Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nature ; 598(7881): 451-456, 2021 10.
Article in English | MEDLINE | ID: mdl-34464959

ABSTRACT

Metal-catalysed cross-couplings are a mainstay of organic synthesis and are widely used for the formation of C-C bonds, particularly in the production of unsaturated scaffolds1. However, alkyl cross-couplings using native sp3-hybridized functional groups such as alcohols remain relatively underdeveloped2. In particular, a robust and general method for the direct deoxygenative coupling of alcohols would have major implications for the field of organic synthesis. A general method for the direct deoxygenative cross-coupling of free alcohols must overcome several challenges, most notably the in situ cleavage of strong C-O bonds3, but would allow access to the vast collection of commercially available, structurally diverse alcohols as coupling partners4. We report herein a metallaphotoredox-based cross-coupling platform in which free alcohols are activated in situ by N-heterocyclic carbene salts for carbon-carbon bond formation with aryl halide coupling partners. This method is mild, robust, selective and most importantly, capable of accommodating a wide range of primary, secondary and tertiary alcohols as well as pharmaceutically relevant aryl and heteroaryl bromides and chlorides. The power of the transformation has been demonstrated in a number of complex settings, including the late-stage functionalization of Taxol and a modular synthesis of Januvia, an antidiabetic medication. This technology represents a general strategy for the merger of in situ alcohol activation with transition metal catalysis.


Subject(s)
Alcohols/chemistry , Bromides/chemistry , Carbon/chemistry , Chlorides/chemistry , Metals/chemistry , Oxygen/chemistry , Photochemistry , Catalysis , Methane/analogs & derivatives , Methane/chemistry , Nitrogen/chemistry , Oxidation-Reduction , Paclitaxel/chemistry , Simvastatin/chemical synthesis , Simvastatin/chemistry
2.
Drug Des Devel Ther ; 15: 2193-2210, 2021.
Article in English | MEDLINE | ID: mdl-34079222

ABSTRACT

PURPOSE: A differential release fixed dose matrix tablet of amlodipine besylate (AML-B) and simvastatin (SIM) was formulated to enhance patient compliance. MATERIAL AND METHOD: In the first phase, release controlling parameters of AML-B and SIM granules were identified and in the second phase a fixed dose AML-B and SIM tablet formulation was prepared and optimized for a differential release of the drugs using a quality by design (QbD) and risk assessment approach. A validated HPLC method was employed for simultaneous determination of AML-B and SIM for FDC formulation. A pharmacokinetics of the above drugs was studied in healthy dogs in the third phase. RESULTS: In QbD-based optimized formulation, Eudragit® RSPO-dicalcium phosphate (DCP) blend controlled the release of AML-B over 8 h, though this diffusion-controlled release assumed first order kinetics. DCP and Eudragit® RS 100 also retarded release of SIM causing SIM release over 8 h after AML-B release from the optimized FDC tablet formulation. The HPLC retention times of AML-B and SIM were 2.10 and 15.52 min, respectively. Linearity for AML-B was 5.0-50 ng/mL and 0.01-2.0 µg/mL for SIM with percent recoveries of 92.85-101.53% and 94.51-117.75% for AML-B and SIM. AUC0-∞ of AML-B was increased 3 fold, while AUC0-∞ of SIM was decreased 2 fold. The tmax values for AML-B and SIM were 12 and 6 h, respectively. AML-B was absorbed without any lag time (tlag) while tlag was 6.33 ± 0.81 h for SIM, thus met the study objective. CONCLUSION: The pharmacokinetic study showed an immediate absorption of AML-B while that of SIM was withheld for 6 h, close to the desired delay time of 8 h.


Subject(s)
Amlodipine/pharmacokinetics , Simvastatin/pharmacokinetics , Amlodipine/chemical synthesis , Amlodipine/chemistry , Dose-Response Relationship, Drug , Drug Compounding , Drug Design , Drug Liberation , Humans , Risk Assessment , Simvastatin/chemical synthesis , Simvastatin/chemistry , Tablets
3.
AAPS PharmSciTech ; 21(4): 119, 2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32318974

