ArticlePDF Available

New Synthesis of Simvastatin

Authors:

Abstract and Figures

A noninfringing synthesis of simvastatin 1, starting from lovastatin 2, is presented. This synthesis features the protection of the free hydroxyl group of the lovastatin with 3,4-dihydro-2H-pyran (DHP) and opening of the lactone ring with n-BuNH2 to afford amide 4 as a key intermediate.
Content may be subject to copyright.
PLEASE SCROLL DOWN FOR ARTICLE
This article was downloaded by:
[North Carolina State University]
On:
13 November 2008
Access details:
Access Details: [subscription number 783677828]
Publisher
Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,
37-41 Mortimer Street, London W1T 3JH, UK
Synthetic Communications
Publication details, including instructions for authors and subscription information:
http://www.informaworld.com/smpp/title~content=t713597304
New Synthesis of Simvastatin
Radha Krishna Singamsetty a; Satish Kumar Vujjini a; Nagaraju Manne a; Brahmeshwara Rao Mandava
Venkata Naga a; Vurimidi Himabindu b; Apurba Battacharya a; Mahesh Reddy Ghanta a; Rakeshwar
Bandichhor a
a Innovation Plaza, Dr. Reddy's Laboratories Ltd., Bachupally, Qutubullapur, India b Department of Chemistry,
Institute of Science and Technology, J. N. T. University, Kukatpally, Hyderabad, India
Online Publication Date: 01 January 2008
To cite this Article Singamsetty, Radha Krishna, Vujjini, Satish Kumar, Manne, Nagaraju, Venkata Naga, Brahmeshwara Rao
Mandava, Himabindu, Vurimidi, Battacharya, Apurba, Ghanta, Mahesh Reddy and Bandichhor, Rakeshwar(2008)'New Synthesis of
Simvastatin',Synthetic Communications,38:24,4452 — 4459
To link to this Article: DOI: 10.1080/00397910802369570
URL: http://dx.doi.org/10.1080/00397910802369570
Full terms and conditions of use: http://www.informaworld.com/terms-and-conditions-of-access.pdf
This article may be used for research, teaching and private study purposes. Any substantial or
systematic reproduction, re-distribution, re-selling, loan or sub-licensing, systematic supply or
distribution in any form to anyone is expressly forbidden.
The publisher does not give any warranty express or implied or make any representation that the contents
will be complete or accurate or up to date. The accuracy of any instructions, formulae and drug doses
should be independently verified with primary sources. The publisher shall not be liable for any loss,
actions, claims, proceedings, demand or costs or damages whatsoever or howsoever caused arising directly
or indirectly in connection with or arising out of the use of this material.
New Synthesis of Simvastatin
Radha Krishna Singamsetty,
1
Satish Kumar Vujjini,
1
Nagaraju Manne,
1
Brahmeshwara Rao Mandava Venkata Naga,
1
Vurimidi Himabindu,
2
Apurba Battacharya,
1
Mahesh Reddy
Ghanta,
1
and Rakeshwar Bandichhor
1
1
Innovation Plaza, Dr. Reddy’s Laboratories Ltd., Bachupally,
Qutubullapur, India
2
Department of Chemistry, Institute of Science and Technology,
J. N. T. University, Kukatpally, Hyderabad, India
Abstract: A noninfringing synthesis of simvastatin 1, starting from lovastatin 2,is
presented. This synthesis features the protection of the free hydroxyl group of the
lovastatin with 3,4-dihydro-2H-pyran (DHP) and opening of the lactone ring with
n-BuNH
2
to afford amide 4as a key intermediate.
Keywords: 3,4-Dihydro-2H-pyran, low-density lipoprotein, methylation,
simvastatin
INTRODUCTION
Simvastatin
[1]
1is a semisynthetic version of a fermentation product 2of
Aspergillus terreus. Orally active simvastatin 1(Fig. 1), a prodrug that
gets hydrolyzed in vivo to the corresponding hydroxy acid, which is
responsible for eliciting pharmacological effects.
