S-oxybutynin (Registry Number 1 19618-22-3) is shown in formula I. S- oxybutynin is not commercially available at the present time.

 

Administration of racemic oxybutynin may result in a number of adverse effects. These adverse effects include, but are not limited to, xerostomia, mydriasis, drowsiness, nausea, constipation, palpitations and tachycardia. The amelioration of cardiovascular side effects of racemic oxybutynin, such as tachycardia and palpitations, is of particular therapeutic value.

The synthesis of S-oxybutynin has been described in the literature by Kacher et al, J. Pharmacol. Exp. Ther., 247, 867-872 (1988). An improved synthetic method is disclosed in copending U.S. patent application, serial number 09/21 1,646, the contents of which are incorporated in their entirety. In this method, an activated derivative of cyclohexylphenylglycolic acid (CHPGA), the mixed anhydride I, is prepared.

isobutylchloroforrnate

 

The mixed anhydride I is coupled with the propargyl alcohol derivative 4-N,N-diethylamino butynol (4-N,N-DEB)( III where R1 is -CH2R2; R2 is -ΝR3R4; and R3 and R4 are each ethyl.) Reaction of the optically active mixed anhydride with 4-NN-DEB produces a single enantiomer of oxybutynin, in this case, (S)-4-diethylamino-2- butynylphenylcyclohexylglycolate.

Improved syntheses of starting material CHPGA have been described in two copending U.S. Patent Applications, Serial Numbers 09/050,825 and 09/050,832. The contents of both are incorporated by reference in their entirety. In the first (09/050,825), phenylglyoxylic acid or cyclohexylglyoxylic acid is condensed with a single enantiomer of a cyclic vicinal aminoalcohol to form an ester of the phenylglyoxylic acid or the cyclohexylglyoxylic acid. The ester is reacted with an appropriate Grignard reagent to provide an α-cyclohexylphenylglycolate ester. A single diastereomer of the product ester is separated from the reaction mixture, and hydrolyzed to provide S-α- cyclohexylphenylglycolic acid (S-CHPGA). The second (09/050,832) discloses an alternate stereoselective process for preparing CHPGA. A substituted acetaldehyde is condensed with mandelic acid to provide a 5-phenyl-l,3-dioxolan-4-one, which is subsequently reacted with cyclohexanone to provide a 5-(l-hydroxy cyclohexyl)-5-phenyl-l,3-dioxolan-4-one. The product is dehydrated to a 5-(l-cyclohexenyl)-5-phenyl-l,3-dioxolan-4-one, hydrolyzed and reduced to CHPGA.

 

……………………………..

SYNTHESIS

Racemic cyclohexylphenyl glycolic acid (CHPGA) (I) is dissolved with (L)-tyrosine methyl ester (II) in refluxing acetonitrile/water to yield a mixture of diastereomeric salts, which is resolved by crystallization to afford the desired diastereomeric salt [(S)-CHPGA-(L)-TME] (III). Finally, the hydrolysis of salt (III) with HCl or H2SO4 at 40-50篊 in toluene yields the enantiomer (IV). Alternatively intermediate (IV) can be obtained as follows: acetalization of (S)-mandelic acid (V) with pivaldehyde (VI) in pentane and catalytic TfOH provides derivative (VII), which is then treated with LHMDS and then condensed with cyclohexanone (VIII) in THF to furnish aldol adduct (IX). Elimination of tertiary alcohol in (IX) with SOCl2 and pyridine in THF gives derivative (X), which is then converted into intermediate (IV) either by first hydrolysis of lactone (X) with KOH in MeOH and subsequent hydrogenation of the obtained derivative (XI) over Pd/C in MeOH, or by first hydrogenation of (X) over Pd/C in MeOH to give (XII), followed by hydrolysis with KOH in MeOH. On turn, derivative (XII) can alternatively be synthesized by treatment of derivative (VII) with LHMDS, followed by reaction with 3-bromocyclohexene (XIII) in THF to provide derivative (XIV), which is then hydrogenated over Pd/C.

US 5973182; US 6140529; WO 0023414

……………………………………………………………

 

 

The desired product is finally obtained by first formation of a mixed anhydride (XVI) by reaction of the cyclohexylphenyl glycolic acid (IV) with isobutylchloroformate (XV) in cyclohexane in the presence of Et3N, followed by treatment with 4-N,N-diethylamino butynol (XVII) (obtained on turn from reaction of propargyl alcohol (XVIII) with diethylamine (XIX) in the presence of paraformaldehyde and CuCl.

J Org Chem 2000,65(19),6283

Racemic cyclohexylphenyl glycolic acid (CHPGA) (I) is dissolved with (L)-tyrosine methyl ester (II) in refluxing acetonitrile/water to yield a mixture of diastereomeric salts, which is resolved by crystallization to afford the desired diastereomeric salt [(S)-CHPGA-(L)-TME] (III). Finally, the hydrolysis of salt (III) with HCl or H2SO4 at 40-50篊 in toluene yields the enantiomer (IV). Alternatively intermediate (IV) can be obtained as follows: acetalization of (S)-mandelic acid (V) with pivaldehyde (VI) in pentane and catalytic TfOH provides derivative (VII), which is then treated with LHMDS and then condensed with cyclohexanone (VIII) in THF to furnish aldol adduct (IX). Elimination of tertiary alcohol in (IX) with SOCl2 and pyridine in THF gives derivative (X), which is then converted into intermediate (IV) either by first hydrolysis of lactone (X) with KOH in MeOH and subsequent hydrogenation of the obtained derivative (XI) over Pd/C in MeOH, or by first hydrogenation of (X) over Pd/C in MeOH to give (XII), followed by hydrolysis with KOH in MeOH. On turn, derivative (XII) can alternatively be synthesized by treatment of derivative (VII) with LHMDS, followed by reaction with 3-bromocyclohexene (XIII) in THF to provide derivative (XIV), which is then hydrogenated over Pd/C.

……………………………………………

Tetrahedron Lett 2002,43(48),8647

The catalytic enantioselective cyanosilylation of the ketone (I) by means of Tms-CN catalyzed by gadolinium isopropoxide and the chiral ligand (II) in THF/propionitrile gives the silylated cyanohydrin (III), which is reduced by means of DIBAL in toluene to yield the carbaldehyde (IV). The desilylation of (IV) by means of HCl in aqueous THF affords the hydroxyaldehyde (V), which is finally oxidized by means of NaClO2 in tert-butanol/water to provide the target (S)-2-cyclohexyl-2-hydroxy-2-phenylacetic acid intermediate (VI) (see Scheme no. 23604001a, intermediate (IV)).

…………………………

http://www.google.com/patents/WO2009122429A2?cl=en

Example-1 Preparation of 4-diethylamino-2-butyne-ol

A mixture of para formaldehyde (105.Og), N,N-diethyl amine(300g) and copper(II) acetate (7.5g) in 1,4 dioxane (900ml) was heated to 60-65° C. After 1.5 h, 2-propyne-l-ol (150g, 2.7 moles) was added and the mixture was heated at 90-95° C. after 2 hrs; excess solvent, 1,4 dioxane, evaporated at reduced pressure to afford 315g

(84%) of the product as an oil. Example-2

Preparation of diethylamino-2-butvnylacetate

A mixture of 4-diethylamino-2-butyne-l-ol (30Og), acetic acid (600ml); acetic anhydride (300ml) and con.sulphuric acid (15ml) was heated to 65-70° C. After 2hrs.of maintenance excess solvent mixture was evaporated at reduced pressure. The residue was cooled and poured in a mixture of dichloromethane (1800ml) and DM water (3000ml).The reaction mass was saturated with sodium bicarbonate (300g) solid slowly controlling effervescences. The organic layer was separated and washed with 2% sodium bicarbonate and 1% EDTA solution to afford 318g (81%) of product as oil.

Example-3

Preparation of 4-diethylamino-2-butvnyl phenyl cvclohexyl alveolate hydrochloride (Oxybutynin Hydrochloride)

A mixture of 150g of methyl phenyl cyclohexyl glycolate, 133g of 4- diethylamino-2-butynyl acetate was dissolved in 1.8 ltr of n-heptane. The solution was added with 1.2 g of sodium methoxide. The solution was heated with stirring to a temperature of 95-100° C and distillate was collected. After 30min of maintenance at 95-100° C, the solution was cooled to 65-70° C under nitrogen. The solution was added with 3.24 g of sodium methoxide. The solution was heated with stirring to a temperature of 95-100° C and distillate was collected. After 1 hr. maintenance at 95- 100° C, reaction mass cooled to room temperature, washed with water. n-Heptane layer was separated and added 300 ml of 2N Hydrochloric acid to give oxybutynin hydrochloride. The crude was recrystallised from ethyl acetate.

Example-4 Preparation of Oxybutvnin base

A mixture of 150g of methyl phenyl cyclohexyl glycolate, 133g of 4- diethylamino-2-butynyl acetate was dissolved in 1.8 ltr of n-heptane. The solution was added with 1.2 g of sodium methoxide. The solution was heated with stirring to a temperature of 95-100° C and distillate was collected. After 30min of maintenance at 95-100° C, the solution was cooled to 65-70° C under nitrogen. The solution was added with 3.24 g of sodium methoxide. The solution was heated with stirring to a temperature of 95-100° C and distillate was collected. After 1 hr. maintenance at 95-

100° C, reaction mass cooled to room temperature, washed with ‘water. n-Heptane layer was separated, concentrated under reduced pressure to give residue. n-Pentane (250ml) was added to the residue and stirred under nitrogen atmosphere at 25-30° C. The solid product was filtered and washed with chilled n-pentane. Wet cake was dried at 40-42° C. Dry weight = 160.O g

Example-5 Preparation of Oxybutvnin (Base)

Oxybutynin chloride (lOOgm) was treated with DM water (500ml) at 25-30° C and heated to 40-45° C to observe clear solution. n-Heptane (500ml) was added to the solution and adjusted the pH of the mass to 10.0-11.0 using 5% sodium hydroxide solution at 20-25° C. Layers obtained were separated and aqueous layer was extracted with heptane. Organic layers were combined and concentrated under vacuum at 40- 45° C to, give residue. n-Pentane (250ml) was added to the residue and stirred under nitrogen atmosphere at 25-30° C. The solid product was filtered and washed with chilled n-pentane. Wet cake was dried at 40-42° C. Dry weight = 85.0 gm

………………………….

http://www.google.com/patents/US3176019

Example XIX 4-diethylamino-2-butynyl phenylcyclohexylglycolate hydrochl0ride.-A mixture of 394.2 g. of methyl phenylcyclohexylglycolate, 293.1 g. of 4-diethylamino-2-butynyl acetate was dissolved with Warming in 2.6 l. of n-heptane. The solution was heated with stirring to a temperature of 60-70 C. and 8.0 g. of sodium methoxide were added. The temperature of the mixture was then raised until the solvent began to distill. Distillation was continued at a gradual rate and aliquots of the distillate were successively collected and analyzed for the presence of methyl acetate by measurement of the refractive index. The reaction was completed when methyl acetate no longer distilled, and the refractive index observed was that of pure heptane (11 1.3855). About three and one-half hours were required for the reaction to be completed. The reaction mixture was then allowed to cool to room temperature, washed with Water, and extracted with four ml. portions of 2 N hydrochloric acid. The aqueous extracts Were combined and stirred at room temperature to permit crystallization of the hydrochloride salt of the desired product. Crystallization was completed by cooling the slurry in an ice bath, and the product was collected by filtration, pressed dry, and recrystallized from 750 ml. of water. Yield of pure crystalline material, 323 g.

……………………….

http://www.google.com/patents/EP1185498A2?cl=en

 

 

Background of the Invention Cyclohexylphenyl glycolic acid (also referred to herein as “CHPGA”) is used as a starting material for manufacturing compounds that have important biological and therapeutic activities. Such compounds include, for example, oxphencyclimine, oxyphenonium bromide, oxypyrronium bromide, oxysonium iodide, oxybutynin (4- diethylamino-2-butynyl phenylcyclohexylglycolate) and its metabolites, such as desethyloxybutynin (4-ethylamino-2-butynyl phenylcyclohexylglycolate). The important relation between stereochemistry and biological activity is well known. For example, the (S)-enantiomers of oxybutynin and desethyloxybutynin have been shown to provide a superior therapy in treating urinary incontinence, as disclosed in U.S. Patent Nos. 5,532,278 and 5,677,346. The (R) enantiomer of oxybutynin has also been suggested to be a useful drug candidate. [Noronha-Blob et al., J. Pharmacol. Exp. Ther. 256, 562-567 (1991)]. Racemic CHPGA is generally prepared by one of two methods: (1) selective hydrogenation of phenyl mandelic acid or of phenyl mandelate esters, as shown in Scheme 1; or (2) cyclohexyl magnesium halide addition to phenylglyoxylate as shown in Scheme 2. Scheme 1.

R is hydrogen or lower alkyl.

Scheme 2.

Asymmetric synthesis of individual enantiomers of CHPGA has been approached along the lines of Scheme 2, by Grignard addition to a chiral auxiliary ester of glyoxylic acid to give a diastereomeric mixture of esters. In addition, multiple step asymmetric synthesis of (R)-CHPGA from fDJ-arabinose using Grignard reagents has been reported. In general, simple primary alkyl or phenyl Grignard (or alkyllithium) reagents are used for the addition, and the addition of inorganic salts (e.g. ZnCl2) appears to increase the diastereoselectivity of the products.

As outlined in Scheme 3 below, the simple chiral ester wherein R* is the residue of a chiral alcohol, can be directly converted to chiral drugs or drug candidates by trans-esterification (R’=acetate), or hydrolyzed to yield chiral CHPGA (R’=H).

Scheme 3

esterification

(S) or (R)-Oxybutynin

(S) or (R)-CYLOHEXYLPHENYL GLYCOLIC ACID VIA RESOLUTION The resolution process of the present invention provides an inexpensive and efficient method for preparing a single enantiomer from racemic CHPGA via the formation of the diastereomeric salt with (L) or (D) -tyrosine methyl ester, also referred to herein as “(Z) or (D)-TME”. The process consists of three parts, which are depicted and described below: Part 1: Preparation of (S)-CHPGA-(Z)-TME diastereomeric salt or (R)-CHPGA-(D)-TME diastereomeric salt; Part 2:

Preparation of (S) or (R) CHPGA; and Part 3 – Recovery of (L) or (D)-tyrosine methyl ester. The ability to recover the resolving agent in high yield is an advantageous feature of the process of the invention. It greatly reduces cost by allowing recycling of the resolving agent. For ease in understanding, the diastereomeric salt, (<S)-CHPGA-(E)-TME, and the pure enantiomer (S)-CHPGA are depicted in the reactions below. However, the (R) enantiomeric series could instead be depicted and is similarly produced using the opposite enantiomer of TME.

Part 1 : Preparation of (5VCHPGA-(XVTyrosine Methyl Ester Diastereomer Salt

* ( )-TME

(S, R)-CHPGA (S)-CHPGA – (J)-TME (MW= 234.3) (MW = 429.5)

For use in the process of Part 1, the racemic starting material, (S, R)- cyclohexylphenyl glycolic acid (CHPGA) can be prepared by the process described above, i.e. (1) selective hydrogenation of phenyl mandelic acid or of phenyl mandelate esters or (2) cyclohexyl magnesium halide addition to phenylglyoxylate. Mandelic acid and phenylglyoxylic acid, also known as benzoylformic acid, are commercially available. Phenyl mandelic acid may be prepared by Grignard addition of phenyl magnesium bromide to diethyl oxalate followed by hydrolysis. The (L) enantiomer of tyrosine methyl ester is also readily available from commercial sources, as is (Z))-tyrosine, which can then be esterified to produce (_9)-tyrosine methyl ester using conventional techniques, such as acid-catalyzed esterification with methanol. The diastereomer of the present process is produced by dissolving racemic

CHPGA and an appropriate amount of an enantiomer of tyrosine methyl ester in a suitable solvent and then bringing about the insolubilization of one diastereomer. For example, racemic CHPGA and about 0.5 molar equivalents of (Z)-tyrosine methyl ester or (Z))-tyrosine methyl ester can be dissolved in a mixture of acetonitrile and water. When the solvent is about 10 wt % water in acetonitrile, solution may be achieved by heating, preferably by heating to reflux (approximately 78° C). After heating the solution for a sufficient time to achieve complete dissolution, usually about 5 minutes at reflux, followed by cooling, preferably to about 0-5° C, the diastereomeric salt (S)-CHPGA – (E)-TME or (R)-CHPGA – (£>)- TME, depending on the TME enantiomer used, crystallizes from solution. Better yields are obtained when the cooling temperature is maintained until crystallization of the diastereomer salt is complete, typically a period of about four hours. The salt crystals are then separated from the solution, for example by filtration. The crystalline product may be washed with solvent and dried. When the solvent is water/acetonitrile, drying under vacuum at about 40-50° C is effective. The mother liquor stream may be saved for later racemization and recovery of residual CHPGA. Racemization may be effected with aqueous mineral acids, particularly aqueous sulfuric acid in ethanol. Part 2: Preparation of (S.-CHPGA

(S)-CHPGA – (Z)-TME (S)-CHPGA

(MW = 429.5) (MW= 234.3)

In Part 2, the CHPGA enantiomer produced, (S) or (R)-CHPGA, is liberated from the diastereomeric salt. For the preparation of (S)-CHPGA, the (S)-CHPGA-(E)- TME salt from Part 1 is added to and dissolved to form a solution which is about 15 wt % substrate in toluene. The solution is treated with an excess of dilute mineral acid, such as 1.1 equivalents of 0.5 M HC1 or H2SO4. Upon dissolution of the diastereomeric salt, essentially all the TME enantiomer is converted to the hydrochloride salt. The diastereomeric salt mixture may be heated to about 40-50° C for about 10 minutes to facilitate dissolution of the solids. A phase split yields an aqueous solution containing (Z)-TME-HCl and an organic solution of (S)-CHPGA in toluene. The aqueous phase is separated from the organic solution and saved for recovery of the tryrosine methyl ester in Step 3 below. A common method of separation, which may be used throughout the processes described herein, is gravitational settling followed by drainage of the aqueous phase through a tap in the bottom of the reaction vessel.

The toluene organic phase containing (S)-CHPGA may be washed a second time with mineral acid, as specified above, and heated. The organic phase and aqueous phase are then separated, and the aqueous phase is discarded along with the rag layer, i.e. the layer separating the two phases. The retained toluene organic phase is then preferably concentrated, typically by vacuum distillation, to a weight that is about 2.1 to 2.3 times the weight of the diastereomeric salt originally present, followed by gradual cooling to 0-5° C to initiate crystallization of the single (S) enantiomer of CHPGA, as indicated by the formation of a thick slurry. The slurry is cooled for at least an hour to ensure that crystallization is complete, then filtered to isolate (S)-CHPGA. The (S)-CHPGA cake is then dried under vacuum while heating to a temperature of about (40-45° C).

Part 3 : Recovery of (X -Tyrosine Methyl Ester The aqueous phase containing (Z)-TME-HCl or (D)-TME-HCl saved from

Part 2 is cooled, preferably to about 0-5° C. While maintaining the cooling temperature, the aqueous solution is titrated with 0.5M NaOH to a pH of approximately 9.0. Typically, a thin slurry will form as the TME enantiomer precipitates. The TME enantiomer is isolated by filtration, washing with deionized water, and drying under vacuum at a temperature of about (40-50° C).

The resolution process of the present invention set forth above is illustrated by, but not limited to, the following example:

Example 1 Part 1 : Preparation of (S)-CHPGA-(E -Tyrosine Methyl Ester Diastereomer Salt A 2-liter reactor was charged with 100.0 g racemic CHPGA, 41.7 g (L)-

TME (0.5 equiv.), 549.2 g CH3CN, and 54.8 g deionized water. The reaction mixture was heated to reflux at approximately 78° C for about 5 min. The solution was then cooled to a temperature between 0-5° C over a period of 2 hours and remained cooling (0-5 ° C) for about 2 hours. The solution was filtered to isolate the (S)-CHPGA-(Z)-TME diastereomeric salt, and the salt cake was washed with 130 g chilled ( 0-5° C) CH3CN. The salt cake was dried in vacuo at 40-50° C , and the residual solvent remaining in the cake was < 0.5%. Yield = 77.1 g (42.1 mole %); ee > 99.0% (S).

Part 2: Preparation of .S.-CHPGA A 1000 mL reactor was charged with 77. 1 g (S)-CHPGA-(E)-TME from

Part 1, 447.0 g toluene, 339.2 g 0.5M HC1 (1.1 equiv.) and heated to 40-50°C while stirring until the solids dissolved (about 10 min). While maintaining the temperature at 40-50° C, the organic and aqueous phases separated after about 10 minutes. The phases were divided, and the aqueous (bottom) phase containing (L)- TME-HC1 was saved for recovery in Part 3 below. Approximately 370 g aqueous phase was recovered.

To the toluene organic phase an additional 169.6 g 0.5M HC1 (0.6 equiv.) were added, and the solution was heated to a temperature between 40-50° C while stirring for about 10 minutes. The toluene and aqueous phases were allowed to separate (~ 10 min.), while maintaining the temperature between 40-50° C. The phases were divided, and the aqueous (bottom) phase and rag layer were discarded. The organic phase was concentrated by vacuum distillation to a final weight of 168.0 g, then cooled to 0-5 °C over a period of about one hour during which time a thick slurry formed spontaneously. Agitation was adjusted as necessary. The slurry was cooled at 0-5 °C for an additional one hour. The slurry was filtered to recover the (S)-CHPGA. The (S)-CHPGA filter cake was dried in vacuo at 40-45° C , and the residual solvent remaining in the cake was < 0.2%. Yield = 35.8 g (85 mole %); ee > 99.0%; chemical purity (% HPLC area) > 99.0%.

Part 3: Recovery of (Z)-Tyrosine Methyl Ester

A 2-liter vessel was charged with the aqueous phase saved from Part 2 (370 g). The solution was cooled to 0-5 °C, and the cooling temperature was maintained while titrating with 0.5 M NaOH to a pH of 9.0 ±0.5 over approximately 30 min. A thin slurry formed as (Z)-TME precipitated. The slurry was filtered, and the (L)-

TME cake was washed with 154 g deionized water. The cake was dried in vacuo at 40-50°C , and the residual solvent remaining in the cake was < 1.0%. Yield = 30.5 g (E)-TME (87 mole %).

(S) OR fRVOXYBUTYNIN AND RELATED COMPOUNDS VIA DIRECT COUPLING

The synthesis of a single enantiomer of oxybutynin and oxybutynin analogs according to the present invention comprises coupling an enantiomer of cyclohexylphenyl glycolic acid with a propargyl alcohol derivative utilizing carboxylic acid activation. Optically active CHPGA may be prepared either by the resolution process described above or by asymmetric methods. The present invention also provides a process for converting the aforementioned enantiomers of oxybutynin and oxybutynin analogs to their corresponding hydrochloride salts. The synthetic process consists of two reactions, which are depicted and described below: Part 1: Formation of the Mixed Anhydride; Part 2: Formation of (S) or (R) oxybutynin and its related compounds. Again for ease in understanding, the (S) enantiomeric series is depicted, although the (R) series is produced similarly.

Part 1 : Formation of the Mixed Anhydride

isobut lchloroformate

 

(S)-CHPGA Mixed Anhydride MW=234.29

In Part 1, (S) or (R) cyclohexylphenyl glycolic acid (CHPGA) is reacted with an alkyl chloroformate in an organic solvent to form a mixed anhydride enantiomer, as shown above, which can then react to form the desired chiral product in Part 2 below.

It should be noted that, while mixed anhydrides are often employed for the synthesis of amides, their use for ester synthesis is quite unusual. It should also be noted that a surprising and unexpected aspect of the present process is that the mixed anhydride intermediate proceeds to a chiral product without affecting the tertiary carbinol of CHPGA, which would lead to impurity formation or racemization. One would expect reaction with an acyl halide at the benzylic hydroxyl resulting in the formation of a stable, but undesired compound, such as an ester. Alternatively, if the hydroxyl were activated (unintentionally) to form a good leaving group, as, for example, under acidic conditions, the dissociation of the leaving group would form a benzylic carbonium ion, leading to racemization. One would therefore expect a loss in optical activity of the oxybutynin or the extensive production of by-products. Surprisingly, the present process produces a high purity product, and no racemization is observed.

In the preparation of the mixed anhydride, two intermediates, in addition to the mixed anhydride shown above, were detected. The two were isolated and their structures were determined by NMR to be

carbonate-anhydride A carbonate-acid B wherein R5 was isobutyl. Both intermediates were smoothly converted to oxybutynin upon treatment with 4-N,N-DEB.

The reaction is preferably carried out in an inert atmosphere, such as nitrogen or argon, and the reaction solution is stirred using conventional techniques. In the depiction above, isobutyl chloroformate (IBCF) is shown as the preferred alkyl chloroformate for reaction with (S)-CHPGA forming the isobutyloxy mixed anhydride. However, other alkyl chloroformates, such as isopropenylchloroformate and 2-ethylhexylchloroformate, for example, may instead be used. The amount of alkyl chloroformate used in the reaction is preferably about 1.2 equivalents with respect to the CHPGA enantiomer.

Preferably, the reaction proceeds in the presence of a tertiary amine (2.5 equiv.), such as triethylamine (TEA), 4-N,N-dimethylaminopyridine (DMAP), pyridine, diisopropylethylamine, diethylmethylamine, Ν-methylpiperidine or Ν- methylmorpholine, which scavenges the HC1 produced. Organic solvents that may be used include, but are not limited to cyclohexane, heptane, toluene, tetrahydrofuran (THF), ethylene glycol dimethyl ether (DME), diethoxy methane (DEM), and methyl t-butyl ether (MTBE). Part 2: Formation of (S) or (R -Oxybutynin and its Analogs

Mixed Anhydride (S)-Oxybutynin or Analog

A sidechain propargyl alcohol derivative of formula (III), wherein R1 is as previously defined, is added to the mixed anhydride contained in the reaction mixture to produce the single enantiomer of oxybutynin or analog thereof (II). About 1.3 equivalents of the formula (III) compound relative to (S) or (R)-CHPGA is sufficient. Typically, the reaction mixture is heated to reflux at a temperature of about 65-80° C, but more preferably about 70-75° C, until the reaction is complete, as determined by HPLC.

Most preferably, the propargyl alcohol derivative of formula (III) is a 4- amino propargyl alcohol derivative, wherein R1 is represented as -CH2R2; R2 is – NR3R4; and R3 and R4 are each independently lower alkyl, benzyl or methoxybenzyl. For example, the compound of formula (III) is most preferably 4-N,N-diethylamino butynol (4-N,N-DEB), where R3 and R4 are each ethyl. Reaction of the mixed anhydride with 4-N.N-DEB produces the single enantiomer of oxybutynin, i.e. (S) or (R)-4-diethylamino-2-butynyl phenylcyclohexylglycolate. Another preferred embodiment is the reaction using an Ν-protected 4-N-ethylamino butynol, such as Ν-ethyl-Ν-(4-methoxybenzyl)butynol, as the propargyl alcohol derivative and then cleaving the protecting group (by methods well known in the art) to produce (S) or (R)-4-ethylamino-2-butynyl phenylcyclohexylglycolate, also known as desethyloxybutynin. In that case, R3 is ethyl, and R4 is converted to hydrogen in formula (III). Suitable protecting groups are described in Greene and Wuts Protecting Groups in Organic Synthesis. Second Edition Wiley, New York 1991, p. 362-371, which is incorporated herein by reference. In another preferred embodiment, the propargyl alcohol derivative of formula (III) is 4-N,N- ethylmethylamino butynol, which results in the formation of (Sf) or (R)-4- ethylmethylamino-2-butynyl phenylcyclohexylglycolate. In this case, R3 is ethyl, and R4 is methyl.

Other useful sidechain propargyl alcohol compounds in which R1 is -CH2R2 are those wherein R2 is azide, hydroxy, or halo. In addition, propargyl alcohol itself, also known as 2-propyn-l-ol, may be reacted with the mixed anhydride. In this case, R1 is hydrogen in formula (III). 4-N,N-Diethylamino butynol for use as the sidechain propargyl alcohol in the present invention may be prepared by reacting propargyl alcohol, paraformaldehyde, and diethylamine under standard Mannich conditions. Other amino and alkyl amino propargyl alcohol derivatives of structure (III) can be formed by the process disclosed in U.S. Patent No. 5,677,346. Briefly, a secondary amine, in which one or more substituents may be a protecting group, such as N-ethyl-4- methoxybenzenemethanamine for example, is reacted with propargyl alcohol and paraformaldehyde in the presence of cuprous chloride. After condensation with the activated CHPGA, the addition of α-chloroethyl carbonochloridate removes the protecting group. In this example, the 4-N-ethylaminobutynyl ester is the ultimate product. The remaining propargyl alcohol derivatives for use in the present invention are commercially available or can be synthesized by methods known in the art.

As stated above, the progress of the condensation of the mixed anhydride with the propargyl alcohol may be conveniently monitored by periodic HPLC analyses of the reaction mixture until the desired extent of conversion is reached. At >80% conversion, the reaction is preferably quenched by washing with 10-12% aqueous monobasic sodium phosphate and water. About 8.5 g of the phosphate per gram of enantiomeric CHPGA used is typical. After separation of the organic phase, the aqueous washes are then discarded. A final wash using deionized water may then be performed, after which the bottom aqueous phase is discarded. The retained organic phase containing the enantiomer of structure (II) in solution with the organic solvent can then be concentrated to remove most of the solvent, typically by vacuum distillation.

Formation of the Hvdrochloride Salt

(S)-Oxybutynin or Analog (S)-Oxybutynin or Analog-HCl

To promote crystallization, the organic solvent containing the enantiomer of oxybutynin or one of its analogs (II) produced by the process outlined above (also referred to herein as “free base enantiomer (II)”) is exchanged with ethyl acetate (EtOAc). Typically, the organic solvent is removed by vacuum distillation to contain about 20-25 wt % (S) or (R) enantiomer of structure (II), which is based on the theoretical amount of free base (II) formed from the coupling process above. Ethyl acetate is then added to obtain the original solution volume or weight. This step may be repeated substituting the removal of EtOAc for the organic solvent. The EtOAc solution may be filtered through a filtering agent, such as diatomaceous earth. The filter cake is washed with EtOAc as needed.

The filtrate is then concentrated by vacuum distillation, for example, to contain about 20-25 wt % (theoretical) of free base enantiomer (II) and < 0.3 wt % water. To maximize product yield and purity and to encourage crystallization, most of the water should be removed from the solution. If the foregoing concentration processes are insufficient to reduce the water to < 0.3%, the vacuum distillation may be repeated with fresh solvent or a drying agent, such as magnesium sulfate may be employed. Water content can be determined by KF (Karl Fisher method). Methyl t-butyl ethyl (MTBE) is then added to the concentrated EtOAc solution to a volume that reduces the concentration by weight of the free base enantiomer (II) by about one third, or optimally to between about 6.5 and 8.5 wt %. The hydrochloride salt is then formed by the addition of HC1, while stirring. A slight excess of HC1 in ethanol, for example about 1.1 equivalents of 35-40 wt % HC1, is generally sufficient. The temperature may be increased to 35-45° C. To initiate recrystallization, the solution may be seeded with the hydrochloride salt of the enantiomer of structure (II). After about an hour of stirring, which may be done at 35-45° C, a slurry forms. If the slurry is cooled to about 0-5° C and this temperature maintained for about two hours, filtration provides a very good recovery of the hydrochloride salt of the enantiomer of structure (II). The filter cake is typically a white to off-white crystalline solid, which can then be washed with ambient temperature methyl t-butyl ether (at least 2.2 g MTBE per gram free base enantiomer (II)), followed by vacuum drying at 40- 50° C.

The following example is illustrative, but the present invention is not limited to the embodiment described therein:

Example 2 Preparation of (S)-Oxybutvnin-HCl

A 3-neck round bottomed flask was charged with 50.0 g (S)-CHPGA (213.0 mmol) and 780 g (1000 mL) cyclohexane under nitrogen. While stirring, 54 g triethylamine (2.5 equiv.) and 35 g isobutyl chloroformate (IBCF)(1.2 equiv.) were slowly added while maintaining the temperature at 20-30° C. After about 0.5 hour, while continuing to stir the reaction mixture, 39.15 g 4-N,N-DEB (1.3 equiv.) were added, and the mixture was heated to 65 °C to reflux . Mixing continued at reflux until the formation of (S)-oxybutynin was complete by HPLC area normalization. Heating was discontinued, and the reaction mixture was cooled to between

20-30° C. At this time, 425 g of 11.5% ΝaH2PO4Η20 aqueous solution were added to the mixture, and the mixture was stirred for 10 min. Stirring was discontinued, and the organic and aqueous phases separated after about 15 minutes. The aqueous (bottom) phase was discarded. 425 g of 11.5% NaH2PO4Η20 aqueous solution were again added to the retained organic phase, and the mixture was stirred for about 10 min. The phases were then permitted to separate, which took about 15 minutes. The aqueous (bottom) phase was again discarded. To the remaining organic phase, deionized water (400 g) was added. The mixture was stirred for about 10 min, followed by phase separation after about 15 minutes. The aqueous (bottom) phase was discarded.

Cyclohexane was removed from the organic phase by vacuum distillation to about 350 g (~ 22 wt % (S)-oxybutynin based on the theoretical amount (76.29 g) of (S)-oxybutynin free base formed). Ethyl acetate (EtOAc) was added to obtain the original solution volume of about 1000 mL (or about 83 Og), followed by vacuum distillation to 20-25 wt % (S)-oxybutynin. EtOAc was then added a second time to a volume of about 1000 mL (or about 830g). The batch was then polish filtered through about 5.0 g CELITE® while washing the filter cake with EtOAc as needed. The filtered mixture was concentrated and dried by vacuum distillation to 339 g (~ 22.5 wt % (S)-oxybutynin) and < 03 wt % water, as measured by KF. Based on the theoretical amount of (S)-oxybutynin free base (76.29 g), methyl t-butyl ether was added to adjust the (S)-oxybutynin free base concentration to 8.0 wt % (953 g). With agitation, 23 g of 37 wt % HC1 in EtOH (1.1 equiv.) were slowly added to the solution, while maintaining the temperature between 20 and 45° C. The temperature of the solution was then adjusted to 35-45° C, and the solution was seeded with about 500 mg (S)-oxybutynin-HCl crystals (approximately 10 mg of seeds per g (S)-CHPGA ). The temperature was maintained, and the solution was stirred for about one hour. A slurry formed, which was then cooled to 0-5 °C over a minimum of 1 hour and held for 2 hours. The slurry was then filtered to recover the (S)-oxybutynin-HCl. The filter cake was a white to off-white crystalline solid. After washing with MTBE (a minimum of 167.84 g MTBE (2.2 g MTBE/g (S)-oxybutynin free base), the cake was dried in vacuo at 40-45 °C. The residual solvent remaining in the cake was < 0.5%. Dry weight = 57.9 g. Overall yield = 68.9%.

Example 3 Isolation of the two carbonate intermediates A and B To a racemic mixture of cyclohexylphenylglycolic acid [CHPGA] (5.0 g, 0.0213mol) in cyclohexane (100 mL) was added triethylamine (7.4 mL, 0.053 mol) and isobutylchloroformate (5.5mL, 0.0426mol). The slurry was allowed to stir at ambient temperature for approximately 0.5 h, at which time the reaction was quenched with a 10% aq. NaH2PO4 (50 ml). The organic phase was separated from the aqueous phase and washed with 10% aq. NaH2PO4 (50mL) followed by DI water (50mL). The organic phase was dried over anhydrous MgSO4 and concentrated in vacuo to afford a colorless oil. The product was purified by flash chromatography eluting with 95:5 hexane-EtOAc [Rf = 0.2] to afford pure carbonate-anhydride A. The structure was confirmed by H and 13C NMR, IR, in sttw IR and MS.

To a racemic mixture of cyclohexylphenylglycolic acid [CHPGA] (5.0 g, 0.0213 mol) in cyclohexane (100 mL) was added triethylamine (7.4 mL, 0.053mol) or preferably 1-methyl piperidine (O.053mol), and isobutylchloroformate (3.3 mL, 0.026mol). The slurry was allowed to stir at ambient temperature for approximately 0.5 h, at which time the reaction was quenched with a 10% aq. solution of NaH2PO4 (50 mL) followed by DI water (50mL). The organic phase was dried over anhydrous MgSO4 and concentrated in vacuo to afford a colorless oil as a 4:1 mixture of A and B by HPLC. The crude product was purified by passing the mixture through a plug of neutral alumina. Compound A was eluted first using

CHClj B was then washed off the alumina with acetone and concentrated in vacuo to afford pure carbonate-acid B. The structure was confirmed by H and 13C NMR, IR, in situ IR and MS.

 

………………………………………….

http://www.google.com/patents/US6294582

The synthesis of S-oxybutynin has been described in the literature by Kacher et al., J. Pharmacol. Exp. Ther., 247, 867-872 (1988). An improved synthetic method is disclosed in copending U.S. patent application, Ser. No. 09/211,646, now U.S. Pat. No. 6,140,529, the contents of which are incorporated in their entirety. In this method, an activated derivative of cyclohexylphenylglycolic acid (CHPGA), the mixed anhydride I, is prepared.

The mixed anhydride I is coupled with the propargyl alcohol derivative 4-N,N-diethylamino butynol (4-N,N-DEB)(III where R1 is —CH2R2; R2 is —NR3R4; and R3 and R4 are each ethyl.) Reaction of the optically active mixed anhydride with 4-N,N-DEB produces a single enantiomer of oxybutynin, in this case, (S)-4-diethylamino-2-butynylphenylcyclohexylglycolate.

Improved syntheses of starting material CHPGA have been described in two copending U.S. patent applications, Ser. No. 09/050,825, now U.S. Pat. No. 6,013,830, and 09/050,832. The contents of both are incorporated by reference in their entirety. In the first (09/050,825, now U.S. Pat. No. 6,013,830), phenylglyoxylic acid or cyclohexylglyoxylic acid is condensed with a single enantiomer of a cyclic vicinal aminoalcohol to form an ester of the phenylglyoxylic acid or the cyclohexylglyoxylic acid. The ester is reacted with an appropriate Grignard reagent to provide an a-cyclohexylphenylglycolate ester. A single diastereomer of the product ester is separated from the reaction mixture, and hydrolyzed to provide S-α-cyclohexylphenylglycolic acid (S-CHPGA). The second (09/050,832) discloses an alternate stereoselective process for preparing CHPGA. A substituted acetaldehyde is condensed with mandelic acid to provide a 5-phenyl-1,3-dioxolan-4-one, which is subsequently reacted with cyclohexanone to provide a 5-(1-hydroxy cyclohexyl)-5-phenyl-1,3-dioxolan-4-one. The product is dehydrated to a 5-(1-cyclohexenyl)-5-phenyl-1,3-dioxolan-4-one, hydrolyzed and reduced to CHPGA.

The magnitude of a prophylactic or therapeutic dose of S-oxybutynin in the acute or chronic management of disease will vary with the severity of the condition to be treated, and the route of administration. The dose, and perhaps the dose frequency will also vary according to the age, body weight, and the response of the individual patient. In general, the daily dose ranges when administered by inhalation, for the conditions described herein, are from about 0.1 mg to about 100 mg in single or divided dosages. Preferably a daily dose range should be between about 10 mg to about 25 mg, in single or divided dosages, preferably in from 2-4 divided dosages. In managing the patient the therapy should be initiated at a lower dose, perhaps from 5 mg to about 10 mg, and increased up to about 2×20 mg or higher depending on the patient’s global response. When administered orally, preferably as a soft elastic gelatin capsule, the preferred dose range is from about 1 mg to about 1 g per day, more preferably, from about 25 mg to about 700 mg per day, and most preferably, from about 100 mg to about 400 mg per day. It is further recommended that children and patients over 65 years and those with apaired renal, or hepatic function, initially receive low dosages and that they be titrated based on individual responses and blood levels. It may be necessary to use dosages outside these ranges in some cases, as will be apparent to those skilled in the art. Further, it is noted that the clinician or treating physician possesses knowledge of how and when to interrupt, adjust, or terminate therapy in conjunction with individual patient response. The terms “a therapeutically effective quantity”, and “a quantity sufficient to alleviate bronchospasms” are encompassed by the above described dosage amounts and dose frequency schedule.

The methods of the present invention utilize S-oxybutynin, or a pharmaceutically acceptable salt thereof. The term “pharmaceutically acceptable salt” or “a pharmaceutically acceptable salt thereof” refer to salts prepared from pharmaceutically acceptable nontoxic acids including both inorganic and organic acids. Suitable pharmaceutically acceptable acid addition salts for the compound of the present invention include acetic, benzenesulfonic (besylate), benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, and p-toluene sulfonic. The hydrochloride has particular utility.

Preferred unit dosage formulations are those containing an effective dose, as recited, or an appropriate fraction thereof, of S-oxybutynin or pharmaceutically acceptable salts thereof. The formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question. For example, formulations for oral administration may include carriers such as starches, sugars, microcystalline cellulose, diluents, granulating agents, flavoring agents and the like. Formulations suitable for oral, rectal and parenteral administration (including subcutaneous, transdermal, intramuscular, and intravenous) and inhalation may be used for treatment according to the present invention.

Any suitable route of administration may be employed for providing the patient with an effective dosage of S-oxybutynin. For example, oral, rectal, parenteral (subcutaneous, intramuscular, intravenous), transdermal, and like forms of administration may be employed. Transdermal administration may be improved by the inclusion of a permeation enhancer in the transdermal delivery device, for example as described in PCT application WO 92/20377. Dosage forms include troches, dispersions, suspensions, solutions, aerosols, patches, syrups, tablets and capsules, including soft elastic gelatin capsules. Oral and parenteral sustained release dosage forms may also be used.

Because of their ease of administration, tablets and capsules represent one of the more advantageous oral dosage unit forms, in which case solid pharmaceutical carriers are employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques. Soft elastic gel capsules are a preferred form of administration of S-oxybutynin.

Soft elastic gelatin capsules may be prepared by mixing S-oxybutynin with a digestible oil such as soybean oil, lecithin, cottonseed oil, or olive oil. The mixture is then injected into gelatin by means of a positive pressure pump, such that each dosage unit contains an effective dose of S-oxybutynin. The capsules are subsequently washed and dried.

Oral syrups, as well as other oral liquid formulations, are well known to those skilled in the art, and general methods for preparing them are found in most standard pharmacy school textbooks. An exemplary source is Remington: The Science and Practice of Pharmacy. Chapter 86 of the 19th edition of Remington entitled “Solutions, Emulsions, Suspensions and Extracts” describes in complete detail the preparation of syrups (pages 1503-1505) and other oral liquids. Similarly, sustained release formulation is well known in the art, and Chapter 94 of the same reference, entitled “Sustained-Release Drug Delivery Systems”, describes the more common types of oral and parenteral sustained-release dosage forms (pages 1660-1675.) The relevant disclosure, Chapters 86 and 94, is incorporated herein by reference.

Controlled release means and delivery devices are also described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, and in PCT application WO 92/20377. Because they reduce peak plasma concentrations, controlled release dosage forms are particularly useful for providing a therapeutic plasma concentration of S-oxybutynin while avoiding the side effects associated with peak plasma concentrations.

Formulations suitable for inhalation include sterile solutions for nebulization comprising a therapeutically effective amount of S-oxybutynin or a pharmaceutically acceptable salt thereof, dissolved in aqueous saline solution and optionally containing a preservative such as benzalkonium chloride or chlorobutanol, and aerosol formulations comprising a therapeutically effective amount of S-oxybutynin, or a pharmaceutically acceptable salt thereof, dissolved or suspended in an appropriate propellant (e.g., HFA-134a, HFA-227, or a mixture thereof, or a chlorofluorocarbon propellant such as a mixture of Propellants 11, 12 and/or 114) optionally containing a surfactant. Aerosols may be conveniently presented in unit dosage form and prepared by any of the methods well-known in the art of pharmacy. The preparation of a particularly desirable aerosol formulation is described in European Patent No. 556239, the disclosure of which is incorporated herein by reference. Also suitable are dry powder formulations comprising a therapeutically effective amount of S-oxybutynin or a pharmaceutically acceptable salt thereof, blended with an appropriate carrier and adapted for use in connection with a dry-powder inhaler.

 

……………………….

CZ20013826A3 * Title not available
US3176019 * Jun 20, 1961 Mar 30, 1965 Mead Johnson & Co Substituted aminobutynyl acetates
* Cited by examiner
Non-Patent Citations
Reference
1 * DATABASE CAPLUS [Online] 13 November 2010 STN Database accession no. 2006:220682 & CZ 20 013 826 A3 18 June 2003
2 * DATABASE CAPLUS 13 January 2010 STN: ‘Syntheses of oxybutynin hydrochloride‘ Database accession no. 1997:395370 & ZHONGGUO YIYAO GONGYE ZAZHI vol. 27, no. 9, pages 387 – 389

 

………………………….

Racemic

Oxybutynin
Oxybutynin2DCSD.svg
Oxybutynin 3d balls.png
Systematic (IUPAC) name
4-Diethylaminobut-2-ynyl 2-cyclohexyl-2-hydroxy-2-phenylethanoate
Clinical data
Trade names Ditropan
AHFS/Drugs.com monograph
MedlinePlus a682141
Pregnancy cat.
Legal status
Routes oral, transdermal gel, transdermal patch
Pharmacokinetic data
Protein binding 91%-93%
Half-life 12.4-13.2 hours
Identifiers
CAS number 5633-20-5 Yes
ATC code G04BD04
PubChem CID 4634
IUPHAR ligand 359
DrugBank DB01062
ChemSpider 4473 Yes
UNII K9P6MC7092 Yes
KEGG D00465 Yes
ChEBI CHEBI:7856 Yes
ChEMBL CHEMBL1231 Yes
Chemical data
Formula C22H31NO3 
Mol. mass 357.486 g/mol

Oxybutynin (Ditropan, Lyrinel XL, Lenditro (South Africa)) is an anticholinergic medication used to relieve urinary and bladder difficulties, including frequent urination and inability to control urination (urge incontinence), by decreasing muscle spasms of the bladder.[1]

It competitively antagonizes the M1, M2, and M3 subtypes of the muscarinic acetylcholine receptor. It also has direct spasmolytic effects on bladder smooth muscle as a calcium antagonist and local anesthetic, but at concentrations far above those used clinically.

Oxybutynin is also a possible treatment of hyperhidrosis (hyper-active sweating).[2][3][4]

Chemistry

Oxybutynin contains one stereocenter. Commercial formulations are sold as the racemate. The (R)-enantiomer is a more potent anticholinergic than either the racemate or the (S)-enantiomer, which is essentially without anticholinergic activity at doses used in clinical practice.[5][6] However, (R)-oxybutynin administered alone offers little or no clinical benefit above and beyond the racemic mixture. The other actions (calcium antagonism, local anesthesia) of oxybutynin are not stereospecific. (S)-Oxybutynin has not been clinically tested for its spasmolytic effects, but may be clinically useful for the same indications as the racemate, without the unpleasant anticholinergic side effects.

Clinical efficacy

In two trials of patients with overactive bladder, transdermal oxybutynin 3.9 mg/day decreased the number of incontinence episodes and increased average voided volume to a significantly greater extent than placebo. There was no difference in transdermal oxybutynin and extended-release oral tolterodine.[7]

Adverse effects

Common adverse effects associated with oxybutynin and other anticholinergics include: dry mouth, difficulty in urination, constipation, blurred vision, drowsiness, and dizziness.[8] Anticholinergics have also been known to induce delirium.[9]

These are dose-related and sometimes severe. In one population studied—after six months, more than half of the patients had stopped taking the medication because of side effects and calcium defects. An intake of calcium of 800 to 1000 mg is suggested.Dry mouth may be particularly severe; one estimate is that over a quarter of patients who begin oxybutynin treatment may have to stop because of dry mouth.

N-Desethyloxybutynin is an active metabolite of oxybutynin that is thought responsible for much of the adverse effects associated with the use of oxybutynin.[10] N-Desethyloxybutynin plasma levels may reach as much as six times that of the parent drug after administration of the immediate-release oral formulation.[11] Alternative dosage forms have been developed in an effort to reduce blood levels of N-desethyloxybutynin and achieve a steadier concentration of oxybutynin than is possible with the immediate release form. The long-acting formulations also allow once-daily administration instead of the twice-daily dosage required with the immediate-release form. The transdermal patch, in addition to the benefits of the extended-release oral formulations, bypasses the first-pass hepatic effect that the oral formulations are subject to.[12] In those with overflow incontinence because of diabetes or neurological diseases like multiple sclerosis or spinal cord trauma, oxybutynin can worsen overflow incontinence since the fundamental problem is that the bladder is not contracting.

Clinical pharmacology

Oxybutynin chloride exerts direct antispasmodic effect on smooth muscle and inhibits the muscarinic action of acetylcholine on smooth muscle. It exhibits one-fifth of the anticholinergic activity of atropine on the rabbit detrusor muscle, but four to ten times the antispasmodic activity. No blocking effects occur at skeletal neuromuscular junctions or autonomic ganglia (antinicotinic effects).

Sources say the drug is absorbed within one hour and has an elimination half-life of 2 to 5 hours.[13][14][15] There is a wide variation among individuals in the drug’s concentration in blood. This, and its low concentration in urine, suggest that it is eliminated through the liver.[14]

Contraindications

Oxybutynin chloride is contraindicated in patients with untreated angle closure glaucoma, and in patients with untreated narrow anterior chamber angles—since anticholinergic drugs may aggravate these conditions. It is also contraindicated in partial or complete obstruction of the gastrointestinal tract, hiatal hernia, gastroesophageal reflux disease, paralytic ileus, intestinal atony of the elderly or debilitated patient, megacolon, toxic megacolon complicating ulcerative colitis, severe colitis, and myasthenia gravis. It is contraindicated in patients with obstructive uropathy and in patients with unstable cardiovascular status in acute hemorrhage. Oxybutynin chloride is contraindicated in patients who have demonstrated hypersensitivity to the product.

Formulations

It is available orally in generic formulation or as the brand-names Ditropan, Lyrinel XL, or Ditrospam, as a transdermal patch under the brand name Oxytrol, and as a topical gel under the brand name Gelnique.

A 2009 Weill Cornell Medical College study concluded that patients switched to generic oxybutynin experienced a degradation in therapeutic value: “In women, there was a doubling of daytime frequency of urination, a slight 20% increase in nocturia, and a 46.3% increase in urge incontinence. In men, there was a 2.4-fold increase in daytime frequency, a 40% increase in nocturia, and a 40.6% increase in urge incontinence”.[16]

References

  1. Chapple CR. “Muscarinic receptor antagonists in the treatment of overactive bladder”. Urology (55)5, Supp. 1:33-46, 2000.
  2. Tupker RA, Harmsze AM, Deneer VH (2006). “Oxybutynin therapy for generalized hyperhidrosis.”. Arch Dermatol 142 (8): 1065–6. doi:10.1001/archderm.142.8.1065. PMID 16924061.
  3. Mijnhout GS, Kloosterman H, Simsek S, Strack van Schijndel RJ, Netelenbos JC. (2006). “Oxybutynin: dry days for patients with hyperhidrosis.”. Neth J Med 64 (9): 326–8. PMID 17057269.
  4. Schollhammer M, Misery L. (2007). “Treatment of hyperhidrosis with oxybutynin.”. Arch Dermatol. 143 (4): 544–5. doi:10.1001/archderm.143.4.544. PMID 17438194.
  5. Kachur JF, et al. “R and S enantiomers of oxybutynin: pharmacological effects in guinea pig bladder and intestine.” Journal of Pharmacology and Experimental Therapeutics 247:867-72, 1988.
  6. Noronha-Blob L, Kachur JF. “Enantiomers of oxybutynin: in vitro pharmacological characterization at M1, M2 and M3 muscarinic receptors and in vivo effects on urinary bladder contraction, mydriasis and salivary secretion in guinea pigs.” Journal of Pharmacology and Experimental Therapeutics 256:562-7, 1991.
  7. Baldwin C, Keating GM.[1].Drugs 2009;69 (3):327-337. doi:10.2165/00003495-200969030-00008.
  8. Mehta D (Ed.) 2006. British National Formulary 51. Pharmaceutical Press. ISBN 0-85369-668-3
  9. Andreasen NC and Black DW, “Introductory Textbook of Psychiatry.” American Psychiatric Publishing Inc. 2006
  10. Allen B. Reitz, Suneel K. Gupta, Yifang Huang, Michael H. Parker, and Richard R. Ryan (2007). “The preparation and human muscarinic receptor profiling of oxybutynin and N-desethyloxybutynin enantiomers”. Med Chem 3 (6): 543–5. doi:10.2174/157340607782360353. PMID 18045203.
  11. Zobrist RH, et al. “Pharmacokinetics of the R- and S-Enantiomers of Oxybutynin and N-Desethyloxybutynin Following Oral and Transdermal Administration of the Racemate in Healthy Volunteers”. Pharmaceutical Research 18:1029-1034, 2001.
  12. Oki T, et al. “Advantages for Transdermal over Oral Oxybutynin to Treat Overactive Bladder: Muscarinic Receptor Binding, Plasma Drug Concentration, and Salivary Secretion”. Journal of Pharmacology and Experimental Therapeutics Fast Forward 316:1137-1145, 2006.
  13. [2] “Oxybutynin” Retrieved on 30 August 2012.
  14. [3] “The pharmacokinetics of oxybutynin in man. (Abstract)” Retrieved on 30 August 2012.
  15. [4] “Oxybutynin” Retrieved on 30 August 2012.
  16. Kerr, Martha (2009-05-03). “AUA 2009: Generics Not Equal to Brand-Name Drugs for Overactive Bladder”. American Urological Association (AUA) 104th Annual Scientific Meeting (Medscape). Retrieved 2013-04-20.

External links

Title: OxybutyninCAS Registry Number: 5633-20-5CAS Name: a-Cyclohexyl-a-hydroxybenzeneacetic acid 4-(diethylamino)-2-butynyl esterAdditional Names: a-phenylcyclohexaneglycolic acid 4-(diethylamino)-2-butynyl ester; 4-diethylamino-2-butynyl phenylcyclohexylglycolate; oxibutininaMolecular Formula: C22H31NO3Molecular Weight: 357.49Percent Composition: C 73.91%, H 8.74%, N 3.92%, O 13.43%Literature References: Muscarinic receptor antagonist. Prepn: GB 940540 (1963 to Mead Johnson). Physico-chemical properties: E. Miyamoto et al., Analyst 119, 1489 (1994). GC-MS determn in plasma: K. S. Patrick et al., J. Chromatogr. 487, 91 (1989). Toxicity: E. I. Goldenthal, Toxicol. Appl. Pharmacol. 18, 185 (1971). Review of pharmacodynamics and therapeutic use: Y. E. Yarker et al., Drugs Aging 6, 243-262 (1995).Properties: pKa 8.04. Log P (n-octanol/water): 2.9 (pH 6). Soly in water (mg/ml): 77 (pH 1); 0.8 (pH 6); 0.012 (pH >9.6).pKa: pKa 8.04Log P: Log P (n-octanol/water): 2.9 (pH 6) 

Derivative Type: Hydrochloride

CAS Registry Number: 1508-65-2

Additional Names: Oxybutynin chloride

Manufacturers’ Codes: MJ-4309-1

Trademarks: Cystrin (Sanofi-Synthelabo); Ditropan (Sanofi-Synthelabo); Dridase (Sanofi-Synthelabo); Kentera (UCB); Pollakisu (Kodama); Tropax (BMS)

Molecular Formula: C22H31NO3.HCl

Molecular Weight: 393.95

Percent Composition: C 67.07%, H 8.19%, N 3.56%, O 12.18%, Cl 9.00%

Properties: Crystals, mp 129-130°. Sol in water, acids. Practically insol in alkali. LD50 orally in rats: 1220 mg/kg (Goldenthal).

Melting point: mp 129-130°

Toxicity data: LD50 orally in rats: 1220 mg/kg (Goldenthal)

 

Therap-Cat: In treatment of urinary incontinence.

Keywords: Antimuscarinic.

Share
Follow

Get every new post on this blog delivered to your Inbox.

Join other followers: