AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

Phytochemical compounds or their synthetic counterparts? A detailed comparison of the quantitative environmental assessment for the synthesis and extraction of curcumin

 PROCESS, spectroscopy, SYNTHESIS  Comments Off on Phytochemical compounds or their synthetic counterparts? A detailed comparison of the quantitative environmental assessment for the synthesis and extraction of curcumin
Mar 212016
 

 

Green Chem., 2016, 18,1807-1818
DOI: 10.1039/C6GC00090H, Paper
Elisabetta Zerazion, Roberto Rosa, Erika Ferrari, Paolo Veronesi, Cristina Leonelli, Monica Saladini, Anna Maria Ferrari
LCA of the synthesis of curcumin and its direct conventional and microwave assisted extractions fromCurcuma longa L. were compared.

Phytochemical compounds or their synthetic counterparts? A detailed comparison of the quantitative environmental assessment for the synthesis and extraction of curcumin

Phytochemical compounds or their synthetic counterparts? A detailed comparison of the quantitative environmental assessment for the synthesis and extraction of curcumin

*Corresponding authors
aDipartimento di Scienze e Metodi dell’Ingegneria, Università degli Studi di Modena e Reggio Emilia, via Amendola 2, 42100 Reggio Emilia, Italy
bDipartimento di Ingegneria “Enzo Ferrari”, Università degli Studi di Modena e Reggio Emilia, via Pietro Vivarelli 10, 41125 Modena, Italy
E-mail: roberto.rosa@unimore.it
Fax: +390592056243
Tel: +390592056224
c
Dipartimento di Scienze Chimiche e Geologiche, Università degli Studi di Modena e Reggio Emilia, via Campi 103, 41125 Modena, Italy
Green Chem., 2016,18, 1807-1818

DOI: 10.1039/C6GC00090H

Natural compounds represent an extremely wide category to be exploited, in order to develop new pharmaceutical strategies. In this framework, the number of in vitro, in vivo and clinical trials investigating the therapeutic potential of curcumin is exponentially increasing, due to its antioxidant, anti-inflammatory and anticancer properties. The possibility to obtain this molecule by both chemical synthesis and extraction from natural sources makes the environmental assessments of these alternative production processes of paramount importance from a green chemistry perspective, with the aim, for both industries and academia, to pursue a more sustainable development. The present work reports detailed and quantitative environmental assessments of three different curcumin production strategies: synthesis, conventional Soxhlet-based extraction (CE) and microwave-assisted extraction (MAE). The chemical synthesis of curcumin, as recently optimized by the authors, has been firstly evaluated by using the EATOS software followed by a complete “cradle to the grave” study, realized by applying the Life Cycle Assessment (LCA) methodology. The life cycles of CE and MAE were then similarly assessed, considering also the cultivation of Curcuma longa L., the production of the dried rhizomes as well as their commercialization, in order to firstly investigate the widely claimed green character of MAE with respect to more conventional extraction procedures. Secondly, the results related to the two different extraction strategies were compared to those obtained by the chemical synthesis of curcumin, with the aim to determine its greenest preparation procedure among those investigated. This work represents the first example of an environmental assessment comparison between different production strategies of curcumin, thus smoothing the way towards the highly desirable establishment of environmentally friendly rankings, comprising all the existing alternatives to the chemical synthesis of a target chemical compound.

/////Phytochemical compounds,  synthesis,  extraction, curcumin

Share

A pot-economical and diastereoselective synthesis involving catalyst-free click reaction for fused-triazolobenzodiazepines

 PROCESS, SYNTHESIS  Comments Off on A pot-economical and diastereoselective synthesis involving catalyst-free click reaction for fused-triazolobenzodiazepines
Mar 212016
 

 

Green Chem., 2016, Advance Article
DOI: 10.1039/C6GC00497K, Communication
Xiaofeng Zhang, Sanjun Zhi, Wei Wang, Shuai Liu, Jerry P. Jasinski, Wei Zhang
A pot-economical synthesis involving two [3 + 2] cycloadditions for diastereoselective synthesis of novel triazolobenzodiazepine-containing polycyclic compounds

http://pubs.rsc.org/en/Content/ArticleLanding/2016/GC/C6GC00497K?utm_source=feedburner&utm_medium=feed&utm_campaign=Feed%3A+rss%2FGC+%28RSC+-+Green+Chem.+latest+articles%29#!divAbstract

A pot-economical and diastereoselective synthesis involving catalyst-free click reaction for fused-triazolobenzodiazepines

 *Corresponding authors
aCentre for Green Chemistry and Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, USA
E-mail: wei2.zhang@umb.edu
bJiangsu Key Laboratory for the Chemistry of Low-Dimensional Materials, Huaiyin Normal University, Huaian, PR China
cSchool of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an, PR China
dDepartment of Chemistry, Keene State College, Keene, USA
Green Chem., 2016, Advance Article

DOI: 10.1039/C6GC00497K

A pot-economical synthesis involving sequential [3 + 2] cycloadditions of an azomethine ylide and an azide–alkyne (click reaction) has been developed for diastereoselective synthesis of novel triazolobenzodiazepine-containing polycyclic compounds. A new example of catalyst-free click chemistry of non-strained alkynes is also disclosed

str1

 

str1

 

str1

 

str1

 

str1

 

str1

/////A pot-economical, diastereoselective synthesis, catalyst-free click reaction, fused-triazolobenzodiazepines

Share

Cycloaddition of epoxides and CO2 catalyzed by bisimidazole-functionalized porphyrin cobalt(III) complexes

 PROCESS  Comments Off on Cycloaddition of epoxides and CO2 catalyzed by bisimidazole-functionalized porphyrin cobalt(III) complexes
Mar 212016
 

 

 

 

Cycloaddition of epoxides and CO2 catalyzed by bisimidazole-functionalized porphyrin cobalt(III) complexes

Green Chem., 2016, Advance Article
DOI: 10.1039/C6GC00370B, Paper
Xu Jiang, Faliang Gou, Fengjuan Chen, Huanwang Jing
Bisimidazole-functionalized cobaltoporphyrin acted as efficient bifunctional catalysts to facilitate the synthesis of cyclic carbonates from epoxides and CO2.
see

Cycloaddition of epoxides and CO2 catalyzed by bisimidazole-functionalized porphyrin cobalt(III) complexes

Xu Jiang,a   Faliang Gou,a   Fengjuan Chena and  Huanwang Jing*ab  
 *Corresponding authors
aState Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering Lanzhou University, Gansu 730000, PR China
bState Key Laboratory of Coal Conversion, Institute of Coal Chemistry, Chinese Academy of Sciences, Taiyuan 030001, P R China
E-mail: hwjing@lzu.edu.cn
Green Chem., 2016, Advance Article

DOI: 10.1039/C6GC00370B

A series of innovative bisimidazole-functionalized porphyrin cobalt(III) complexes have been devised, synthesized and characterized using NMR, MS and elemental analysis. These homogeneous catalysts were applied to the cycloaddition of epoxides and carbon dioxide without organic solvent and co-catalyst. It was found that the performance of the catalysts deeply relies on their structural features. The alkoxyl chain length of the linkage and the imidazole position relative to the phenyl rings of porphyrin evidently affects the catalyst activities. [5,15-Di(3-((8-imidazolyloctyl)oxy)phenyl)porphyrin] cobalt(III) chloride (J-m8) and [5,15-di(2-((6-imidazolylhexyl)oxy)phenyl)porphyrin] cobalt(III) chloride (J-o6) demonstrated excellent activity under optimal reaction conditions. Synchronously, a preliminary kinetic investigation of this reaction was carried out using three catalysts and illustrated the activation energies of cyclic carbonate formation. Furthermore, a tri-synergistic catalytic mechanism has been carefully proposed in light of the features of the new catalysts and experimental results.
str1

 

str1

 

References [1] L. Jin, H. Jing, T. Chang, X. Bu, L. Wang and Z. Liu, J. Mol. Catal. A: Chem., 2007, 261, 262. [2] X. Jiang, F. Gou and H. Jing, J. Catal., 2014, 313, 159. [3] B. Li, L. Zhang, Y. Song, D. Bai and H. Jing, J. Mol. Catal. A: Chem., 2012, 363– 364, 26.

 

///Cycloaddition of epoxides,   CO2 catalyzed,  bisimidazole-functionalized porphyrin cobalt(III) complexes

Share

Trichloroacetic Acid Removal by a Reductive Spherical Cellulose Adsorbent

 PROCESS  Comments Off on Trichloroacetic Acid Removal by a Reductive Spherical Cellulose Adsorbent
Mar 212016
 

 

Chemical Research in Chinese Universities Vol.32 No.1 February 2016 2016 Vol. 32 (1): 0-0 [Abstract] ( 20 ) [HTML 1KB] [PDF 2198KB] ( 11 )

A novel spherical cellulose adsorbent with amide and sulphinate groups was used for a first reduction of trichloroacetic acid(TCAA) and a subsequent adsorption of generated species, haloacetic acids. The removal mechanism involved TCAA reduction by sulphinate groups and the adsorption of the haloacetic acids through electrostatic interaction with amide group. Investigation of product formation and subsequent disappearance reveals that the reduction reactions proceed viasequential hydrogenolysis, and transform to acetate ultimately. Adsorption of haloacetic acids was ascertained by low chloride mass balances(89.3%) and carbon mass balances(75.1%) in solution. The pseudo-first-order rate constant for TCAA degradation was (0.93±0.12) h-1. Batch experiments were conducted to investigate the effect of pH value on the reduction and adsorption process. The results show that the reduction of TCAA by sulphinate groups requires higher pH values while the electrostatic attraction of haloacetic acids by amino group is favorable in more acidic media.

Trichloroacetic Acid Removal by a Reductive Spherical Cellulose Adsorbent
LIN Chunxiang1,3, TIAN Chen1, LIU Yifan1,3, LUO Wei1, ZHU Moshuqi1, SU Qiaoquan1, LIU Minghua1,2,3
1. College of Environment & Resources, Fuzhou 350108, P. R. China;
2. State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China;
3. Key Laboratory of Eco-materials Advanced Technology(Fuzhou University), Fujian Province University, Fuzhou 350108, P. R. China

LIU Minghua     E-mail: mhliu2000@fzu.edu.cn

Cite this article:
LIN Chunxiang,TIAN Chen,LIU Yifan等. Trichloroacetic Acid Removal by a Reductive Spherical Cellulose Adsorbent[J]. CHEMICAL RESEARCH IN CHINESE UNIVERSITIES, 2016, 32(1): 95-99.
LIN Chunxiang, TIAN Chen, LIU Yifan, LUO Wei, ZHU Moshuqi, SU Qiaoquan, LIU Minghua
Trichloroacetic Acid Removal by a Reductive Spherical Cellulose Adsorbent
2016 Vol. 32 (1): 95-99 [Abstract] ( 9 ) [HTML 1KB] [PDF 0KB] ( 12 )
doi: 10.1007/s40242-016-5304-6

/////Trichloroacetic Acid Removal, Reductive Spherical Cellulose Adsorbent

Share

An Improved Process for the Preparation of Tenofovir Disoproxil Fumarate

 MANUFACTURING, PROCESS, SYNTHESIS  Comments Off on An Improved Process for the Preparation of Tenofovir Disoproxil Fumarate
Mar 152016
 

 

VIREAD® (tenofovir disoproxil fumarate) Structural Formula Illustration

Tenofovir Disoproxil Fumarate

For full details see end of page

 

PAPER

 

 

Abstract Image

The current three-step manufacturing route for the preparation of tenofovir disoproxil fumarate (1) was assessed and optimized leading to a higher yielding, simpler, and greener process. Key improvements in the process route include the refinement of the second stage through the replacement of the problematic magnesium tert-butoxide (MTB) with a 1:1 ratio of a Grignard reagent and tert-butanol. The development of a virtually solvent-free approach and the establishment of a workup and purification protocol which allows the isolation of a pure diethyl phosphonate ester (8) was achieved

 

str1

 

 

see………….http://pubs.acs.org/doi/abs/10.1021/acs.oprd.5b00364

An Improved Process for the Preparation of Tenofovir Disoproxil Fumarate

Department of Chemistry, Natural and Agricultural Sciences, University of Pretoria, 2 Lynnwood Road, Hatfield, 0002, Gauteng, South Africa
Department of Engineering and Technology Management, University of Pretoria, Pretoria, South Africa
§ Pharmaceutical Manufacturing Technology Centre, University of Limerick, Limerick, V94 T9PX, Republic of Ireland
iThemba Pharmaceuticals, Modderfontein, 1645, Gauteng South Africa
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00364
Publication Date (Web): March 04, 2016
Copyright © 2016 American Chemical Society

University of Pretoria

Department of Chemistry, Natural and Agricultural Sciences, University of Pretoria, 2 Lynnwood Road, Hatfield, 0002, Gauteng, South Africa

Map of Department of Chemistry, Natural and Agricultural Sciences, University of Pretoria, 2 Lynnwood Road, Hatfield, 0002, Gauteng, South Africa

///////

Tenofovir Disoproxil Fumarate

5-[[(1R)-2-(6-Amino-9H-purin-9-yl)-1-methylethoxy]methyl]-2,4,6,8-tetraoxa-5-phosphanonanedioic Acid 1,9-Bis(1-methylethyl) Ester 5-Oxide (2E)-2-Butenedioate; GS 4331-05; PMPA Prodrug; Tenofovir DF; Virea; Viread;

GILEAD-4331-300

201341-05-1 – free base, (Tenofovir Disoproxil

 fumarate 202138-50-9
113-115°C (dec.)
CAS No.: 202138-50-9
Name: Tenofovir disoproxil fumarate
Molecular Structure:
Molecular Structure of 202138-50-9 (Tenofovir disoproxil fumarate)
Formula: C19H30N5O10P.C4H4O4
Molecular Weight: 635.51
Synonyms: TDF;PMPA prodrug;Tenofovir Disoproxil Fumarate [USAN];9-((R)-2-((Bis(((isopropoxycarbonyl)oxy)methoxy)phosphinyl)methoxy)propyl)adenine, fumarate;201341-05-1;Bis(NeopentylOC)PMPA;Viread;GS 4331-05 (*1:1 Fumarate salt*);Viread (*1:1 Fumarate salt*);Truvada;Tenofovir DF;[[(2R)-1-(6-aminopurin-9-yl)propan-2-yl]oxymethyl-(propan-2-yloxycarbonyloxymethoxy)phosphoryl]oxymethyl propan-2-yl carbonate;
Usage
tyrosinase inhibitor used for skin lightening and anti-melasma
Usage
An acyclic phosphonate nucleotide analog and selective HIV-1 RT inhibitor
Usage
Acyclic phosphonate nucleotide analogue; reverse transcriptase inhibitor. Used as an anti-HIV agent. Antiviral.

 

Tenofovir disoproxil is an antiretroviral medication used to prevent and treat HIV/AIDS and to treat chronic hepatitis B.[1] The active substance is tenofovir, while tenofovir disoproxil is a prodrug that is used because of its better absorption in the gut.

The drug is on the World Health Organization’s List of Essential Medicines, the most important medications needed in a basic health system.[2] It is marketed by Gilead Sciences under the trade name Viread (as the fumarate, TDF).[3] As of 2015 the cost for a typical month of medication in the United States is more than 200 USD.[4]

http://www.intmedpress.com/journals/avt/iframePopup_fig.cfm?img=c32b4107-6d95-47c7-bb57-45390ba123b1

Medical uses

  • HIV-1 infection: Tenofovir is indicated in combination with other antiretroviral agents for the treatment of HIV-1 infection in adults and pediatric patients 2 years of age and older.[5] This indication is based on analyses of plasma HIV-1 RNA levels and CD4 cell counts in controlled studies of tenofovir in treatment-naive and treatment-experienced adults.
  • Tenofovir is indicated for the treatment of chronic hepatitis B in adults and pediatric patients 12 years of age and older.[5][6]

HIV risk reduction

A Cochrane review examined the use of tenofovir for prevention of HIV before exposure. It found that both tenofovir alone and the tenofovir/emtricitabine combination decreased the risk of contracting HIV.[7]

The U. S. Centers for Disease Control and Prevention (CDC) conducted a study in partnership with the Thailand Ministry of Public Health to ascertain the effectiveness of providing people who inject drugs illicitly with daily doses of the antiretroviral drug tenofovir as a prevention measure. The results of the study were released in mid-June 2013 and revealed a 48.9%-reduced incidence of the virus among the group of subjects who received the drug, in comparison to the control group who received a placebo. The principal investigator of the study stated: “We now know that pre-exposure prophylaxis can be a potentially vital option for HIV prevention in people at very high risk for infection, whether through sexual transmission or injecting drug use.”[8]

Adverse effects

The most common side effects associated with tenofovir include nausea, vomiting, diarrhea, and asthenia. Less frequent side effects include hepatotoxicity, abdominal pain, and flatulence.[9] Tenofovir has also been implicated in causing renal toxicity, particularly at elevated concentrations.[10]

Tenofovir can cause acute renal failure, Fanconi syndrome, proteinuria, or tubular necrosis.[citation needed] These side effects are due to accumulation of the drug in proximal tubules.[citation needed] Tenofovir can interact with didanosine by increasing didanosine’s concentration.[citation needed] It also decreases the concentration of atazanavir sulfate.[citation needed]

Mechanism of action

Tenofovir is a defective adenosine nucleotide that selectively interferes with the action of reverse transcriptase, but only weakly interferes with mammalian DNA polymerases α, β, and mitochondrial DNA polymerase γ.[11] Tenofovir prevents the formation of the 5′ to 3′ phosphodiester linkage essential for DNA chain elongation. A phosphodiester bond cannot be formed because the tenofovir molecule lacks an —OH group on the 3′ carbon of its deoxyribose sugar.[11] Once incorporated into a growing DNA strand, tenofovir causes premature termination of DNA transcription. The drug is classified as a nucleotide analogue reverse transcriptase inhibitor (NRTI), that inhibits reverse transcriptase.[11] Reverse transcriptase is a crucial viral enzyme in retroviruses such as human immunodeficiency virus (HIV) and in hepatitis B virus infections.[5]

History

Tenofovir was initially synthesized by Antonín Holý at the Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic in Prague. The patent[12] filed by Holý in 1984 makes no mention of the potential use of the compound for the treatment of HIV infection, which had only been discovered one year earlier.

In 1985, De Clercq and Holý described the activity of PMPA against HIV in cell culture.[13] Shortly thereafter, a collaboration with the biotechnology company Gilead Sciences led to the investigation of PMPA’s potential as a treatment for HIV infected patients. In 1997 researchers from Gilead and the University of California, San Francisco demonstrated that tenofovir exhibits anti-HIV effects in humans when dosed by subcutaneous injection.[14]

The initial form of tenofovir used in these studies had limited potential for widespread use because it was not absorbed when administered orally. A medicinal chemistry team at Gilead developed a modified version of tenofovir, tenofovir disoproxil.[15] This version of tenofovir is often referred to simply as “tenofovir”. In this version of the drug, the two negative charges of the tenofovir phosphonic acid group are masked, thus enhancing oral absorption.

Tenofovir disoproxil was approved by the U.S. FDA on October 26, 2001, for the treatment of HIV, and on August 11, 2008, for the treatment of chronic hepatitis B.[16][17]

Drug forms

Tenofovir disoproxil is a prodrug form of tenofovir. It is also marketed under the brand name Reviro by Dr. Reddy’s Laboratories. Tenofovir is also available in a fixed-dose combination with emtricitabine in a product with the brand name Truvada for once-a-day dosing. Efavirenz/emtricitabine/tenofovir disoproxil (brand name Atripla) — a fixed-dose triple combination of tenofovir, emtricitabine, and efavirenz, was approved by the FDA on 12 July 2006 and is now available, providing a single daily dose for the treatment of HIV.

Therapeutic drug monitoring

Tenofovir may be measured in plasma by liquid chromatography. Such testing is useful for monitoring therapy and to prevent drug accumulation and toxicity in people with kidney or liver problems.[18][19][20]

PATENT

http://www.google.com/patents/EP2545063A2?cl=en

Tenofovir Disoproxil is chemically known as 9-[-2-(R)-[[bis [[(isopropoxycarbonyl) oxy]methoxy] phosphinoyl]methoxy]propyl]-adenine, having the following structural formula-I.

Formula-I

Tenofovir is a highly potent antiviral agent, particularly for the therapy or prophylaxis of retroviral infections and belongs to a class of drugs called Nucleotide Reverse Transcriptase Inhibitors (NRTI) which blocks reverse transcriptase an enzyme crucial to viral production in HIV-infected people.

Tenofovir Disoproxil and its pharmaceutically acceptable salts were first disclosed in US 5,922,695. This patent discloses the preparation of Tenofovir Disoproxil by the esterification of Tenofovir with chloromethyl isopropyl carbonate using l-methyl-2- pyrrolidinone and triethylamine. In this patent Tenofovir Disoproxil is converted into its Fumarate salt without isolation. PCT Publication WO 2008007392 discloses process for the preparation of Tenofovir Disoproxil fumarate, wherein the isolated crystalline Tenofovir Disoproxil is converted into fumarate salt.

Tenofovir Disoproxil processes in the prior art are similar to process disclosed in product patent US 5,922,695. According to the prior art processes, Tenofovir Disoproxil fumarate obtained is having low yields and also show the presence of impurities such as dimers.

scheme- 1.

Tenofovir disoproxil chloromethyl isopropyl carbonate

Tenofovir disoproxil fumarate

Example 1 : Process for the preparation of Tenofovir Disoproxil fumarate

Toluene (500 ml) was added to the Tenofovir (100 gm) and stirred at room temperature. To this triethylamine (66.31 gm) was added, temperature was raised to 90° C and water was collected by azeotropic distillation at 110°C. Toluene was completely distilled under vacuum at same temperature. The reaction mixture was cooled to room temperature and to this a mixture of N-methyl pyrrolidine (300 gm), triethylamine (66.31 gm), Tetrabutyl ammonium bromide (52.8 gm) and trimethyl silyl chloride (17.8 gm) were added. The above reaction mixture was heated to 50-55 °C and was added slowly chloromethyl. isopropyl carbonate (CMIC) and maintained the reaction mixture at 50-55°C for 5 hrs. (Qualitative HPLC analysis shows about 85% product formation). The above reaction mixture was cooled to room temperature and filtered. The filtrate was added to DM water at 5-10°C and extract with dichloromethane. The combined dichloromethane layer was concentrated under vacuum and the crude was Co-distilled with cyclohexane and this crude was taken into isopropyl alcohol (1000 ml). To this fumaric acid (38 gm) was added and temperature was raised to 50° C. The reaction mixture was filtered and filtrate was cooled to 5-10° C. The obtained solid was filtered and washed with isopropyl alcohol. The compound was dried under vacuum to yield Tenofovir Disoproxil fumarate (140 gm).

Example-2 : Preparation of Tenofovir

N-methyl-2-pyrrolidone (25 gm) was taken along with toluene (150 gm) into a reaction vessel. l-(6-amino-purin-9-yl)-propan-2-ol (100 gm); toluene-4-sulfonic acid diethoxy phosphoryl methyl ester (200 gm) and magnesium ter-butoxide (71.2 gm) were also taken at’ 25-35°C. Temperature was raised to 74-75 °C and maintained for 5-6hrs. After completion of reaction, acetic acid (60 gm) was added and maintained for 1 hr. Later aq.HBr (332 gm) was taken and heated to 90-95 °C. After reaction completion, salts were filtered and filtrate was subjected to washings with water and extracted into methylene dichloride. Later pH was adjusted using CS lye below 10 °C. Tenofovir product was isolated using acetone.

Yield: 110 gm.

Example 3 : Preparation of Tenofovir disoproxil

(R)-9-[2-(phosphonomethoxy)propyl]adenine (25 gm), triethyl amine (25 ml) and cyclohexane (200 ml) were combined and heated to remove water and the solvent was distilled off under vacuum. The reaction mass was cooled to room temperature N-methyl pyrrolidinone (55 ml), triethyl amine (25 ml) and tetra butyl ammonium bromide(54 gms) were added to the reaction mixture. The reaction mass was heated to 50-60°C and chloromethyl isopropyl carbonate (65 gm) was added and maintained for 4-8 hrs at 50- 60°C and then cooled to 0°C. The reaction mass was diluted with chilled water or ice and precipitated solid product was filtered. The mother liquor was extracted with methylene chloride (150 ml). The methylene chloride layer was washed with water (200 ml). The filtered solid and the methylene chloride layer were combined and washed with water and the solvent was distilled under vacuum. Ethyl acetate was charged to the precipitated solid. The reaction mass was then cooled to 0-5 °C and maintained for 6 hrs. The solid was filtered and dried to produce Tenofovir disoproxil (45 gm).

CLIPS

The reaction of chloromethyl chloroformate (I) with isopropyl alcohol (II) by means of pyridine or triethylamine in ether gives the mixed carbonate (III), which is then condensed with (R)-PMPA (IV) by means of diisopropyl ethyl-amine in DMF.

US 5922695; WO 9804569

CLIP 2

1) The protection of isobutyl D-(+)-lactate (I) with dihydropyran (DHP)/HCl in DMF gives the tetrahydropyranyloxy derivative (II), which is reduced with bis(2-methoxyethoxy)aluminum hydride in refluxing ether/ toluene yielding 2(R)-(tetrahydropyranyloxy)-1-propanol (III). The tosylation of (III) with tosyl chloride as usual affords the expected tosylate (VI), which is condensed with adenine (V) by means of Cs2CO3 in hot DMF, affording 9-[2(R)-(tetrahydropyranyloxy)propyl]adenine (VI). The deprotection of (VI) with sulfuric acid affords 9-[2(R)-hydroxypropyl]adenine (VII), which is N-benzoylated with benzoyl chloride/chlorotrimethylsilane in pyridine to give the benzamide (VIII), which is condensed with tosyl-oxymethylphosphonic acid diisopropyl ester (IX) by means of NaH in DMF to yield 9-[2(R)-(diisopropoxyphosphorylmethoxy)propyl]adenine (X). Finally, this compound is hydrolyzed by means of bromotrimethylsilane in acetonotrile.