[2]
Simvastatin 1targets
a specific enzyme that is responsible for catalyzing the conversion of 3-
hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) to mevalonate.
Received April 15, 2008.
DRL-IPD Communication Number IPDO-IPM-00085.
Address correspondence to Rakeshwar Bandichhor, Innovation Plaza, IPD,
R&D, Dr. Reddy’s Laboratories Ltd., Survey Nos. 42, 45, 46, & 54, Bachupally,
Qutubullapur, R. R. Dist. 500073, A. P., India. E-mail: rakeshwarb@drreddys.com
Synthetic Communications
1
, 38: 4452–4459, 2008
Copyright #Taylor & Francis Group, LLC
ISSN: 0039-7911 print=1532-2432 online
DOI: 10.1080/00397910802369570
4452
Downloaded By: [North Carolina State University] At: 14:57 13 November 2008
The mode of action involves the inhibition of (HMG-CoA) reductase and
eventually lowers the low-density lipoprotein (LDL) level.
[2]
The discovery of lovastatin 2
[3]
led to the development of a more
potent LDL lowering agent, simvastatin (synvinolin) 1, way back in
1991 by Merck. Syntheses of lovastatin 2and simvastatin 1have been
extensively explored.
[4]
However, most of the developed syntheses except
a few appear to be industrially incompatible. Askin et al.
[1]
reported an
elegant synthesis of 1, although it involves expensive bis-silyloxy protec-
tions of naked hydroxy groups. Thaper et al.
[5]
published a relatively
better large-scale preparation method that incorporates four steps, how-
ever, the use of more expensive amine (cyclopropyl amine) for amidation
appears to be less attractive. Herein we report a cost-effective and
noninfringing semisynthetic route for 1on a large scale.
RESULTS AND DISCUSSION
Methylation at the 2-position in lovastatin 2to obtain simvastatin 1,
without a protecting hydroxy group and using excess lithium pyrrolidide
and methyl iodide, also allows the formation of side products that make
the synthesis cumbersome. However, in the prior process, different pro-
tecting groups
[1]
such as tert-butyldimethylsilyl (TBDMS) (expensive), tri-
methylsilyl (TMS), and dimethoxy propane were successfully employed.
In our efforts to develop a robust and scaleable route for simvastatin
1as shown in Scheme 1, we chose lovastatin 2, a commercially available
starting material. Use of 3,4-dihydro-2H-pyran (DHP), one of the suita-
ble protecting groups in hydroxyl protection, was found to be cost-effec-
tive, nonhazardous, and easy to handle, and overall the transformation
was high yielding.
Parallel to the development of the synthesis of 1, we also prepared all
the reported impurities 710 (Fig. 2) in significant quantities. The spec-
troscopic data of these impurities are in complete agreement with the
related known values.
Figure 1. Structure of simvastatin 1.
New Synthesis of Simvastatin 4453
Downloaded By: [North Carolina State University] At: 14:57 13 November 2008
Tetrahydro pyran (THP) protection in the first step was found to be
concentration dependent. The reaction time in this step was less at lower
dilution, and the transformation was also efficient.
It was also observed that 2.2 eq. of DHP with respect to 3was
more than sufficient to achieve better yield and purity. Optimization
studies of hydroxy protection stage revealed that the 1.4 to 1.8 eq. of
DHP was not enough to effect the optimal conversion. Eventually 2.0
eq. of the same was found to be ideal for this transformation as shown
in Table 1 (entry 4).
Scheme 1. Synthesis of simvastatin 1.
4454 R. K. Singamsetty et al.
Downloaded By: [North Carolina State University] At: 14:57 13 November 2008
The number of equivalence of n-butyl amine was also investigated.
An amount of 1.4 eq. of n-butyl amine was optimal for the preparation
of amide 4. Optimization results are summarized in Table 2 (see entry 2).
In further transformation, a solution of 4.0 eq. n-BuLi and pyrroli-
dine each was added to a solution of 4in THF. After stirring for 1 h,
methyl iodide was added slowly to the reaction mass and subsequently
quenched upon completion of reaction. The pyrrolidine hydrochloride
salt was removed by extraction, and the organic layer was concentrated
Figure 2. Structure of reported impurities 7–10.
Table 1. Quantification of DHP in the conversion of 2to 3
Purity by HPLC
(%)
Entry Input 2(g) H
2
SO
4
(eq.) DHP (eq.) 32
1 5.0 0.05 1.4 71.27 27.82
2 5.0 0.05 1.6 94.48 4.34
3 5.0 0.05 1.8 96.98 2.72
4 5.0 0.05 2.0 98.93 0.46
5 10.0 0.05 2.2 97.46 0.09
New Synthesis of Simvastatin 4455
Downloaded By: [North Carolina State University] At: 14:57 13 November 2008
to afford amide 5as syrup with excellent yield (>99%) and purity (high
pressure liquid chromatography [HPLC]). Optimization results regarding
number of equivalence of bases, alkylating agent, and solvent are sum-
marized in Table 3 (entry 6; best result at 100-g scale).
Deprotection, hydrolysis, and ammonium salt formation to obtain 6
were accomplished in one pot with an overall yield of 86%, and the opti-
mization results are summarized in Table 4. Optimization of number of
equivalents of HCl involved in the deprotection was essential to effect
transformation efficiently. An amount of 1.3 eq. of HCl was optimal
for the preparation of 6(see Table 4, entry 7).
Finally, p-TsOH-catalyzed lactonization of 6in toluene and acetoni-
trile afforded 1, with 93% yield and 99% purity.
EXPERIMENTAL
The
1
H and
13
C NMR spectra were recorded in DMSO-d6 at 400 MHz,
on a Varian Gemini NMR spectrometer. The chemical shifts are reported
in dppm relative to TMS. The mass spectrum (70 eV) was recorded on
HP-5989a LC-MS spectrometer. The solvents and reagents were used
without any purification.
Table 2. Optimization of n-butyl amine quantity in the conversion of 3to 4
Purity by
HPLC (%)
Entry Input 2(g) Residue (g) n-Butyl amine (eq.) 43
1 5.0 7.1 1.2 93.33 3.18
2 5.0 7.8 1.4 97.72 0.21
Table 3. Quantification of reagents and solvent
Entry Input 4(g)
Residue
(g)
n-Buli
(eq.)
Pyrrolidine
(eq.)
MeI
(eq.)
THF
(volumes with
respect to 4)
Conversion
of 5(%)
1 10.0 11.0 7.5 8.8 7.16 13.0 99.67
2 10.0 11.5 7.5 7.5 7.16 13.0 99.67
3 10.0 11.4 4.5 5.8 7.16 10.0 99.57
4 10.0 11.1 4.0 5.3 7.16 10.0 99.32
5 10.0 11.0 3.5 4.8 5.0 6.0 98.90
6 100.0 115.0 4.0 4.0 4.0 6.0 99.60
4456 R. K. Singamsetty et al.
Downloaded By: [North Carolina State University] At: 14:57 13 November 2008
2-Methyl-butyricacid-8-[6-butylcarbamoyl-3-hydroxy-5-(tetrahydro-
pyran-2-yloxy)-hexyl]-3,7-dimethyl-1,2,3,7,8,8a-hexahydro-
naphthalen-1-yl Ester 4
A solution of sulphuric acid (0.06 mL) in THF (10 mL) was added to a
solution of 3,4-dihydro-2H-pyran (0.54 mol 49.4 mL) in THF (40 mL).
After the addition over a period of 10 min and stirring for 5 min, lovas-
tatin 2(0.25 mol, 100 g) was added at 28 C. After stirring for 45 min at
36 C, n-BuNH
2
(0.32 mol, 34 mL) was added. After additional stirring
for 4 h at 49 C, the reaction mixture was concentrated under reduced
pressure to afford crude material 4as brown syrup. Crude yield:
148.3 g (>100%); purity by HPLC: 98%;
1
H NMR (400 MHz, DMSO-
d6): d7.77 and 7.69 (t, J¼6.4 Hz, 1H), 5.94 (d, J¼9.6 Hz, 1H), 5.78
(dd, J¼9.6, 6.0 Hz, 1H), 5.48 (s, 1H), 5.24–5.18 (m, 1H), 4.63–4.59 (m,
1H), 4.36 (d, J¼6.0 Hz, 1H), 4.26 (d, J¼5.6 Hz, 1H), 4.12–4.00 (m,
1H), 3.82–3.70 (m, 1H), 3.10–2.94 (m, 2H), 2.50 (t, J¼4.0 Hz, 1H),
2.44–2.20 (m, 4H), 1.98–1.22 (m, 22H), 1.03–1.00 (m, 6H), 0.88–0.80
(m, 9H); MS: m=e¼584.4 (M þNa
þ
).
2,2-Dimethyl-butyricacid-8-[6-butylcarbamoyl-3-hydroxy-5-
(tetrahydro-pyran-2-yloxy)-hexyl]-3,7-dimethyl-1,2,3,7,8,8a-
hexahydro-naphthalen-1-yl Ester 5
To a solution of pyrrolidine (1 mol, 82mL) in THF (278 mL), 1.6 M n-
BuLi (1 mol, 622 mL) was added at 25 C. After the addition over a per-
iod of 1 h and stirring for 25 min, a solution of 4(0.25 mol, 139 g) in THF
(556 mL) was added. After stirring for additional 1 h at 45 C, methyl
iodide (1 mol, 63mL) was added to the reaction mixture and stirred for
1.5 h. The reaction mixture was quenched with 1N aq. HCl (915 mL),
and the organic layer was separated, concentrated, and dried under
Table 4. Optimization of acid=base on THP deprotection and amide hydrolysis
Entry 5 (g) Quantity=yield HCl (eq.) NaOH (eq.) HPLC purity (%)
1 5.0 1.7=49 4.0 4.0 94.97
2 5.0 0.4=11 0.53 4.0 94.97
3 5.0 1.9=54 0.45 4.0 93.16
4 5.0 2.7=77 1.0 4.0 95.37
5 5.0 1.2=34 1.0 3.6 97.58
6 5.0 2.8=80 1.3 4.0 98.11
7 50.0 30.5=87 1.3 4.0 98.09
New Synthesis of Simvastatin 4457
Downloaded By: [North Carolina State University] At: 14:57 13 November 2008
reduced pressure to afford crude material 5as a thick brown syrup.
Crude yield: 154 g (>100%);
1
H NMR (400 MHz, DMSO-d6): d7.77
and 7.69 (t, J¼6.4 Hz, 1H), 5.94 (d, J¼9.6 Hz, 1H), 5.77 (dd, J¼9.6,
6.0 Hz, 1H), 5.48 (s, 1H), 5.20–5.18 (m, 1H), 4.63–4.59 (m, 1H), 4.36
(d, J¼6.0 Hz, 1H), 4.27 (d, J¼5.6 Hz, 1H), 4.10–4.02 (m, 1H), 3.79–
3.71 (m, 1H), 3.04–2.95 (m, 2H), 2.50 (t, J¼4.0 Hz, 1H), 2.39–2.23 (m,
4H), 1.99–1.20 (m, 21H), 1.26–1.00 (m, 9H), 0.88–0.75 (m, 9H); MS:
m=e¼598.4 (M þNa
þ
).
(3R,5R)-7-[(1S,2S,6R,8S,8aR)-8-[(2,2-Dimethylbutanoyl]oxy]-2,6-
dimethyl-1,2,6,7,8,8a-hexahydronaphthalen-1-yl]-3,5-dihydroxyhepta-
noicacidammonium Salt 6
[5]
HCl (0.15 mol, 15 mL) was added to a solution of 5(0.12 mol, 67.6 g) in
methanol (149 mL) at 2 C. After stirring for 4 C at 45 min to afford in
situ amide intermediate, 10% aq. NaOH (0.5 mol, 188 mL) was added
to the reaction mixture. After additional stirring for 4 h at 77 C, the reac-
tion mixture was cooled to 33 C and water (135 mL) was added. After
adjusting the pH to 4.7 (by adding 10% HCl) at 15 C, the reaction mix-
ture was extracted with EtOAc (338 mL), and 28% ammonium hydroxide
solution (0.24 mol, 30mL) in methanol (30 mL) was added to the organic
layer. After stirring for 30 min at 28 C, the reaction mixture was cooled
to 3 C and stirred additionally for 1 h to afford 6as a light-brown-
colored solid in 87% isolated yield (39.5 g) and 98% purity (HPLC).
1
H
NMR was found to be satisfactory as per literature.
2,2-Dimethylbutanoicacid(1S,2R,7S,8S,8aR)1,2,3,7,8a-hexahydro-
3,7,dimethyl-8-[2-[(2R,4R)-tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-
yl]ethyl]-1-naphthalenyl Ester 1
[5]
A solution of p-TsOH (0.003 mol, 0.5 g) in toluene (421 mL) was added to
a solution of 6(0.055 mol, 25 g) in acetonitrile (173 mL) after stirring for
8 h at 83 C, water, a by-product, was azeotropically distilled out. The
reaction mixture was cooled to 43 C and filtered. The solvent was com-
pletely distilled from the filtrate at 60 C under vacuum, and methanol
(300 mL) was added to the residue at 45 C. After stirring for 20 min,
activated carbon (2.5 g) was added and stirred for an additional period
of 0.5 h. The reaction mixture was filtered, washed with methanol
(50 mL), and heated to 38 C. Water (350 mL) was added to the filtrate
and cooled to 13 C over a period of 2 h. The obtained solid mass was
filtered, followed by washing with a precooled mixture (110 mL) of
4458 R. K. Singamsetty et al.
Downloaded By: [North Carolina State University] At: 14:57 13 November 2008
methanol=water (1:1). The wet material was dried over a period of 2 h at
54 C to afford 1(21.5 g) in 93% yield and 99% purity (HPLC).
1
HNMR
was found to be satisfactory as per literature.
ACKNOWLEDGMENTS
We acknowledge Dr. Reddy’s Laboratories for financial and analytical
support.
REFERENCES
1. Askin, D.; Verhoeven, T. R.; Liu, T. M.-H.; Shinkai, I. Synthesis of synvino-
lin: Extremely high conversion alkylation of an ester enolate. J. Org. Chem.
1991,56, 4929–4932.
2. Vickers, S.; Duncan, C. A.; Chen, I.-W.; Rosegy, A.; Duggan, E. Metabolic
disposition studies on simvastatin, a cholesterol-lowering prodrug. Drug
Metab. Dispos. 1989,51, 138–145.
3. (a) Kysika, R.; Kren, V. Determination of lovastatin (mevinolin) and mevino-
linic acid in fermentation liquids. J. Chromatogr. 1993,630, 415–417. (b)
Moore, R. N.; Bigam, G.; Chan, J. K.; Hogg, A. M.; Thomas, T.; Verderas,
J. C. Biosynthesis of the hypocholesterolemic agent mevinolin by Aspergillus
terreus: Determination of the origin of carbon, hydrogen, and oxygen atoms
by carbon-13 NMR and mass spectrometry. J. Am. Chem. Soc. 1985,107,
3694–3701. (c) Rosen, T.; Heathcock, C. H. The synthesis of mevinic acids.
Tetrahedron 1986,42, 4909–4951.
4. Hoffman, W. F.; Alberts, A. W.; Anderson, P. S.; Chem, J. S.; Smith, R.L.;
Willward, A. K. 3-Hydroxy-3-methylglutaryl-coenzyme A reductase inhibi-
tors, 4: Side-chain ester derivatives of mevinolin. J. Med. Chem. 1986,29,
849–852. Also see Ref. 1.
5. Thaper, R. K.; Kumar, Y.; Kumar, S. M. D.; Misra, S.; Khanna, J. M. A cost-
efficient synthesis of simvastatin via high-conversion methylation of an alkox-
ide ester enolate. Org. Process Res. Dev. 1999,3, 476–479.
New Synthesis of Simvastatin 4459
Downloaded By: [North Carolina State University] At: 14:57 13 November 2008
Chapter
Nowadays, much attention is devoted to develop green, safe, and energy-efficient chemistry for drug synthesis with minimal impact on environment and health. Drugs are typically synthesized by chemical means using solvents and chemical reagents generating toxic wastes and by-products which pollute the environment. Therefore, pharmaceutical industries are gradually adopting environment-friendly green chemistry for manufacturing drugs. These efforts led to the development of novel aqueous-based chemistry with biocatalysts that reduce solvent and reagent consumption resulting in minimum impact on environment. Such chemistry has been successfully implemented to manufacture life-saving drugs. These include cholesterol-lowering statin drugs like lovastatin, simvastatin, atorvastatin, and rosuvastatin which inhibit HMG-Co-A reductase responsible for cholesterol synthesis in the liver. Increased LDL-cholesterol in blood is the key risk factor for cardiovascular disease as it leads to atherosclerotic plaque deposit inside blood vessels causing blockage. These can result in stroke causing death or lifetime disability. This chapter provides an overall review of industry progress and accomplishments in embracing green chemistry for commercial synthesis of statin drugs.
Article
ChemInform is a weekly Abstracting Service, delivering concise information at a glance that was extracted from about 200 leading journals. To access a ChemInform Abstract of an article which was published elsewhere, please select a “Full Text” option. The original article is trackable via the “References” option.
Article
Die 3-Hydroxy-3-methylglutaryl-Coenzym-A-Reduktase (HMG- CoA-Reduktase) katalysiert die Bildung von Mevalonsäure, einer frühen Zwischenstufe in der Biosynthese von Cholesterol. Die Statine sind HMG-CoA-Reduktase-Inhibitoren, die sehr effizient die Cholesterol-Serumwerte senken und sehr häufig zur Behandlung der Hypercholesterolämie, der Ursache der Arteriosklerose, verordnet werden. Dieser Review beschreibt den medizinischen Hintergrund dieser Krankheit, die Entdeckung der Statine, ihre Biosynthese und die Methoden der industriellen Herstellung der weltweit umsatzstärksten Arzneiwirkstoffe. The 3-Hydroxy-3-methylglutaryl-coenzyme A reductase (HMG- CoA reductase) catalyses the formation of mevalonic acid, an early intermediate in the biosynthesis of cholesterol. Statins are HMG-CoA reductase inhibitors that efficiently lower serum cholesterol levels and are widely prescribed in the treatment of hypercholesterolemia, which can cause arteriosclerosis. This review describes the medical background of this disease, the discovery of the statins, their biosynthesis and the methods of industrial production of some of the best selling active pharmaceutical ingredients worldwide.
Article
Typescript. Thesis (Ph. D.)--University of South Carolina, 1989. Includes bibliographical references.
Article
A rapid and simple HPLC method for the determination of Lovastatin (mevinolin) and mevinolinic acid in fermentation fluids of Aspergillus terreus using a Separon SGX C18 column and methanol-18 mM orthophosphoric acid (77.5:22.5, v/v) as mobile phase with detection at 238 nm is described. The detection limit of Lovastatin and mevinolinic acid was 20–30 ng/ml.
Article
Modification of the 2(S)-methylbutyryl moiety of mevinolin led to a series of side chain ester derivatives. A systematic exploration of the structure-activity relationships showed that the introduction of an additional aliphatic group on the carbon alpha to the carbonyl group increased potency. This observation led to the synthesis of compound 16, which has about 2.5 times the intrinsic inhibitory activity of mevinolin.
Article
A cost-efficient synthesis of simvastatin (2), starting from mevinolin (lovastatin) (1a) or its precursor mevinolinic acid (1b), is reported. This synthesis involves the use of a new intermediate, lovastatin cyclopropylamide (3), eliminating two chemical steps of protection and deprotection of the open dihydroxy form of (1a). Synthesis is based on the high-conversion methylation of an alkoxide ester enolate and involves only four chemical steps. Methylation reaction conditions have been optimized to get >99.5% conversion. Process is economical on large-scale and product (2) is obtained in 85% overall yield.
Article
A efficient process for the commercial preparation of the therapeutically important cholesterol lowering drug synvinolin (2: simvastatin, ZOCOR) from mevinolin (1: lovastatin, MEVACOR) is reported. The synthesis relies upon deactivation of the delta-lactone carbonyl toward enolization via conversion to the bis[(tert-butyldimethylsilyl)oxy] butylamide 7. An extremely high conversion (99.7%) ester enolate alkylation of 7 affords 8 and 9. Subsequent desilylation and intramolecularly assisted basic amide hydrolysis in the presence of the dimethylbutyrate ester moiety yields 12, which is lactonized to 2. The overall yield from 1 to 2 is 86%.
Article
The 1H and 13C NMR spectra of mevinolin (1), an inhibitor of hydroxymethylglutaryl-coenzyme A reductase, were fully assigned by using selective homonuclear and heteronuclear decoupling and two-dimensional DEPT heteronuclear shift correlation. Sodium [1-13C]-, [2-13C]-, [1,2-13C2]-, [1-13C,18O2]-, [1-13C,2H3]-, and [2-13C,2H3]acetate as well as 18O2 and [methyl-13C]methionine were incorporated into mevinolin (1) by cultures of Aspergillus terreus ATCC 20542. Double quantum coherence (2D INADEQUATE) NMR spectra of 1 derived from sodium [1,2-13C]acetate independently confirmed the 13C NMR assignment and provided the location of intact carbon-carbon bonds from acetate. Mevinolin (1) is formed from two polyketide chains (4-carbon and 18-carbon) of acetate units coupled in head to tail fashion, with each chain bearing a methionine-derived methyl group. Material obtained from 2H and 18O incorporations was analyzed by mass spectrometry and by 13C NMR detection of α and β isotope shifts. The results show that oxygen atoms on the main chain are introduced by aerobic oxidation of a deoxygenated precursor. Several mechanisms, including a biological Diels-Alder reaction, are proposed to account for formation of bicyclic ring systems in mevinolin (1), compactin (2), and related metabolites.
Article
The biosynthesis of cholesterol is mainly regulated by 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. Because the liver is the major site of cholesterol synthesis, it is the primary target of the class of drugs known as HMG-CoA reductase inhibitors. Simvastatin (SV) is a lactone prodrug which undergoes reversible metabolism. In the hydroxy acid form (SVA) it is a potent inhibitor of HMG-CoA reductase. SV is well absorbed by rats, dogs, and humans. After an oral dose of SV, tissue distribution studies were consistent with high hepatic extraction of SV and relatively poor tissue penetration of SVA. The majority of a radioactive dose of SV is eliminated in bile. A high portal/systemic gradient for 6'-OH-SVA, an active biliary metabolite, suggests its probable reentry and indicates potential for prolongation of HMG-CoA reductase inhibition. AUC comparisons in dogs after simultaneous iv (3H) and intraportal (14C) infusions indicate that hepatic extraction is high with only 8% of SV reaching the systemic circulation unchanged. Approximately 98% and 96% of SV was bound to human and dog plasma protein, respectively. The physiological disposition of SV in dog appears to be a suitable paradigm for man. Because of its high hepatic extraction SV should be both specific and selective with respect to the inhibition of HMG-CoA reductase.
Article
Modification of the 2(S)-methylbutyryl moiety of mevinolin led to a series of side chain ester derivatives. A systematic exploration of the structure-activity relationships showed that the introduction of an additional aliphatic group on the carbon alpha to the carbonyl group increased potency. This observation led to the synthesis of compound 16, which has about 2.5 times the intrinsic inhibitory activity of mevinolin.
Determination of lovastatin (mevinolin) and mevinolinic acid in fermentation liquids
  • R Kysika
  • V Kren
Kysika, R.; Kren, V. Determination of lovastatin (mevinolin) and mevinolinic acid in fermentation liquids. J. Chromatogr. 1993, 630, 415-417. (b)