ABSTRACT

In the presented study, insight into the development and optimisation of the dry emulsion formulation and spray drying process is provided. The aim was to facilitate the dissolution of the poorly soluble, highly lipophilic drug, simvastatin, by forming spray-dried dry emulsion particles having adequate powder flow properties, while assuring sufficient drug content. Simvastatin and a mixture of caprylic, capric triglyceride and 1-oleoyl-rac-glycerol were employed as a model drug and solubilising oils, respectively. A matrix of the dry emulsions was composed at a fixed ratio mixture of mannitol and HPMC. Tween 20 was used in low amounts as the primary emulsion stabiliser. To facilitate process optimisation, a DoE surface response design was used to study the influence of formulation and process parameters on the particle size distribution, powder bulk properties, emulsion reconstitution ability, drug stability and process yield of spray-dried products. Two-fluid nozzle geometry was identified, studied and confirmed to be important for most product critical quality attributes. Models obtained after the study showed acceptable coefficients of determination and provided good insight in the relationship governing the process and product characteristics. Five model optimised products showed adequate process yield, suitable particle size distribution, good reconstitution ability and improved dissolution profile, when compared to a non-lipid-based tablet and the pure drug. However, the obtained dry emulsion powders exhibited poor flow character according to the Carr index. The optimised product was further analysed with NMR during lipolysis to gain insight into the species formed during digestion and the kinetics of their formation.


Subject(s)
Drug Delivery Systems/methods , Emulsions/chemical synthesis , Simvastatin/chemical synthesis , Technology, Pharmaceutical/methods , Chemical Phenomena , Desiccation/methods , Drug Stability , Emulsions/administration & dosage , Glycerides/administration & dosage , Glycerides/chemical synthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemical synthesis , Particle Size , Polysorbates/administration & dosage , Polysorbates/chemical synthesis , Simvastatin/administration & dosage , Solubility , Tablets
4.
Curr Org Synth ; 16(5): 652-670, 2019.
Article in English | MEDLINE | ID: mdl-31984887

ABSTRACT

Cardiovascular diseases are among the most threatening problems being faced by twenty-first century humans. The core cause of these diseases is high cholesterol level. Simvastatin (1: Synvinolin) is a well-known cholesterol-lowering drug marketed under the trade name Zocor®, which significantly reduces the risk of cardiovascular diseases related to hypercholesterolemia and is effective in lowering the total plasma cholesterol, low-density and very low-density lipoprotein cholesterol. It also enhances the high-density lipoprotein cholesterol. This review article aims to provide an overview of several chemical and biological methods utilized for the production of simvastatin in high yields and purity. Many robust and scalable methods have been described using lovastatin (2: Mevinolin) as a starting material, produced by the fungal strain of Aspergelius terreus. Enzymatic synthesis of simvastatin is also highlighted in this review. In addition, detailed experimental conditions, as well as the compatibility for industrial-scale preparations of simvastatin are also discussed.


Subject(s)
Anticholesteremic Agents/chemical synthesis , Simvastatin/chemical synthesis
5.
Drug Dev Ind Pharm ; 44(10): 1583-1590, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29847182

ABSTRACT

Simvastatin (Sim) is a widely known drug in the treatment of hyperlipidemia, which has attracted so much attention in bone regeneration due to its potential osteoanabolic effect. However, repurposing of Sim in bone regeneration will require suitable delivery systems that can negate undesirable off-target/side effects. In this study, we have investigated a new lipid nanoparticle (NP) platform that was fabricated using a binary blend of emulsifying wax (Ewax) and glyceryl monooleate (GMO). Using the binary matrix materials, NPs loaded with Sim (0-500 µg/mL) were prepared and showed an average particle size of about 150 nm. NP size stability was dependent on Sim concentration loaded in NPs. The suitability of NPs prepared with the binary matrix materials in Sim delivery for potential application in bone regeneration was supported by biocompatibility in pre-osteoclastic and pre-osteoblastic cells. Additional data demonstrated that biofunctional Sim was released from NPs that facilitated differentiation of osteoblasts (cells that form bones) while inhibiting differentiation of osteoclasts (cells that resorb bones). The overall work demonstrated the preparation of NPs from Ewax/GMO blends and characterization to ascertain potential suitability in Sim delivery for bone regeneration. Additional studies on osteoblast and osteoclast functions are warranted to fully evaluate the efficacy of Sim-loaded Ewax/GMO NPs using in-vitro and in-vivo approaches.


Subject(s)
Bone Regeneration/drug effects , Drug Delivery Systems/methods , Emulsifying Agents/chemical synthesis , Glycerides/chemical synthesis , Nanoparticles/chemistry , Simvastatin/chemical synthesis , Animals , Bone Regeneration/physiology , Drug Evaluation, Preclinical/methods , Drug Repositioning/methods , Emulsifying Agents/administration & dosage , Glycerides/administration & dosage , Mice , Nanoparticles/administration & dosage , Osteoblasts/drug effects , Osteoblasts/physiology , RAW 264.7 Cells , Simvastatin/administration & dosage , Waxes/chemical synthesis , Waxes/pharmacology
6.
Drug Dev Ind Pharm ; 41(1): 22-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24160569

ABSTRACT

Simvastatin has low aqueous solubility resulting in low oral bioavailability (5%) and thus presents a challenge in formulating a suitable dosage form. To improve the aqueous solubility, a solid dispersion formulation of Simvastatin was prepared by lyophilization utilizing skimmed milk as a carrier. Six different formulations were prepared with varying ratios of drug and carrier and the corresponding physical mixtures were also prepared. The improvement of amorphous state through solid dispersion was confirmed by differential scanning calorimetry and X-ray diffraction studies. The optimum drug-to-carrier ratio of 1:9 enhanced solubility nearly 30-fold as compared to pure drug. In-vitro drug release studies exhibited a cumulative release of 86.69% as compared to 25.19% for the pure drug. Additionally, scanning electron microscopy studies suggested the conversion of crystalline Simvastatin to an amorphous form. In a Triton-induced hyperlipidemia model, a 3-fold increase in the lipid lowering potential was obtained with the reformulated drug as compared to pure drug. These results suggest that solid dispersion of Simvastatin using skimmed milk as carrier is a promising approach for oral delivery of Simvastatin.


Subject(s)
Chemistry, Pharmaceutical/methods , Milk/chemistry , Simvastatin/chemical synthesis , Animals , Calorimetry, Differential Scanning/methods , Microscopy, Electron, Scanning/methods , Milk/metabolism , Simvastatin/metabolism , Solubility , X-Ray Diffraction/methods
8.
Biopolymers ; 93(9): 755-63, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20577995

ABSTRACT

The fungal polyketide lovastatin is a cholesterol lowering agent that is an immediate precursor to a multi-billion dollar drug, simvastatin (Zocor). Lovastatin is produced by an iterative type I polyketide synthase known as LovB and a partner enoyl reductase (LovC). There is evidence that a Diels-Alderase enzyme activity is utilized in its biosynthesis. This review examines the biosynthesis of lovastatin, as well as of compactin, equisetin, cytochalasins, and solanapyrones, which are other structurally related polyketides that appear to utilize a Diels-Alderase.


Subject(s)
Lovastatin/biosynthesis , Polyketide Synthases/metabolism , RNA, Catalytic/metabolism , Cytochalasins/biosynthesis , Cytochalasins/chemistry , Lactones/chemistry , Lactones/metabolism , Lovastatin/analogs & derivatives , Lovastatin/chemistry , Pyrrolidinones/chemistry , Pyrrolidinones/metabolism , Simvastatin/chemical synthesis , Simvastatin/chemistry , Tetrahydronaphthalenes/chemistry , Tetrahydronaphthalenes/metabolism
9.
Biomed Khim ; 54(3): 301-13, 2008.
Article in Russian | MEDLINE | ID: mdl-18712086

ABSTRACT

A molecular complex of simvastatin (SV) and glycyrrhyzic acid (GA) (at the ratio of 1 : 4), has been synthesized. The complex named "simvaglyzin" (SVG) was stable in aqeous and aqua-alcohol solutions at GA concentrations exceeding 0.2 mM. In vitro SVG acted as uncompetitive inhibitor of 3-hydroxy-3-methyl-glutaryl-CoA reductase (Ki = 94 nM). Appearance of this inhibitory activity is associated with the cytochrome P450-dependent conversion of SVG. The addition of 1 mM methyrapone into incubation medium fully prevented the inhibition of 3-HMG-CoA reductase. SV and SVG (used at 300 nM concentration) inhibited mevalonate synthesis rate by 39.15+/-8.27% and 38.85+/-3.04%, respectively. In vivo SVG showed dose-dependent cholesterol-lowering effect. In rats the cholesterol-lowering effect of SVG used at daily doses corresponding to 66 and 100 mg/kg of SV was equal to the effect of the daily dose 200 mg/kg of SV. The decreases of total cholesterol level in blood serum were 7%, 9% and 8%, respectively. Myotoxicity of those SVG doses estimated by creatine phosphokinase (CPK) activity in blood serum was lower than that of SV. In rats treated with SV the activity of CPK increased by 79% (p<0.01), while in SVG treated rats by 30% and 36% (p<0.05). Any increase of hepatotoxicity markers alanine aminotransferse or aspartate aminotransferase in blood serum was not observed. The data suggest pharmacological synergism attributed to the SV-GA complex formation and elevated safety of the resultant complex compared with the parent compound.


Subject(s)
Glycyrrhizic Acid/analogs & derivatives , Glycyrrhizic Acid/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/analogs & derivatives , Simvastatin/pharmacology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Creatine Kinase/blood , Glycyrrhizic Acid/chemical synthesis , Glycyrrhizic Acid/toxicity , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemical synthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Liver/drug effects , Liver/enzymology , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Rats , Rats, Wistar , Simvastatin/chemical synthesis , Simvastatin/toxicity
10.
J Pharm Biomed Anal ; 45(4): 642-7, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17697761

ABSTRACT

Unknown by-product in Simvastatin synthesis from Lovastatin was found. The elucidation of this molecular structure by means of (1)H and (13)C NMR spectroscopy, HPLC/MS, MS/MS and FT-IR was shown. The mentioned by-product, originated during Merck Sharp and Dhome synthesis scheme was isolated in the second-last step replacing butylamine with benzylamine. The spectroscopic results agreed with a molecular formula C(32)H(43)NO(3). The proposed structure of this compound, characterised by the presence of a conjugated dienic system in the heptanoic acid amide residue, was alpha,beta,gamma,delta unsaturated Simvastatin N-benzylamide.


Subject(s)
Lovastatin/chemistry , Simvastatin/analogs & derivatives , Simvastatin/chemical synthesis , Chromatography, High Pressure Liquid , Magnetic Resonance Spectroscopy , Molecular Conformation , Simvastatin/chemistry , Spectroscopy, Fourier Transform Infrared , Tandem Mass Spectrometry
11.
Appl Environ Microbiol ; 73(7): 2054-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17277201

ABSTRACT

Simvastatin is a semisynthetic derivative of the fungal polyketide lovastatin and is an important drug for lowering cholesterol levels in adults. We have developed a one-step, whole-cell biocatalytic process for the synthesis of simvastatin from monacolin J. By using an Escherichia coli strain overexpressing the previously discovered acyltransferase LovD (X. Xie, K. Watanabe, W. A. Wojcicki, C. C. Wang, and Y. Tang, Chem. Biol. 13:1161-1169, 2006), we were able to achieve >99% conversion of monacolin J to simvastatin without the use of any chemical protection steps. The key finding was a membrane-permeable substrate, alpha-dimethylbutyryl-S-methyl-mercaptopropionate, that was efficiently utilized by LovD as the acyl donor. The process was scaled up for gram-scale synthesis of simvastatin. We also demonstrated that simvastatin synthesized via this method can be readily purified from the fermentation broth with >90% recovery and >98% purity as determined by high-performance liquid chromatography. Bioconversion using high-cell-density, fed-batch fermentation was also examined. The whole-cell biocatalysis can therefore be an attractive alternative to currently used multistep semisynthetic transformations.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemical synthesis , Simvastatin/chemical synthesis , Acyltransferases/metabolism , Catalysis , Chromatography, High Pressure Liquid , Escherichia coli/metabolism , Fermentation , Naphthalenes/metabolism
12.
Chem Biol ; 13(11): 1161-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17113998

ABSTRACT

The natural product lovastatin and its semisynthetic, more effective derivative, simvastatin, are important drugs for the treatment of hypercholesterolemia. Here, we report the biochemical characterization of a dedicated acyltransferase, LovD, encoded in the lovastatin biosynthetic pathway. We demonstrate that LovD has broad substrate specificity towards the acyl carrier, the acyl substrate, and the decalin acyl acceptor. LovD can efficiently catalyze the acyl transfer from coenzyme A thioesters or N-acetylcysteamine (SNAC) thioesters to monacolin J. When alpha-dimethylbutyryl-SNAC was used as the acyl donor, LovD was able to convert monacolin J and 6-hydroxyl-6-desmethylmonacolin J into simvastatin and huvastatin, respectively. Using the Escherichia coli LovD overexpression strain as a whole-cell biocatalyst, preparative amounts of simvastatin were synthesized in a single fermentation step. Our results demonstrate LovD is an attractive enzyme for engineered biosynthesis of pharmaceutically important cholesterol-lowering drugs.


Subject(s)
Acyltransferases/metabolism , Anticholesteremic Agents , Fungal Proteins/metabolism , Lovastatin/biosynthesis , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Acyltransferases/genetics , Aspergillus/genetics , Catalysis , Escherichia coli/metabolism , Fungal Proteins/genetics , Lovastatin/analogs & derivatives , Mutation , Simvastatin/chemical synthesis , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...