 

 

2) The reaction of the previously described (R)-2-(2-tetrahydropyranyloxy)-1-propanol (III) with benzyl bromide (XI) by means of NaH in DMF, followed by a treatment with Dowex 50X, gives 1-benzyloxy-2(R)-propanol (XII), which is condensed with tosyloxymethylphosphonic acid diisopropyl ester (IX) by means of NaH in THF, yielding 2-benzyloxy-1(R)-methylethoxymethylphosphonic acid diisopropyl ester (XIII). The hydrogenolysis of (XIII) over Pd/C in methanol affords 2-hydroxy-1(R)-methylethoxymethylphosphonic acid diisopropyl ester (XIV), which is tosylated with tosyl chloride/dimethyl-aminopyridine in pyridine to give the expected tosylate (XV). The condensation of (XV) with adenine (VI) by means of Cs2CO3 in hot DMF yields 9-[2(R)-(diisopropoxyphosphorylmethoxy)propyl]adenine (X), which is finally hydrolyzed as before.

 

3) The catalytic hydrogenation of (S)-glycidol (XVI) over Pd/C gives the (R)-1,2-propanediol (XVII), which is esterified with diethyl carbonate (XVIII)/NaOEt, yielding the cyclic carbonate (XIX). The reaction of (XIX) with adenine (V) by means of NaOH in DMF affords 9-[2(R)-hydroxypropyl]adenine (VII), which is condensed with tosyloxymethylphosphonic acid diethyl ester (XX) by means of lithium tert-butoxide in THF, giving 9-[2(R)-(diethoxyphosphorylmethoxy)propyl]adenine (XXI). Finally, this compound is hydrolyzed with bromotrimethylsilane as before. Compound (XX) is obtained by reaction of diethyl phosphite (XXII) with paraformaldehyde, yielding hydroxy- methylphosphonic acid diethyl ester (XXIII), which is finally tosylated as usual.

 

References

  1. R. Baselt, Disposition of Toxic Drugs and Chemicals in Man, 8th edition, Biomedical Publications, Foster City, California, 2008, pp. 1490–1492.

External links

WO2008007392A2 Jul 11, 2007 Jan 17, 2008 Matrix Lab Ltd Process for the preparation of tenofovir
US5922695 Jul 25, 1997 Jul 13, 1999 Gilead Sciences, Inc. Antiviral phosphonomethyoxy nucleotide analogs having increased oral bioavarilability
WO2015051874A1 Sep 22, 2014 Apr 16, 2015 Zentiva, K.S. An improved process for the preparation of tenofovir disoproxil and pharmaceutically acceptable salts thereof
CN103360425A * Apr 1, 2012 Oct 23, 2013 安徽贝克联合制药有限公司 Synthesis method of tenofovir disoproxil and fumarate thereof
CN103374038A * Apr 11, 2012 Oct 30, 2013 广州白云山制药股份有限公司广州白云山制药总厂 Preparation method of antiviral medicine
CN103848868A * Dec 4, 2012 Jun 11, 2014 蚌埠丰原涂山制药有限公司 Method for preparing tenofovir
CN103848869A * Dec 4, 2012 Jun 11, 2014 上海医药工业研究院 Method for preparing tenofovir
CN103980319A * Apr 24, 2014 Aug 13, 2014 浙江外国语学院 Preparation method of tenofovir
CN103980319B * Apr 24, 2014 Dec 2, 2015 浙江外国语学院 一种泰诺福韦的制备方法
EP2860185A1 Oct 9, 2013 Apr 15, 2015 Zentiva, k.s. An improved process for the preparation of Tenofovir disoproxil and pharmaceutically acceptable salts thereof

 

 

The chemical name of tenofovir disoproxil fumarate is 9-[(R)-2[[bis[[(isopropoxycarbonyl)oxy]methoxy]phosphinyl]methoxy]propyl]adenine fumarate (1:1). It has a molecular formula of C19H30N5O10P • C4H4O4 and a molecular weight of 635.52. It has the following structural formula:

 

VIREAD® (tenofovir disoproxil fumarate) Structural Formula Illustration

Tenofovir disoproxil fumarate is a white to off-white crystalline powder with a solubility of 13.4 mg/mL in distilled water at 25 °C. It has an octanol/phosphate buffer (pH 6.5) partition coefficient (log p) of 1.25 at 25 °C.

VIREAD is available as tablets or as an oral powder.

VIREAD tablets are for oral administration in strengths of 150, 200, 250, and 300 mg of tenofovir disoproxil fumarate, which are equivalent to 123, 163, 204 and 245 mg of tenofovir disoproxil, respectively. Each tablet contains the following inactive ingredients: croscarmellose sodium, lactose monohydrate, magnesium stearate, microcrystalline cellulose, and pregelatinized starch. The 300 mg tablets are coated with Opadry II Y-3010671-A, which contains FD&C blue #2 aluminum lake, hypromellose 2910, lactose monohydrate, titanium dioxide, and triacetin. The 150, 200, and 250 mg tablets are coated with Opadry II 32K-18425, which contains hypromellose 2910, lactose monohydrate, titanium dioxide, and triacetin.

VIREAD oral powder is available for oral administration as white, taste-masked, coated granules containing 40 mg of tenofovir disoproxil fumarate per gram of oral powder, which is equivalent to 33 mg of tenofovir disoproxil. The oral powder contains the following inactive ingredients: mannitol, hydroxypropyl cellulose, ethylcellulose, and silicon dioxide.

enofovir disoproxil
Tenofovir disoproxil structure.svg
Systematic (IUPAC) name
Bis{[(isopropoxycarbonyl)oxy]methyl} ({[(2R)-1-(6-amino-9H-purin-9-yl)-2-propanyl]oxy}methyl)phosphonate
Clinical data
Trade names Viread
AHFS/Drugs.com monograph
Pregnancy
category
  • AU: B3
  • US: B (No risk in non-human studies)
Routes of
administration
Oral (tablets)
Legal status
Legal status
Pharmacokinetic data
Bioavailability 25%
Identifiers
CAS Number 201341-05-1
ATC code J05AF07 (WHO)
PubChem CID 5481350
ChemSpider 4587262
UNII F4YU4LON7I
ChEBI CHEBI:63717
NIAID ChemDB 080741
Chemical data
Formula C19H30N5O10P
Molar mass 519.443 g/mol
Tenofovir
Tenofovir structure.svg
Systematic (IUPAC) name
({[(2R)-1-(6-amino-9H-purin-9-yl)propan-2-yl]oxy}methyl)phosphonic acid
Clinical data
MedlinePlus a602018
Routes of
administration
In form of prodrugs
Pharmacokinetic data
Protein binding < 1%
Biological half-life 17 hours
Excretion Renal
Identifiers
CAS Number 147127-20-6 Yes
ATC code None
PubChem CID 464205
DrugBank DB00300 Yes
ChemSpider 408154 Yes
UNII 99YXE507IL Yes
KEGG D06074 Yes
ChEBI CHEBI:63625
ChEMBL CHEMBL483 Yes
Synonyms 9-(2-Phosphonyl-methoxypropyly)adenine (PMPA)
Chemical data
Formula C9H14N5O4P
Molar mass 287.213 g/mol

///////

Share

Nanopalladium-catalyzed conjugate reduction of Michael acceptors – application in flow

 PROCESS, SYNTHESIS  Comments Off on Nanopalladium-catalyzed conjugate reduction of Michael acceptors – application in flow
Mar 102016
 

 

Green Chem., 2016, Advance Article
DOI: 10.1039/C5GC02920A, Communication
Anuja Nagendiran, Henrik Sorensen, Magnus J. Johansson, Cheuk-Wai Tai, Jan-E. Backvall
A continuous-flow approach towards the selective nanopalladium-catalyzed hydrogenation of the olefinic bond in various Michael acceptors, which could lead to a greener and more sustainable process, has been developed.
A continuous-flow approach towards the selective nanopalladium-catalyzed hydrogenation of the olefinic bond in various Michael acceptors, which could lead to a greener and more sustainable process, has been developed. The nanopalladium is supported on aminofunctionalized mesocellular foam. Both aromatic and aliphatic substrates, covering a variation of functional groups such as acids, aldehydes, esters, ketones, and nitriles were selectively hydrogenated in high to excellent yields using two different flow-devices (H-Cube® and Vapourtec). The catalyst was able to hydrogenate cinnamaldehyde continuously for 24 h (in total hydrogenating 19 g cinnanmaldehyde using 70 mg of catalyst in the H-cube®) without showing any significant decrease in activity or selectivity. Furthermore, the metal leaching of the catalyst was found to be very low (ppb amounts) in the two flow devices
str1
3 Gottlieb, H. E.; Kotlyar, V; Nudelman, A. J. Org. Chem. 1997, 62, 7512-7515.
str1

Nanopalladium-catalyzed conjugate reduction of Michael acceptors – application in flow

*Corresponding authors
aDepartment of Organic Chemistry, Arrhenius Laboratory, Stockholm University, SE-106 91 Stockholm, Sweden
E-mail: jeb@organ.su.se
bBerzelii Centre EXSELENT on Porous Materials, Arrhenius Laboratory, Stockholm University, SE-106 91 Stockholm, Sweden
cAstraZeneca R&D, Innovative Medicines, Cardiovascular and Metabolic Disorders, Medicinal Chemistry, Pepparedsleden 1, SE-431 83 Mölndal, Sweden
dDepartment of Materials and Environmental Chemistry, Arrhenius Laboratory, Stockholm University, SE-106 91, Stockholm, Sweden
Green Chem., 2016, Advance Article

DOI: 10.1039/C5GC02920A

///////

Share

Efficient formation of nitriles in the vapor-phase catalytic dehydration of aldoximes

 PROCESS, SYNTHESIS  Comments Off on Efficient formation of nitriles in the vapor-phase catalytic dehydration of aldoximes
Mar 102016
 

 

Efficient formation of nitriles in the vapor-phase catalytic dehydration of aldoximes

Green Chem., 2016, Advance Article
DOI: 10.1039/C6GC00384B, Paper
Daolai Sun, Eisyun Kitamura, Yasuhiro Yamada, Satoshi Sato
Nitriles were efficiently produced in a vapor-phase dehydration of aldoximes over SiO2 catalysts without external heat supply.
A vapor-phase dehydration of acetaldoxime to acetonitrile was investigated over various solid catalysts. Among the tested catalysts, ZrO2, Al2O3 and SiO2 showed high catalytic activity for the formation of acetonitrile from acetaldoxime, while the correlation between catalytic activity and the acid property of the catalysts was not observed. Weak acidic sites such as silanols sufficiently work as catalytic sites for the dehydration, which does not require strong acids such as zeolites. Several SiO2 catalysts with different physical properties were tested, and the SiO2with the smallest pore size and the highest specific surface area showed the highest catalytic activity for the formation of acetonitrile. Because the dehydration of acetaldoxime to acetonitrile is exothermic, a large amount of reaction heat was generated during the reaction, and the reaction temperature was found to be significantly affected by the feed rate of the reactant and the flow rate of the carrier gas. In order to effectively utilize the in situ generated reaction heat, the dehydration of acetaldoxime to acetonitrile without using the external heat supply was conducted. The temperature was controllable even in the absence of the external heat, and the acetonitrile yield higher than 90% could be achieved in such a green operation under the environment-friendly adiabatic conditions.

Efficient formation of nitriles in the vapor-phase catalytic dehydration of aldoximes

*Corresponding authors
aGraduate School of Engineering, Chiba University, Chiba, Japan
E-mail: satoshi@faculty.chiba-u.jp
Fax: +81 43 290 3401
Tel: +81 43 290 3377
Green Chem., 2016, Advance Article

DOI: 10.1039/C6GC00384B

//////

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

Application of On-Line NIR for Process Control during the Manufacture of Sitagliptin

 PROCESS, spectroscopy, SYNTHESIS  Comments Off on Application of On-Line NIR for Process Control during the Manufacture of Sitagliptin
Mar 062016
 
Abstract Image

The transamination-chemistry-based process for sitagliptin is a through-process, which challenges the crystallization of the active pharmaceutical ingredient (API) in a batch stream composed of multiple components. Risk-assessment-based design of experiment (DoE) studies of particle size distribution (PSD) and crystallization showed that the final API PSD strongly depends on the seeding-point temperature, which in turn relies on the solution composition.

To determine the solution composition, near-infrared (NIR) methods had been developed with partial least squares (PLS) regression on spectra of simulated process samples whose compositions were made by spiking each pure component, either sitagliptin free base (FB), water, isopropyl alcohol (IPA), dimethyl sulfoxide (DMSO), or isopropyl acetate (IPAc), into the process stream according to a DoE. An additional update to the PLS models was made by incorporating the matrix difference between simulated samples in lab and factory batches.

Overall, at temperatures of 20–35 °C, the NIR models provided a standard error of prediction (SEP) of less than 0.23 wt % for FB in 10.56–32.91 wt %, 0.22 wt % for DMSO in 3.77–19.18 wt %, 0.32 wt % for IPAc in 0.00–5.70 wt %, and 0.23 wt % for water in 11.20–28.58 wt %. After passing the performance qualification, these on-line NIR methods were successfully established and applied for the on-line analysis of production batches for compositions prior to the seeding point of sitagliptin crystallization.

see……..http://pubs.acs.org/doi/abs/10.1021/acs.oprd.5b00409

Application of On-Line NIR for Process Control during the Manufacture of Sitagliptin

Global Science, Technology and Commercialization, Merck Sharp & Dohme Corporation P.O. Box 2000, Rahway, New Jersey 07065, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.5b00409
Publication Date (Web): February 12, 2016
Copyright © 2016 American Chemical Society

////

Share

Current Practices of Process Validation for Drug Substances and Intermediates

 PROCESS, regulatory  Comments Off on Current Practices of Process Validation for Drug Substances and Intermediates
Jan 132016
 
Abstract Image

Process validation includes laboratory optimization, pilot-plant introduction, and process implementation on manufacturing scale, as well as monitoring batches after implementation and continuously improving the manufacturing processes. There are many opportunities to change and optimize operations. The background information in this contribution describes current guidance and terminology for validation, including the integration of validation over the development lifecycle of drug substances. Various examples illustrate challenges and success stories of implementation as part of the overall approach to process validation.

 

Current Practices of Process Validation for Drug Substances and Intermediates

Anderson’s Process Solutions, 7400 Griffin Lane, Jacksonville, Oregon 97530, United States, Creative Innovation Partners, 1971 Western Avenue, Albany, New York 12203, United States, and Rib-X Pharmaceuticals Inc., 300 George Street, New Haven, Connecticut 06511, United States
Org. Process Res. Dev., 2011, 15 (1), pp 162–172
DOI: 10.1021/op1002825
Publication Date (Web): December 21, 2010
Copyright © 2010 American Chemical Society
* To whom correspondence should be addressed: E-mail: nganderson@dishmail.net., †

Creative Innovation Partners., ‡Rib-X Pharmaceuticals Inc.

http://pubs.acs.org/doi/abs/10.1021/op1002825

////////

Share
Follow

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

Join other followers: