AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

Ultrasonic Assisted Extraction of Artemisinin from Artemisia Annua L. Using Monoether based Solvents

 PROCESS  Comments Off on Ultrasonic Assisted Extraction of Artemisinin from Artemisia Annua L. Using Monoether based Solvents
Dec 252017
 

Image result for Artemisia Annua L

Artemisia Annua L

Ultrasonic Assisted Extraction of Artemisinin from Artemisia Annua L. Using Monoether based Solvents

Abstract

Artemisinin is a kind of natural antimalarial drug exhibiting low toxicity with a very fast action against malaria. Solvent extraction is the most widely used method to separate artemisinin from the Chinese medicinal herb Artemisia annua L. In this study, a series of monoether based solvents have been proposed to extract artemisinin and propylene glycol methyl ether (PGME) was found to be the most appropriate one for this extraction. Ultrasonic irradiation was demonstrated to be able to assist artemisinin extraction. Influences of extraction conditions, including liquid/solid ratio, extraction temperature, ultrasonic time, ultrasonic power, on the extraction efficiency were discussed by single factor experiments, and the main influence factors were optimized by responds surface method. The extraction mechanism was explored with spectroscopic characterizations, and kinetics of this process was also studied. Results indicate that ultrasonic assisted extraction using PGME has faster extraction rate than conventional solvents, and ultrasonic can significantly enhance mass transfer. Compared with conventional extraction, the process developed here exhibited higher efficiency (13.79 mg/g vs. 13.29 mg/g) and short extraction time (decreased from 8 h to 0.5 h) at a relatively low temperature. In addition, PGME has low toxicity and volatility, making the extraction process more safe and reliable. Therefore, this proposed method demonstrates that PGME based ultrasonic assisted extraction is a rapid, efficient, simple and safe technique for natural product extraction.

http://pubs.rsc.org/en/Content/ArticleLanding/2017/GC/C7GC03191B?utm_source=feedburner&utm_medium=feed&utm_campaign=Feed%3A+rss%2FGC+%28RSC+-+Green+Chem.+latest+articles%29#!divAbstract

/////////// Artemisinin,  Artemisia Annua L, extraction

Share

Commercial Production of Semi-Synthetic Artemisinin

 Uncategorized  Comments Off on Commercial Production of Semi-Synthetic Artemisinin
Aug 072016
 

STR1

Figure 1. Production of artemisinic acid or β-farnesene by engineered yeast. The sesquiterpene alkenes β-farnesene and amorphadiene are both derived from FPP (farnesyl diphosphate) by the action of specific enzymes introduced from plants: amorphadiene synthase (ADS) generates amorphadiene and β-farnesene synthase (FS) generates β-farnesene. Production strains express either ADS or FS, not both. Oxidation of amorphadiene to artemisinic acid is accomplished by the action of five plant enzymes expressed in the engineered yeast.17 Conversion of purified artemisinic acid to artemisinin is accomplished by in vitro organic chemistry. Isoprenoid production strains make little ethanol.

The antimalarial drug artemisinin and the specialty chemical β-farnesene are examples of natural product isoprenoids that can help solve global challenges, but whose usage has previously been limited by supply and cost impediments. This review describes the path to commercial production of these compounds utilizing fermentation of engineered yeast. Development of commercially viable yeast strains was a substantial challenge that was addressed by creation and implementation of an industrial synthetic biology pipeline. Using the engineered strains, production of β-farnesene from Brazilian sugarcane offers several environmental advantages. Among the many commercial applications of β-farnesene, its use as a feedstock for making biodegradable lubricants is highlighted. This example, along with others, highlight a powerful new suite of technologies that will become increasingly important for production of chemicals, spanning from pharmaceuticals through commodity chemicals.

STR1

Figure 2. Sanofi industrial semi-synthesis of artemisinin. The process starts with a moderate pressure catalytic diastereoselective hydrogenation of artemisinic acid to produce a high (95:5) ratio of the desired (R)-isomer. To avoid formation of a lactone byproduct, dihydro-epi-deoxyarteannuin B, during the photooxidation, the carboxylic acid is protected as a mixed anhydride. The final step combines formation of the intermediate hydroperoxide via photoxidation using a Hg vapor lamp and commercially available tetraphenylporphyin (TPP) as sensitizer with a Hock cleavage and rearrangement catalyzed by trifluoroacetic acid to give, after workup, the best yield reported to date of pure isolated artemisinin (55%).

Synthetic Biology and the Development of Commercial β-Farnesene Production Strains Semi-synthetic artemisinin is a pharmaceutical with a price point comparable to plant-derived artemisinin,20 namely above $150 per kg. β-Farnesene, however, is a specialty chemical with multiple uses (more details below); most specialty and commodity chemicals have significantly lower price points, often below $10 per kg. For these product categories, it is of paramount importance that fermentative production be as efficient as possible, with high yields (namely, grams of product made per gram of feed substrate), productivities (grams of product/liter of culture/hour) and concentration (also known as titer; grams of product per liter of culture). Developing yeast strains capable of the yield, productivity and titer required for chemical production requires extensive development, and has been enabled over the last decade by the new discipline of synthetic biology. Synthetic biology seeks to extend approaches and concepts from engineering and computation to redesign biology for a chosen function;21recent advances in the application of design automation, i.e., the use of software, hardware and robotics22 have enabled the creation and screening of hundreds of thousands of strain variants (created by both design and random mutagenesis) for the properties required for commercial production of β-farnesene. Notable enabling technologies developed for routine usage include rapid and reliable assembly of large (i.e., multiple kilobase) deoxyribonucleic acid (DNA) constructs;23-25 high throughput, cost effective, verification of structural DNA assemblies by both initial restriction digest26 and by low-cost DNA sequencing;27 and whole genome sequencing of yeast strains.28 In addition, there is a need to effectively identify the best new strains (akin to panning for gold!) through high throughput, rapid, and accurate methods to screen thousands of strains. Further, the results of small-scale (< 1 milliliter) tests must correspond to the results of large-scale (> 50,000 liter) production. Development and implementation of these technologies required considerable investment by Amyris. The outcome is a robust pipeline for efficient, cost-effective strain generation allied with screening for the properties required for commercial production of β-farnesene by fermentation (i.e., at a price point required for its use as a specialty chemical).

As the world’s population and economies grow, the demand for a wide variety of specialty, commodity, and pharmaceutical chemicals will outpace the supply available from current sources. There is an urgent need to develop alternative, sustainable sources of many existing chemicals and to develop abundant sources of currently scarce chemicals with novel beneficial properties. Synthetic biology and industrial fermentation, combined with synthetic chemistry, will be an increasingly important source of chemicals in the decades ahead; artemisinin and β-farnesene provide good examples of this relatively new approach to chemical production. Brazil’s plentiful sugar cane feedstock and fermentation expertise make it an excellent location for this type of manufacturing, which can expand and diversify the nation’s industrial base and international importance.

J. Braz. Chem. Soc. 2016, 27(8), 1339-1345

Developing Commercial Production of Semi-Synthetic Artemisinin, and of β-Farnesene, an Isoprenoid Produced by Fermentation of Brazilian Sugar

Kirsten R. Benjamin; Iris R. Silva; João P. Cherubim; Derek McPhee; Chris J. Paddon

How to cite this article

Genes encoding the biosynthetic pathway for production of a valuable product (e.g., farnesene) in a native organism are expressed in a heterologous microbial host (e.g., yeast). The engineered yeast produces farnesene by commercial fermentation. Copyright © 2016 Amyris, inc. All rights reserved.

http://dx.doi.org/10.5935/0103-5053.20160119

http://jbcs.sbq.org.br/imagebank/pdf/v27n8a04.pdf

Benjamin KR, Silva IR, Cherubim JP, Mcphee D, Paddon CJ. Developing Commercial Production of Semi-Synthetic Artemisinin, and of β-Farnesene, an Isoprenoid Produced by Fermentation of Brazilian Sugar. J. Braz. Chem. Soc. 2016;27(8):1339-1345

Kirsten R. Benjamin,a Iris R. Silva,b João P. Cherubim,c Derek McPheea and Chris J. Paddon*,a a Amyris, Inc., 5885 Hollis Street, Suite 100, CA 94608 Emeryville, USA b Amyris Brasil Ltda, Rua John Dalton 301-Bloco B-Edificio 3, Condominio Techno Plaza, 13069-330 Campinas-SP, Brazil c Amyris Brasil Ltda, Rodovia Brotas/Torrinha-km 7.5, 17380-000 Brotas-SP, Brazil

*e-mail: paddon@amyris.com
Chris Paddon

Chris Paddon, PhD

Dr. Paddon has a PhD in Biochemistry from Imperial College, London, but now considers himself a synthetic biologist. After postdoctoral work at the National Institutes of Health in Bethesda, MD, he worked in the pharmaceutical industry (GlaxoSmithKline), and then for two Bay Area biopharmaceutical companies (Affymax and Xenoport) before joining Amyris, Inc. in 2005 as its sixth employee and first scientist. He was project leader for the semi-synthetic artemisinin project at Amyris, Inc. and has subsequently led a number of other projects and programs there.

Chris Paddon is a Principal Scientist at Amyris, Inc. in Emeryville, CA. He was project leader for the Semi-Synthetic Artemisinin project, and subsequently led a number of projects at Amyris using synthetic biology for the production of natural products. He received his Bachelor’s degree in Microbiology from The University of Surrey (UK), and doctorate in Biochemistry from Imperial College (London, UK). Following postdoctoral work at The National Institutes for Health (Bethesda, MD) he joined the pharmaceutical industry, working for GSK (London, UK). He subsequently worked for Affymax (Palo Alto, CA) and Xenoport (Santa Clara, CA) before joining Amyris.

//////////// Commercial Production, Semi-Synthetic , Artemisinin,  farnesene, fermentation, natural product, lubricant

Share

New Patent, WO 2016110874, Artemisinin , IPCA Laboratories Ltd

 PATENTS, Uncategorized  Comments Off on New Patent, WO 2016110874, Artemisinin , IPCA Laboratories Ltd
Jul 182016
 

 

New Patent, WO 2016110874, Artemisinin , IPCA Laboratories Ltd

FOR Cancer; Parasitic infection; Plasmodium falciparum infection; Viral infection

WO-2016110874

KUMAR, Ashok; (IN).
SINGH, Dharmendra; (IN).
MAURYA, Ghanshyam; (IN).
WAKCHAURE, Yogesh; (IN)

 

Dr. Ashok Kumar, President – Research and Development (Chemical) at IPCA LABORATORIES LTD

IPCA LABORATORIES LIMITED [IN/IN]; 48, Kandivli Industrial Estate, Charkop, Kandivali (West), Mumbai 400067 (IN)

Novel process for preparing artemisinin or its derivatives such as dihydroartemisinin, artemether, arteether and artesunate. Also claims novel intermediates of artemesinin such as artemisinic acid or dihydroartemisinic acid. Discloses the use of artemisinin or its derivatives, for treating malaria, cancer, viral and parasitic infections.

In July 2016, Newport Premium™ reported that IPCA was capable of producing commercial quantities of artemether, arteether and artesunate; and holds an inactive US DMF for artemether since February 2009. In July 2016, IPCA’s website lists artemether, arteether and artesunate under its products and also lists artemether and artesunate as having EDMF and WHO certificates. The assignee also has Canada HPFB certificate for artemether.

The Central Drug Research Institute (CDRI) in collaboration with IPCA is developing CDRI-97/78 (1,2,4 trioxane derivative), a synthetic artemisinin substitute for treating drug resistant Plasmodium falciparum infection. In July 2016, CDRI-97/78 was reported to be in phase 1 clinical development. IPCA in collaboration with CDRI was also investigating CDRI-99/411, a synthetic artemisinin substitute for treating malaria; but its development had been presumed to have been discontinued; however, this application’s publication would suggest otherwise.

Writeup

Artemisinin is an active phytoconstituent of Chinese medicinal herb Artemisia annua, useful for the treatment of malaria. Generally, artemisinin/artemisinic acid is obtained by extraction of the plant, Artemisia annua. The plant Artemisia annua was first mentioned in an ancient Chinese medicine book written on silk in the West Han Dynasty at around 200 B.C. The plant’s anti-malarial application was first described in a Chinese pharmacopeia, titled “Chinese Handbook of Prescriptions for Emergency Treatments,” written at around 340 A.D.

Artemisinin being poorly bioavailable limits its effectiveness. Therefore semisynthetic derivatives of artemisinin such as artesunate, dihydroartemisinin, artelinate, artemether, arteether have been developed to improve the bioavailability of Artemisinin.

Artemisinin and its derivatives – dihydroartemisinin, artemether, arteether, and artesunate being a class of antimalarials compounds used for the treatment of uncomplicated, severe complicated/cerebral and multi drug resistant malaria. Additionally, there are research findings that artemisinin and its derivatives show anti-parasite, anti-cancer, and anti-viral activities.

Dihydroartemisinin Artesunate

The content of Artemisinin in the plant Artemisia annua varies significantly according to the climate and region/geographical area where it is cultivated. Further, the extraction methods provide artemisinin or artemisinic acid from the plant in very poor yields and therefore not sufficient to accommodate the ever-growing need for this important drug. Consequently, widespread use of these valuable drugs has been hampered due to the low availability of this natural product. Therefore, research has focused on the syntheses of this valuable drug in a larger scale to meet the increasing global demand and accordingly ample literature is available on the synthesis of artemisinin or its derivatives, but no commercial success being reported / known till date.

Artemisinin can be prepared synthetically from its precursors such as artemisinic acid or dihydroartemisinic acid according to literature methods known to skilled artisans. For example, dihydroartemisinic acid can be converted to artemisinin by a combination of photooxidation and air-oxidation processes as described in U.S. Patent No. 4,992,561.

Amorphadiene is an early starting material for synthesis of Artemisinic acid or dihydroartemisinic acid, which is an important intermediate for producing Artemisinin commercially, and WO2006128126 reported a preparation method as mentioned in scheme- 1.


acid

In accordance with the scheme 1, the amorphadiene is treated with di(cyclohexyl)borane ( δΗι ΒΗ followed by reaction with H2O2 in presence of NaOH to obtain the amorph-4-ene 12-ol which is further oxidized to dihydroartemisinic acid using CrCb/ifcSC^. The formation of amorph-4-ene 12-ol is taking place via epoxidation of the exocyclic double bond. However, the reported yields of this synthesis are very low, making it unviable to produce artemisinic acid at a cheaper cost than natural extraction, for commercial use.

Amorpha -4, 11-diene

A similar method is published in, WO2009088404, for synthesis of dihydroartemisinic acid through preparation of amorph-4-ene-12-ol via epoxide formation, albeit, predominantly at exo position by reacting the amorpha-4,11-diene with H2O2 in presence of porphyrin catalyst (TDCPPMnCl). During reaction, epoxidation also occurred at endo position leading to formation of Amorphadiene- 4,5- epoxide that remain as impurity. The formed exo epoxide (amorphadiene – 11, 12 – epoxide) is further reduced to get amorph- 4-ene 12-ol and then converted to dihydroartemisinic acid and finally converted into artemisinin.

Amorphadiene-11,12-epoxide

This process involves expensive & industry unfriendly reagents. Moreover, desired stereo isomers were obtained only in poor yields, because several purification steps were needed to get desired stereo isomers leading to escalated production/operational costs.

Therefore there remains a need in the art to improve the yield of Dihydroartemisinic acid, which could potentially reduce the cost of production of Artemisinin and/or its derivatives. Consequently it is the need of the hour to provide a synthetic and economically viable process to meet the growing worldwide demand by improving the process for Artemisinin and/or its derivatives to obtain them in substantially higher yields with good purity by plant friendly operations like crystallization/extractions rather than column chromatography/other cost constraint procedures.

Therefore, the object of the invention is to prepare Artemisinic acid of formula-II, Dihydroartemisinic acid of formula-IIa, Artemisinin and its derivatives through Amorphadiene- 4,5- epoxide.

DHAA methyl ester

Scheme 2

 

Method 4 (From compound of formula IV (R = CI)):

In the 4-neck round bottom flask was charged Diphenyl sulfoxide (23.8 g), NaHC03 (32.96 g) and DMSO (80 ml) at 30°C. Further a solution of compound of formula IV (R = CI) (10 g) in DMSO (20 ml) was charged to the reaction mass at 30°C followed by heating and maintaining the temperature for 40 hours at 80°C till completion. DMSO was distilled out under vacuum. The reaction mass was cooled followed by charging water

(100 ml) and toluene (100 ml) to the reaction mass with stirring for 30 minutes at 28°C. The layers were separated out and aqueous layer was back extracted with toluene (2 X 100 ml). The organic layer was washed with water (100 ml) and saturated brine solution (100 ml). Solvent was distilled out under vacuum at 50°C, and the crude mass degassed under vacuum at 50-55°C. IPA (40 ml) was charged to the mass. Simultaneous addition of hydrazine hydrate (65% in aqueous solution) (3.8 g) and hydrogen peroxide (50% in aqueous solution) (2.5 ml) was done at 30-32°C over a period of 3.25 hours. After completion, reaction mass was cooled up to 5-10°C and water (100ml) was added to the reaction mass. The pH of the reaction mass was adjusted to 3.8 with dilute 8% aqueous HC1 (24 ml) at 10°C. Ethyl acetate (60 ml) was added to the reaction mass at 10°C and stirred for 15 minutes at 15-20°C. The layers were separated. Aqueous layer was back extracted with ethyl acetate (2 X 20 ml). The combined organic layer was washed with 10%) sodium metabisulfite solution (50 ml), water (50 ml) and saturated brine solution (50 ml). The organic layer was distilled out under vacuum at 45°C and the obtained crude mass was degassed at 50-55°C. To this was added DME (40 ml), Biphenyl (0.9 g) and Li-metal (1.63 g) and the reaction mass was maintained for 10 hours at 80-85°C till reaction completion. The reaction mass was cooled up to 0-5°C followed by drop wise addition of water within one hour, and the reaction stirred for two hours at 20-25°C. Toluene (35 ml) was charged with stirring and layers were separated. The aqueous layer was washed with toluene (35 ml) and the combined toluene layer was washed with water (20 ml). The combined aqueous layer was again washed with toluene (20 ml). The aqueous layer was cooled to 10-15°C and pH adjusted to 3.5-4 with dilute 16% aqueous HC1. MDC (50 ml) was charged and stirred 30 minutes at 20-25°C followed by separation of layers. The aqueous layer extracted with MDC (25 ml) and the combined MDC layer was washed with water (50 ml), then with saturated NaCl solution (25 ml). The solvent was distilled out under vacuum at 40-45°C and the crude mass (Purity: 70-80%>) was degassed at 65-70°C. The crude product (10 g) was dissolved in ethyl acetate (200 ml). 10%> aqueous NaOH (100 ml) was charged to the reaction mass and stirred one hour at 20°C followed by layer separation. Again 10%> aqueous NaOH (100ml) was added to the organic layer, stirred for 30 minutes and layers were separated out. The pH of the combined NaOH solution wash was adjusted to 4.0 with dilute 16%> aqueous HC1 at 5-10°C under stirring. Ethyl acetate (850 ml) was charged to aqueous acidic mass, stirred 30 minutes and layers were separated out. The aqueous layer was back extracted with ethyl acetate (2 X 30 ml) and the combined organic layer was washed with water (100 ml) and saturated brine (50 ml). The organic layer was dried over sodium chloride, solvent was distilled out under vacuum and the purified mass was degassed under vacuum at 50-55°C to obtain Dihydroartemisinic acid (Purity: 90-95%).

b) Methyl ester of Dihydroartemisinic acid:

To a clear solution of Dihydroartemisinic acid (40 g) dissolved in MDC (120 ml) was added thionyl chloride (SOCh) (14.85 ml) at 10±2°C and reaction mass was heated to reflux temperature 40±2°C. After the completion of reaction, solvent was distilled out and excess SOCh was removed under reduced pressure. The resulting concentrated mass of acid chloride was dissolved in MDC (200 ml). In another RBF was taken triethylamine (30.6 ml) and methanol (120 ml). To this solution was added above acid chloride solution at 30±2°C and maintained till completion of reaction. To the reaction mass was added water (400 ml) and organic layer was separated. The aqueous layer was washed with MDC and mixed with main organic layer and the combined organic layer was back washed with water till neutral pH. Then organic layer was concentrated to give methyl ester of Dihydroartemisinic acid as a brown color oily mass.

Weight: 41.88 gm

Yield = 98%

c) Artemisinin:

Methyl ester of dihydroartemisinic acid (67.7 g) was dissolved in methanol (338 ml). To this solution was added Sodium molybdate (29.5 g), 50% hydrogen peroxide (147.3 g) was added at 30±2°C and reaction was maintained for 3-4 hours. After completion of reaction was added water (300 ml) and MDC (300 ml) to the reaction mass. The organic layer was separated and aqueous layer washed with MDC (100 ml). The combined organic layer was concentrated to 475 ml containing hydroperoxide intermediate and directly used for next stage reaction. In another RBF containing MDC (475 ml) was added benzene sulfonic acid (1.27 g) and Indion resin (6.7 g). This heterogeneous solution was saturated with oxygen by passing O2 gas for 10 min at 0±2°C. To this was added previous stage hydroperoxide solution at same temperature with continuous 02 gas purging within 30-40 minutes. The oxygen gas was passed at same temp for 4 hours and temperature raised to 15±2°C with continued passing of oxygen for 5 hours. The

mixture was stirred at 25-30°C for 8-10 hours followed by filtration of resin. The filtrate was washed with water (200 ml X 3) and the combined aqueous layer back washed with MDC (50 ml). The combined organic layer was concentrated to give crude Artemisinin. Weight: 54 gm

Yield= 70.7%

Purification of Artemisinin:

Crude Artemisinin (10 g) was dissolved in ethyl acetate (25 ml) at 45-50°C. The solution was cooled to 30-35°C followed by addition of n-Hexane (100 ml). The material was isolated, stirred for 2 hours, filtered and vacuum dried at 45°C.

Weight: 4 gm

Yield: 40%

THE VIEWS EXPRESSED ARE MY PERSONAL AND IN NO-WAY SUGGEST THE VIEWS OF THE PROFESSIONAL BODY OR THE COMPANY THAT I REPRESENT, amcrasto@gmail.com, +91 9323115463 India

////////New Patent, WO 2016110874, Artemisinin , IPCA Laboratories Ltd, malaria, Cancer,  Parasitic infection,  Plasmodium falciparum infection,  Viral infection, artemether artemisinin,  artemotil,  artenimol,  artesunate,

Share

Crystallization of Artemisinin from Chromatography Fractions of Artemisia annua Extract

 PROCESS  Comments Off on Crystallization of Artemisinin from Chromatography Fractions of Artemisia annua Extract
Mar 102016
 
Abstract Image

Crystallization is an inevitable step in the purification of artemisinin either from the plantArtemisia annua or from reaction mixtures of semisynthetically produced artemisinin.

Rational design of crystallization process requires knowledge about the solid–liquid equilibrium in a given solvent system and effect of impurities on it.

In the present work, a crystallization process was designed to purify artemisinin from fractions of a flash chromatography column effluent collected after injecting extracts of Artemisia annua leaves.

The fractions from chromatography containing artemisinin were combined together into one fraction, and the impurities present in this fraction were identified.

The solubility of artemisinin in the mobile phase used for chromatography, i.e.,n-hexane–ethyl acetate mixture of varying compositions, was measured at 25, 15, and 5 °C, respectively. The collective effect of impurities present in the combined fraction on the solid–liquid equilibrium of artemisinin was evaluated by measuring the solubility of artemisinin in the combined fraction at same temperatures. The results show that the impurities present in the combined fraction increase the solubility of artemisinin.

Finally, the crystallization of artemisinin from the combined fraction designed on the basis of artemisinin solubility data was carried out in two steps by adding an antisolvent and cooling crystallization.

The yield of artemisinin obtained in the process was 50%, and it was found that the impurities present in the combined fraction at a given concentration do not affect the crystallization of artemisinin.

Figure

Figure

Figure 2. Chemical structure of artemisinin (1) and impurities [artemisitene (2), dihydroartemisinic acid (3), artemisinic acid (4), arteannuin B (5), and coumarin (6)] found in the combined fraction.

Crystallization of Artemisinin from Chromatography Fractions ofArtemisia annua Extract

Department of Chemical Engineering, Biotechnology and Environmental Technology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
§ Institute of Process Engineering, Otto von Guericke University, 39106 Magdeburg, Germany
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00399
Publication Date (Web): February 09, 2016
Copyright © 2016 American Chemical Society
*E-mail: crm@kbm.sdu.dk; phone: 0045 65508669.
Chandrakant Malwade

https://www.researchgate.net/profile/Chandrakant_Malwade/info

////////

Share

Synthetic chemistry fuels interdisciplinary approaches to the production of artemisinin

 SYNTHESIS  Comments Off on Synthetic chemistry fuels interdisciplinary approaches to the production of artemisinin
Jan 242015
 

7 Semi-synthesis of artemisinin using continuous flow. The Seeberger group has recently developed a continuous flow approach to the production of …

In the developing world, multi-drug resistant malaria caused by the parasite Plasmodium falciparum is an epidemic that claims the lives of 1–3 million people per year. Artemisinin, a naturally occurring small molecule that has seen little resistance from malarial parasites, is a valuable weapon in the fight against this disease. Several easily accessible artemisinin derivatives, including artesunate and artemether, display potent antimalarial activity against drug-resistant malaria strains; however, the global supply of artemisinin from natural sources alone remains highly inconsistent and unreliable. As a result, several approaches to artemisinin production have been developed, spanning areas such as total synthesis, flow chemistry, synthetic biology, and semi-synthesis. This review highlights achievements in all areas, in addition to the interplay between synthetic biology and synthetic chemistry that has fueled the recent industrial-scale production of artemisinin.

Graphical abstract: Synthetic chemistry fuels interdisciplinary approaches to the production of artemisinin

Synthetic chemistry fuels interdisciplinary approaches to the production of artemisinin

*

Corresponding authors
aDepartment of Chemistry and Biochemistry, University of California, Los Angeles, USA
Nat. Prod. Rep., 2015, Advance Article

DOI: 10.1039/C4NP00113C

Neil garg

http://www.chem.ucla.edu/dept/Faculty/garg/Garg_Group/About_Neil.html

Michael A. Corsello

Share

Sanofi and PATH launch large-scale malaria drug production

 Uncategorized  Comments Off on Sanofi and PATH launch large-scale malaria drug production
Aug 202014
 
Sanofi image

Sanofi and global health charity PATH have come together to launch a large-scale production line of malaria jab semisynthetic artemisinin at Sanofi’s Garessio site in Italy.

Global demand for artemisinin, the key ingredient of artemisinin-based combination therapies (ACTs) for malaria, has increased since the World Health Organization identified ACTs as the most effective malaria treatment available.

Because the existing botanical supply of artemisinin – derived from the sweet wormwood plant – is inconsistent, having multiple sources of high-quality product will strengthen its supply chain, contribute to a more stable price, and ultimately ensure greater availability of treatment to people suffering from malaria, according to Sanofi.

read at

http://www.pharmafile.com/news/192711/sanofi-and-path-launch-large-scale-malaria-drug-production

Share
Follow

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

Join other followers: