throbber
H M G-CoA Reductase Inhibitors
`
`A. Endo and K. Hasumi
`Department of Applied Biological Science, Tokyo Noko University, Fuchu, Tokyo, 183 Japan
`
`Selectively reviewing the literature published up t o October 1992
`
`Introduction
`1
`Biochemical and Pharmacological Mechanisms
`2
`HMG-CoA Reductase
`2.1
`Plasma Cholesterol
`2.2
`Mevinic Acids of Fungal Origin
`3
`3.1
`Isolation
`Biosynthesis
`3.2
`Modification
`3.3
`4
`Synthetic Analogues
`5
`References
`
`1 Introduction
`Extensive epidemiological studies performed in many countries
`have shown that increased blood cholesterol levels, or, more
`specifically, increased levels of low density lipoprotein (LDL)
`cholesterol, are a major cause of coronary heart disease. There
`is also substantial evidence that lowering total and LDL
`cholesterol levels will reduce the incidence of coronary heart
`disease.l
`In 1976, Endo et al. reported the isolation of mevastatin
`(formerly called ML 236B or compactin) (1) as a potent
`inhibitor of hydroxymethylglutaryl coenzyme A (HMG-CoA)
`reductase, the rate-limiting enzyme in endogenous cholesterol
`synthesis.2. They elucidated biochemical mechanisms for the
`
`action of me~astatin~-~ and by 1980, had shown that mevastatin
`strikingly lowers total and LDL cholesterol in patients with
`hyperch~lesterolemia.~ These findings stimulated the world-
`wide development of mevastatin analogues in the 1980s and by
`1990, three drugs, lovastatin (formerly called mevinolinlO or
`monacolin K11,12) (2), simvastatin13 (3) and pravastatin14 (4),
`
`had been marketed in many c o ~ n t r i e s . ~ ~ , ~ ~ In addition to these,
`many other mevastatin analogues have been synthesized, some
`of which are now under clinical development.
`
`2 Biochemical and Pharmacological
`Mechanisms
`2.1 HMG-CoA Reductase
`HMG-CoA reductase, a 97 kDa glycoprotein," catalyses the
`reductive deacylation of HMG-CoA to mevalonate in a two-
`step reaction (Scheme 1).
`The stereospecificity of HMG-CoA reductase is illustrated in
`Scheme 1. Only the 3s isomer of HMG-CoA is utilized in the
`reaction.1s-20 Each hydride transfer involves the pro-R or 'A'
`side of the NADPH pyridine ring,21.22 the first forming the 3S,
`
`5R-thiohemiacetal, and the second incorporating the hydrogen
`into the 5-pro-S position of (3S)-me~alonate.~~.
`23. 24
`Mevastatin analogues are reversible competitive inhibitors of
`The K, value for mammalian HMG-
`HMG-CoA r e d ~ c t a s e . ~ ~ ' ~
`
`CoA reductase is - 10 ,UM, while the Ki value for the ring-
`
`opened acids of mevastatin (5) and lovastatin (6) are in the
`range of 0.2-1 n
`
`
`
` ~~.~Thus, the affinity of HMG-CoA reductase
`
`
`for mevastatin analogues is 10000-fold or more than its affinity
`for the natural substrate, HMG-CoA. The 3,5-dihydroxy-
`heptanoic acid portion of these compounds resembles the
`HMG portion of HMG-CoA (Scheme 1).
`The 3,5-dihydroxyheptanoic acid chain of mevastatin inter-
`acts with the HMG binding domain of the enzyme's active site.
`It has been postulated that the tight binding of mevastatin is the
`result of its ability to simultaneously interact with both the
`HMG binding domain and an adjacent hydrophobic pocket
`which is not utilized in substrate binding.26
`Kinetic studies have shown that the enzymatic reaction is
`consistent with the general chemical mechanism postulated for
`
`(1) R = H
`(2) R = Me
`
`(3)
`
`C02Na
`
`" ' Y O H
`
`0
`
`(4)
`
`(5) R = H
`(6) R = Me
`
`HO ,CH3
`
`CoA-S &02H
`
`3s-HMG-COA
`
`NADPH NADP+
`
`NADPH" NADP'
`
`3 S,5 R-Mevakfyl-CoA
`thiohemiacetal
`
`Scheme 1
`
`54 1
`
`3 R-Mevalonate
`
`38-2
`
`Published on 01 January 1993. Downloaded by Reprints Desk on 19/05/2015 16:57:02.
`
`View Article Online
`
` / Journal Homepage
`
` / Table of Contents for this issue
`
`NCI Exhibit 2022
`Page 1 of 10
`
`

`
`542
`
`0 +&
`
`R'-
`
`ti I N
`
`"Do "vo "U0
`& & & &
`
`NATURAL PRODUCT REPORTS, 1993
`
`H 0 q C 0 2 H
`OH
`
`R'-
`
`(8) 0 0
`R = H
`
`R.*
`
`(9) R = 0 H
`
`,.-'
`
`(1 2) 0 ..*-
`
`\ (13) "OH
`
`(7) R = H
`(14) R = M e
`
`(10) R =Me
`
`(11) R = Me
`
`dehydrogenase catalysis in assisting direct transfer of a hydride
`ion between nucleotide and
`The pKa of this
`catalytic group is dependent on whether reduced or oxidized
`cofactor (NADPH or NADP', respectively) is bound at the
`active site. Mevastatin (1) apparently owes its inhibitory
`activity to the ability to mimic the half-reduced substrate
`mevalonate hemithioacetal. Therefore, by analogy, the 5-
`hydroxy group of mevastatin must interact with the un-
`protonated form of this catalytic group.
`Mevastatin analogues inhibit cholesterol biosynthesis in a
`variety of mammalian cell cultures at nM concentration^.^. 6 , 25
`In human skin fibroblast cultures, sterol synthesis from
`
`[14C]acetate is inhibited by 50% at - 1 nM. When mevastatin
`
`analogues are given orally to rats, sterol synthesis in the liver,
`the major organ of sterol synthesis, is acutely inhibited.,
`Mevastatin and lovastatin do not lower plasma cholesterol
`levels in rats and mice,28 but these agents are highly effective in
`reducing plasma cholesterol in dogs,29, 30
` rabbit^^^,^^
`and h ~ m a n s . ~ . ~ ~ * ~ ~ , ~ ~
`
`2.2 Plasma Cholesterol
`Cholesterol is transported in plasma in the form of lipoproteins.
`By means of ultracentrifugation six distinct classes of lipo-
`protein can be isolated from plasma. Most of the cholesterol in
`humans is carried by LDL.
`Mammalian cells from normal subjects possesses LDL
`receptors on the cell surface which bind LDL with a high-
`affinity.35 The bound LDL is incorporated into the cells and
`undergoes lysosomal digestion, leading to hydrolysis of
`cholesterol esters. The free cholesterol that is released serves to
`control the rate of cholesterol synthesis within the cells by
`down-regulating HMG-CoA reductase.l' The chief role of the
`LDL receptor is to provide a constantly available source of
`cholesterol throughout the body. Mutations of the gene
`encoding the LDL receptor results in impaired degradation of
`LDL and thus cause familial hypercholesterolemia (FH).36
`Endogenous cholesterol synthesis is decreased by exposing
`cells to LDL which facilitates delivery of exogenous cholesterol
`and thus down-regulates HMG-CoA r e d ~ c t a s e . ~ ~
`The co-ordinate regulation of LDL receptor expression and
`
`_.
`CH3CCOONa
`A
`CH3SCH2CH&H(NH*)COOH
`
`Scheme 2
`
`HMG-CoA reductase activity provides a homeostatic mech-
`anism for ensuring an adequate supply of cholesterol to cells
`such as hepatocytes which metabolize large amounts of LDL
`each day.36 HMG-CoA reductase inhibitors typified by
`mevastatin (1) block endogenous cholesterol synthesis, es-
`pecially in the liver which requires cholesterol as a substrate for
`bile acid synthesis4 To overcome the short-fall, hepatocytes
`express a greater number of LDL receptors and thereby
`
`promote influx of LDL cholesterol from ~ l a s m a . ~ ~ , ~ ' The net
`result is a decrease in plasma LDL cholesterol. Lovastatin (2)
`and simvastatin (3) are both lactones and inactive until
`metabolized in the liver to the open-ring hydroxy acids.
`
`3 Mevinic Acids of Fungal Origin
`3.1 Isolation
`Mevastatin (l), a metabolite of Penicillium citrinum, was
`isolated in 1973, filed for patent in 1974, and first described in
`the literature in 1976.2. This compound was independently
`isolated from P. brevicompactum as an antibiotic by Brown et
`al.38 Subsequently, lovastatin (2) was isolated from Monascus
`ruberll. l 2 and Aspergillus terreus,1° respectively. Lovastatin is
`slightly more potent than mevastatin in inhibiting HMG-CoA
`reductase. These compounds can be easily converted to the
`respective open-chain dihydroxy acids (5) and (6).
`Along with mevastatin, dihydrocompactin (7),39 ML-236A
`(S),2 and ML-236C (9)2 have been isolated from P. citrinum.
`Monacolin J
`monacolin L (1 l),,O dihydromonacolin L
`( l2),,l and 3a-hydroxy-3,5-dihydromonacolin L acid (1 3)42 are
`minor metabolites of M . ruber. Dihydromevinolin (14) is a
`product of A . t e r r e ~ s . , ~
`The class of compounds mentioned above, distinguished by
`a highly functionalized hexalin or octalin unit and a /3-hydroxy-
`6-lactone portion linked by an ethylene bridge, are collectively
`referred to as mevinic
`Dihydrocompactin (7) and dihydromevinolin (14) are com-
`parable to mevastatin and lovastatin, respectively, in inhibiting
`HMG-CoA reductase, while other metabolites that lack the
`side chain ester are far less active.25
`
`3.2 Biosynthesis
`[13C]Acetate, [methy1-l3C]methionine and 1802 are incorporated
`into mevastatin and lovastatin in cultures of P. citrinum, M .
`ruber and A . terreus, and the 'H and 13C NMR spectra of the
`two products have been fully assigned by a combination of
`spectral analyses. Both compounds are formed by the head-to-
`tail coupling of two polyketide chains (C, and C18) each derived
`from acetate units. The C, chain has one methionine derived
`methyl group, and a methyl group at C-6 in the bicyclic ring
`system of lovastatin is also derived from methionine (Scheme
`2).4547
`The above data suggest involvement of a biological Diels-
`Alder cyclization to generate the correct ring stereochemistry in
`a single step. Such ring forming processes have been suggested
`for a number of reduced polyketide metabolites.
`Mevinic acids isolated from M . ruber by Endo and co-
`workers include dihydromonacolin L (1 2), 3a-hydroxy-3,5-
`
`Published on 01 January 1993. Downloaded by Reprints Desk on 19/05/2015 16:57:02.
`
`View Article Online
`
`NCI Exhibit 2022
`Page 2 of 10
`
`

`
`NATURAL PRODUCT REPORTS, 1993-A. END0 AND K. HASUMI
`
`543
`
`Methionine
`
`(2)
`
`(10)
`
`(3)
`
`Reagents: i, LiOH, H,O, 100 "C; H,O+, toluene, 110°C; ii, TBSCl; iii, 2,2-dimethylbutyryl chloride; Bu"NF, HOAc
`Scheme 4
`
`Table 1 Effects of modification of the side chain ester moiety
`of lovastatin.
`
`R
`
`Relative potency"
`
`254
`
`K acid (6) can be derived from monacolin J acid (17) by M .
`r ~ b e r . ~ ~
`One possible mechanism for this conversion is the
`esterification of monacolin J acid with a-methylbutyryl-CoA to
`monacolin K acid. These results are summarized in Scheme 3.
`
`3.3 Modification
`The 2s-methylbutyryl ester of mevastatin and lovastatin, gave
`ML-236A and monacolin J, respectively, by hydrolysis with
`either alkali or carboxylesterase of the fungus Emericellu
`
`ur~guis.~O,~~ Hoffman et ul. at Merck, Sharp & Dohme (MSD)
`synthesized a series of the side chain ester analogues of
`lovastatin from monacolin J (Scheme 4).13 A systematic
`exploration of the structure-activity relationships showed that
`the introduction of an additional aliphatic group on the carbon
`01 to the carbonyl group increased potency (Table l).13 This
`observation led to the synthesis of simvastatin (3) (Scheme 4),
`which has about 2.5 times the intrinsic inhibitory activity of
`lovastatin (2).13 A process involving enolization/methylation
`of the 2s-methylbutyrate side chain of lovastatin also affords
`simvastatin highly effi~iently.~, Simvastatin has been marketed
`since 1988.
`Side chain ether analogues of lovastatin are weaker inhibitors
`of HMG-CoA reductase than the corresponding side chain
`ester analogues. Of the ether analogues prepared by Lee et al.,
`the 4-fluorobenzyl ether analogue (1 8) proved to enhance the
`potency.53
`
`" O U 0
`
`F
`
`A
`
`
`
`w
`a Potency of mevastatin is assigned a value of 100.
`
`15
`
`dihydromonacolin L acid (1 3), monacolin L (1 I), monacolin J
`(10) and monacolin K (lovastatin) (2). They have shown that
`dihydromonacolin L acid (1 5) is converted to 3a-hydroxy-3,5-
`dihydromonacolin L acid (13) by M . ruber in the presence of
`molecular oxygen.42 The
`latter can be spontaneously
`dehydrated to monacolin L acid (16), although conversion of
`exogenously added 301-hydroxy-3,5-dihydromonacolin L acid
`to monacolin L acid by M. ruber has not been successful.
`Monacolin L acid is hydroxylated to monacolin J acid (17) by
`the action of a mono~xygenase.~~ The end-product monacolin
`
`Published on 01 January 1993. Downloaded by Reprints Desk on 19/05/2015 16:57:02.
`
`View Article Online
`
`NCI Exhibit 2022
`Page 3 of 10
`
`

`
`544
`
`NATURAL PRODUCT REPORTS, 1993
`
`0
`
`0
`
`, O q r O
`
`(19) R = P-OH
`(20) R = a-OH
`
`(22) R = H
`(23) R =Me
`
`,
`
`(27) R' = CI, R2 = H
`(28) R'=H, R2=CI
`
`Modification of the hexahydronaphthalene ring 6-position in
`simvastatin (3) via oxygenation and oxa replacement by Duggan
`et al. led to the selection of (19) and (20) for pharmacological
`
`e v a l ~ a t i o n . ~ ~ These two compounds proved to be orally active
`as hypocholesterolemic agents in cholestyramine-primed dogs
`and also exhibit low peripheral plasma drug activity levels,
`which may minimize pharmacologically related side effects
`since the major site of cholesterol synthesis is the liver. (+)-6-
`Ethylmevastatin (21) is comparable to lovastatin in inhibitory
`PO tency . 55
`Pravastatin (4)14 is prepared by microbial transformati~n.~~.
`57
`The enzyme cytochrome P-450,,,
`is responsible for this
`conversion by Streptomyces carbophil~s.~~ Pravastatin is struc-
`turally different from lovastatin in that it contains a hydroxyl
`group in the hexahydronaphthalene ring, making pravastatin
`more hydrophilic than lovastatin. Pravastatin is comparable to
`mevastatin and lovastatin in inhibiting HMG-CoA reductase
`and in lowering plasma cholesterol. It has been on the market
`since 1989.
`The phosphorylated derivatives (22) and (23) are produced
`by the action of several fungal
`These derivatives are
`converted to the respective parent compounds ( 5 ) and (6) in the
`liver, when administered to rats.
`Compound L-669262 (24) is derived from simvastatin (3) by
`microbial conversion, and is 6-7
`times more active than
`simvastatin in inhibiting HMG-CoA reductase.60
`
`4 Synthetic Analogues
`Since the mid 1980s a plethora of work has been directed
`towards the preparation of synthetic analogues of mevastatin
`(1) and lovastatin (2) by many pharmaceutical companies. Of
`these studies, initial investigations at MSD served to delineate
`key structure-activity relationship for mevastatin-like mimics
`and afforded a series of moderately effective HMG-CoA
`reductase inhibitors bearing a monocyclic substituent, of which
`the ring-opened form of lactone (25) was the most potent.61.62
`In general, unless the hydroxy groups remain unsubstituted
`in an eryfhro relationship, inhibitory activity is greatly reduced.
`Furthermore, only one enantiomer of the ring-opened form of
`the lactone possesses the activity displayed by the racemate.61
`
`These findings reveal that the chiral lactone moiety is essential
`for biological activity.
`Insertion of a bridging unit other than ethyl or (E)-ethenyl
`between the Scarbin01 moiety and an appropriate lipophilic
`moiety (e.g. 2,4-dichlorphenyl) attenuates activity.61
`Further studies of a series of substituted derivatives of (25) at
`MSD provided a series of 7-[3,5-disubstituted( 1,l '-biphenyl)-2-
`yl]-3,5-dihydroxy-6-heptanoic acids, of which (26) possessed
`2.8 times the inhibitory activity of m e ~ a s t a t i n . ~ ~ X-ray
`crystallography studies on compound (26) showed it to possess
`the same chirality in the lactone ring as mevastatin. Potent
`inhibitory activity was not retained without concomitant sub-
`stitution at the 3- and 5-positions of the central phenyl ring of
`The type
`the biphenyl moiety with methyl or chloro
`and position of substituents on the external phenyl ring is
`critical. An electron-donating group (CH, or CH,O) in the 4'-
`position is detrimental, whereas a halogen (C1 or F) in this
`position is beneficial. Ring fusion of substituted biphenyls
`afforded products that were substantially less active, indicating
`that the dihedral angle between phenyl rings must be greater
`than 0" to maintain a high level of inhibitory potency.64
`Substituted naphthalene derivatives, (27) and (28), display
`about the same potency as m e ~ a s t a t i n . ~ ~
`Investigations at the Sandoz Research Institute further
`extended the findings obtained at MSD. The researchers chose
`indolyl derivatives by considering structures and molecular
`shapes of both coenzyme A and mevastatin. An extensive and
`rapid analogue program led to the choice of fluvastatin (XU62-
`320) (29) (Scheme 5) as a candidate for extensive biological
`testing.66 As compared to the respective sodium salts of
`mevastatin and lovastatin, fluvastatin is 22- and 10-fold more
`potent in inhibiting HMG-CoA reductase. However, fluvastatin
`is comparable to mevastatin in cholesterol-lowering activity in
`patients.67 This drug is now under development and expected to
`be marketed in 1993.
`Many synthetic analogues of mevastatin have been prepared
`at Hoechst AG. Baader et al. prepared compound (30), which
`is comparable to lovastatin with respect to inhibition of HMG-
`CoA reductase.68 However, the cholesterol-lowering activity of
`(30) in rabbits is slightly lower when compared to that of
`lovastatin. The same group prepared the pyridine analogue HR
`780 (31) (Scheme 6),69370 which exceeded the activity of
`
`Published on 01 January 1993. Downloaded by Reprints Desk on 19/05/2015 16:57:02.
`
`View Article Online
`
`NCI Exhibit 2022
`Page 4 of 10
`
`

`
`NATURAL PRODUCT REPORTS, 1993-A. END0 AND K. HASUMI
`
`+k
`F alp
`i -
`
`ii
`
`___L
`
`F
`
`F
`
`545
`
`iii
`
`__c
`
`CI
`
`I A
`
`F
`
`C02Na 2
`
`COOR
`
`v
`
`Reagents: i, EtOH, A, ii, ZnC1,; iii, Me,NCH=CHCHO, POCl,; NaOH; iv, NaH, Bu"Li; v, Et,B, THF; NaBH,, -78 "C; vi, MeOH, NaOH.
`Scheme 5
`
`(31)
`Reagents: i, Bu'Ph,SiCl; imidozole; ii, CH,CO,But, LDA; iii, Et,B, NaBH,; iv, MeC(OMe),H, H+; v, Bu,NF; vi, Swern oxidation; vii, base; viii,
`CF,CO,H
`
`Scheme 6
`
`Published on 01 January 1993. Downloaded by Reprints Desk on 19/05/2015 16:57:02.
`
`View Article Online
`
`NCI Exhibit 2022
`Page 5 of 10
`
`

`
`546
`
`NATURAL PRODUCT REPORTS, 1993
`
`" " ' C O H Hoqo
`
`C02Na
`
`C02Na
`H o q o
`I ONa
`
`F
`
`Fw
`'w
`
`R
`
`(33) R=Pr'
`(34) R = pCeH4F
`(35) R = S@-CeH4F)
`
`i
`(36)
`
`(37)
`
`(38)
`
`F
`
`\ /
`\ / - coNHQ
`
`(39)
`
`F
`
`(40) R = CI
`(41) R=OMe
`
`F
`
`F
`
`F
`
`v
`
`
`
`/ 0
`
`F
`
`t
`F
`
`F 6
`V -
`
`H3c0&wE'
`
`F
`
`.COOCH3
`
`H3C0
`
`vii
`f--
`
`viii
`
`N
`H
`
`vi c--
`
`H3c0*
`
`C02Na
`
`""-C,H
`
`(+)-Enantiomer
`(6)
`Reagents: i, neat, 180 "C; ii, CH,Cl,, DDQ; iii, THF, Red-Al; iv, THF, CH,I; v, THF, LiAlH,, reflux; CH2C12, PCC, rt; vi, THF,
`(EtO),POCH=CHNHCy, NaH, rt; vii, THF, pentan-2,4-dione, NaH, BuLi, 0 "C; THF, BEt,, NaBH,, -65 "C; viii, THF, S-( +>-
`phenylglycinol, 50 "C ; chromatography; ix, EtOH, NaOH, reflux.
`Scheme 7
`
`Published on 01 January 1993. Downloaded by Reprints Desk on 19/05/2015 16:57:02.
`
`View Article Online
`
`NCI Exhibit 2022
`Page 6 of 10
`
`

`
`A1
`NATURAL PRODUCT REPORTS, 1993-A. END0 AND K. HASU3
`
`547
`
`is several times higher than that of lovastatin analogues,
`enhancing the in vivo efficacy of the drug. Clinical trials with
`HR 780 are in progress.
`Compound (32), prepared by the investigators at Hoechst, is
`3-5 times more active than lovastatin in both in vitro and in vivo
`assays and is a promising candidate for development as a
`choles terol-lowering agent. The phenoxy- type inhibitors (3 3),
`(34) and (35) show more pronounced changes in their
`pharmacological
`These compounds are considerably
`stronger in vitro than lovastatin and highly efficacious in
`rabbits, whilst they have only moderate activity in dogs.
`Lovastatin exhibits comparable activity in these animal species.
`The analogues having 1 -N-methyltetrazol-5-y1 attached to
`the C-8 position of 9,9-bis(substituted aryl)-3,5-dihydroxy-6,8-
`nonadienoic acids have been prepared at Bristol-Meyers
`Squibb, of which BMY 22089 (36) is a promising candidate for
`
`d e ~ e l o p m e n t . ' ~ , ~ ~ The open chain salt form of the 4R,6S
`enantiomer of (36) inhibits HMG-CoA reductase with a K,
`value of 4.3 nM, a value comparable to that for lovastatin (2).75
`The hydroxyphosphinyl-containing inhibitor SQ 33 600 (37)
`is about as effective as pravastatin and lovastatin in inhibiting
`intravenous ad-
`de novo cholesterol biosynthesis on
`
`mini~tration.~~ SQ 33 600 also shows oral activity equivalent to
`that of pravastatin and lovastatin and is also effective as a
`hypocholesterolemic agent in rabbits, dogs and monkey^.'^ It
`has been chosen for clinical study in humans.
`A series of substituted pyridines containing a hydroxy-
`phosphinyl functionality have been prepared at Bristol-Meyers
`Squibb, leading to the synthesis of compound (38).77 Compound
`(38) exhibited acute in vivo activity in rats comparable to that
`of lovastatin and pravastatin.
`Investigators at
`the Warner-Lambert Company have
`
`reported a variety of substituted p y r r ~ l e , ~ ~ , ~ ~ quinoline,'O and
`pyrazolea1,82 mevalonolactones. Compound (39) is 5 times
`more potent
`than mevastatin
`
`in ~ i t r o . ~ ~ Two quinoline
`mevalonolactones, (40) and (4 l), are comparable to mevastatin
`both in vitro and in vivo.'O The optically active form of
`compound (42) is 5-10 times more potent than mevastatin both
`in vitro and in vivo.'2 Thus, the hypocholesterolemic activity of
`compound (42) in dogs was significant at doses of 0.1 mg/kg,
`while lovastatin did not show a statistically significant lowering
`at doses lower than 1 mg/kg.82
`Investigators at Rhone-Poulenc Rorer reported a series of
`mevastatin analogues containing a phenylcyclohexane group.
`Of these compounds, RG 12561 (dalvastatin) (43) was slightly
`more potent in inhibitory potency than 10vastatin.~~ RG 12561
`was evaluated in clinical trials for the treatment of hyper-
`cholesterolemia. The analogue (44) is 5 times more potent in
`vitro than l~vastatin.'~
`A research group at SmithKline Beecham reported alterna-
`tives to the 5-hydroxymethylene group of mevastatin analogues
`including a,a-difluoroketonesa4 and phosphinic
`Highly potent HMG-CoA reductase inhibitors have recently
`been discovered via synthetic variation of the substitution
`pattern of functionalized pyridines by investigators at Bayer
`AG. The 5-methoxymethyl derivative BAY W 6228 (45) is the
`most potent compound and has been selected for further
`development (Scheme 7).86 In the in vitro assay BAY W 6228 is
`110 times more potent than lovastatin. The absolute 3R,5S-
`stereochemistry in the 3,5-dihydroxy acid side chain is necessary
`for the inhibitory activity (Table 2). For optimal activity, a
`branched substituent, i.e. isopropyl, cycloakyl or aryl, at C-6 of
`the pyridine ring, in combination with an isopropyl (or
`cyclopropyl) group at C-2 and a p-fluorophenyl substituent at
`C-4, is required. Substitution at position 5 of the pyridine ring
`increased the potency, and the 5-methoxymethyl group of BAY
`W 6228 is essential for its outstanding in vitro and in vivo
`activity (Table 3).
`BAY W 6228 significantly reduces serum cholesterol levels in
`dogs at a daily dose of 30 ,ug/kg, and is thus at least 200 times
`more potent than lovastatin under the same condition^.'^ BAY
`W 6228 is under clinical development.
`
`Table 2 HMG-CoA reductase inhibitory activities of
`substituted pyridines (I). Stereochemistry of the side chain.
`F
`
`R
`OH OH
`&&COONa
`
`?H
`
`?H
`
`4-4-
`COONa
`
`x c
`
`,
`
`, a
`
`OH OH
`&Coma
`
`COON a
`
`Relative potency"
`
`110
`
`0.2
`
`0.2
`
`0.03
`
`40
`
`0.4
`
`a Potency of lovastatin is assigned a value of 1.
`
`lovastatin (2) in inhibiting sterol synthesis in HEP G2 cells. HR
`780 effectively lowers plasma cholesterol levels in normo-
`lipidemic and hyperlipidemic animals (rats, dogs and monkeys).
`Based on equipotent doses, the potency of HR 780 appears to
`be 5-10 times that of lovastatin. The plasma half-life of HR 780
`
`Published on 01 January 1993. Downloaded by Reprints Desk on 19/05/2015 16:57:02.
`
`View Article Online
`
`NCI Exhibit 2022
`Page 7 of 10
`
`

`
`548
`
`NATURAL PRODUCT REPORTS, 1993
`
`Table 3 HMG-CoA reductase inhibitory activities of substituted pyridines (11).
`
`F
`
`COONa
`
`H3C0
`
`Substitution in position 5
`Relative
`R5
`potency"
`H
`0.8
`CH3
`12
`
`CH,OH
`
`CH,OCH,
`
`0"""
`2o Q,, 10
`0"""
`
`20
`20
`
`50
`
`3
`
`Substitution in positions 2 and 6
`Relative
`potencya
`< 0.01
`0.1
`
`R2
`CH3
`CH(CH,),
`
`R6
`CH3
`CH3
`
`CH(CH3)2 CH3
`
`CH(CH,), CH(CH3),
`
`CH(CH3)2
`
`CH(CH3)2
`
`CH(CH,), '0
`
`0.1
`
`50
`
`40
`
`2o
`
`1.6
`
`CH(CH,), '0 0.02
`
`CH(CH3)2
`
`l6
`
`Linker modifications
`
`A-B
`
`CH,-CH,
`
`CH=CH ( E )
`
`c=c
`
`Relative
`potency"
`
`0.7
`
`50
`
`20
`
`COOCH,
`
`30
`
`Substitution in position 4
`Relative
`potency"
`
`R4
`
`
`
`6
`
`Q
`
`Q
`F Q
`
`1
`
`50
`
`a Potency of lovastatin is assigned a value of 1.
`
`5 References
`1 Lipid Research Clinics Program, JAMA, 1988, 251, 351.
`2 A. Endo, M. Kuroda, and Y . Tsujita, J . Antibiot. (Japan), 1976,
`29, 1346.
`3 A. Endo, M. Kuroda, and K. Tanzawa, FEBS Lett., 1976,72,323.
`
`4 A. Endo, Y. Tsujita, M. Kuroda, and K. Tanzawa, Eur. J.
`Biochern., 1977, 77, 31.
`5 I. Kaneko, Y. Hazama-Shimada, and A. Endo, Eur. J . Biochem.,
`1978, 87, 313.
`6 M. S . Brown, J. R. Faust, J. L. Goldstein, I. Kaneko, and A.
`Endo, J. Biol. Chem., 1978, 253, 1121.
`
`Published on 01 January 1993. Downloaded by Reprints Desk on 19/05/2015 16:57:02.
`
`View Article Online
`
`NCI Exhibit 2022
`Page 8 of 10
`
`

`
`NATURAL PRODUCT REPORTS, 1993-A. END0 AND K. HASUMI
`
`549
`
`7 0. Doi and A. Endo, J. Med. Sci. Biol. (Japan), 1978, 31, 225.
`8 K. Tanzawa and A. Endo, Eur. J. Biochem., 1979,98, 195.
`9 A. Yamamoto, H. Sudo, and A. Endo, Atherosclerosis, 1980, 35,
`259.
`10 A. W. Alberts, J. Chen, G. Curon, V. Hunt, J. Huff, C. Hoffman,
`J. Rothrock, M. Lopez, H. Joshua, E. Harris, A. Patchett, R.
`Monaghan, S. Currie, E. Stapley, G. Albers-Schonberg, 0.
`Hensens, J. Hirshfield, K. Hoogsteen, J. Liesch, and J. Springer,
`Proc. Natl. Acad. Sci. USA, 1980, 77, 3957.
`11 A. Endo, J. Antibiot. (Japan), 1979, 32, 852.
`12 A. Endo, J. Antibiot. (Japan), 1980, 33, 334.
`13 W. F. Hoffman, A. W. Alberts, P. S. Anderson, J. S. Chen, R. L.
`Smith, and A. K. Willard, J. Med. Chem., 1986, 29, 849.
`14 Y. Tsujita, M. Kuroda, Y. Shimada, K. Tanzawa, M. Arai, I.
`Kaneko, M. Tanaka, H. Masuda, C. Tarumi, Y. Watanabe, and
`S. Fujii, Biochim. Biophys. Acta, 1986, 877, 50.
`15 S. M. Grundy, N . Engl. J. Med. 1988, 319, 24.
`16 D. B. Hunninghake, Curr. Opin. Lipidol., 1992, 3, 22.
`17 J. L. Goldstein and M. S. Brown, J. Lipid. Res., 1984, 25, 1450.
`18 D. Veloso, W. W. Cleland, and J. W. Porter, Biochemistry, 1981,
`20, 887.
`19 A. S . Beedle, K. A. Munday, and D. C. Wilton, FEBS Lett., 1972,
`28, 13.
`20 T. C. Linn, J. Biol. Chem., 1967, 242, 984.
`21 R. E. Dugan and J. W. Porter, J. Biol. Chem., 1971, 246, 5361.
`22 A. S. Beedle, K. A. Munday, and D. C. Wilton, Eur. J. Biochem.,
`1972, 28, 151.
`23 P. Blattmann and J. Retey, J. Chem. Soc., Dalton Trans., 1970,
`1394.
`24 P. Blattmann and J. Retey, Hoppe-Seyler’s Z . Physiol. Chem.,
`1971, 352, 369.
`25 A. Endo, J . Med. Chem., 1985, 28, 401.
`26 R. H. Abeles and H. Nakamura, Biochemistry, 1985, 24, 1364.
`27 W. E. Brown and V. W. Rodwell, in ‘Dehydrogenases Requiring
`Nicotinamide Coenzymes’, ed. J. Jeffery, Birkhauser Verlag,
`Basel, 1980, p. 232.
`28 A. Endo, Y. Tsujita, M. Kuroda, and K. Tanzawa, Biochim.
`Biophys. Acta, 1979, 575, 266.
`29 Y. Tsujita, M. Kuroda, K. Tanzawa, N. Kitano, and A. Endo,
`Atherosclerosis, 1979, 32, 307.
`30 P. T. Kovanen, D. W. Bilheimer, J. L. Goldstein, J. J. Jaramillo,
`and M. S. Brown, Proc. Natl. Acad. Sci. USA, 1981, 78, 1194.
`31 M. Kuroda, Y. Tsujita, K. Tanzawa, and A. Endo, Lipids, 1979,
`14, 585.
`32 Y. Watanabe, T. Ito, M. Saeki, M. Kuroda, K. Tanzawa, M.
`Mochizuki, Y. Tsujita, and M. Arai, Atherosclerosis, 1981,38,27.
`33 P. A. Kroon, K. M. Hand, J. W. Huff, and A. W. Alberts,
`Atherosclerosis, 1982, 44, 41.
`34 A. Endo, J. Lipid Res., 1992, 33, 1569.
`35 M. S. Brown and J. L. Goldstein, Proc. Natl. Acad. Sci. USA,
`1979, 76, 3330.
`36 M. S. Brown and J. L. Goldstein, Science, 1986, 232, 34.
`37 M. S. Brown and J. L. Goldstein, N . Engl. J . Med., 1981,305,515.
`38 A. G. Brown, T. C. Smale, T. J. King, R. Hasenkamp, and R. H.
`Thompson, J. Chem. SOC., Perkin Trans. I , 1976, 1165.
`39 Y. K. T. Lam, V. P. Gullo, R. T. Goegelman, D. Jorn, L. Huang,
`C. De Riso, R. L. Monagham, and I. Putter, J. Antibiot. (Japan),
`1981, 34, 614.
`40 A. Endo, K. Hasumi, and S. Negishi, J. Antibiot. (Japan), 1985,
`38, 420.
`41 A. Endo, K. Hasumi, T. Nakamura, M. Kunishima, and M.
`Masuda, J. Antibiot. (Japan), 1985, 38, 321.
`42 T. Nakamura, D. Komagata, S. Murakawa, K. Sakai, and A.
`Endo, J. Antibiot. (Japan), 1990, 43, 1597.
`43 G. A. Schonberg, H. Joshua, M. B. Lopez, 0. D. Hensens, J. P.
`Springer, J. Chen, S. Osterove, C. H. Hoffman, A. W. Alberts,
`and A. A. Patchett, J. Antibiot. (Japan), 1981, 34, 507.
`44 T. Rosen and C. H. Heathcock, Tetrahedron, 1986, 42, 4909.
`45 J. K. Chan, R. N. Moor, T. T. Nakashima, and J. C. Vederas,
`J. Am. Chem. SOC., 1983, 105, 3334.
`46 R. N. Moor, G. Bigam, J. K. Chan, et al., J. Am. Chem. SOC.,
`1985, 107, 3694.
`47 A. Endo, Y. Negishi, T. Iwashita, K. Mizukawa, and M. Hirama,
`J. Antibiot. (Japan), 1985, 38, 444.
`48 D. Komagata, H. Shimada, S. Murakawa, and A. Endo, J.
`Antibiot. (Japan), 1989, 42, 407.
`49 K. Kimura, D. Komagata, S. Murakawa, and A. Endo. J.
`Antibiot. (Japan), 1990, 43, 780.
`50 D. Komagata, H. Yamashita, and A. Endo, J. Antibiot. (Japan),
`1986, 39, 1574.
`
`51 S. Murakawa, T. Nakamura, D. Komagata, E. Sunagawa, and A.
`Endo, Agric. Biol. Chem., 1987, 51, 1879.
`52 D. Askin, T. R. Verhoeven, T. M.-H. Liu, and I. Shinkai, J. Org.
`Chem., 1991, 56, 4929.
`53 T.-J. Lee, W. J. Hotz, R. L. Smith, A. W. Alberts, and J. L.
`Gilfillan, J. Med. Chem., 1991, 34, 2474.
`54 M. E. Duggan, A. W. Alberts, R. Bostedor, Y.-S. Chao, J. I.
`Gemershausen, J. L. Gilfillan, W. Halczenko, G. D. Hartman, V.
`Hunt, J. S. Imagire, M. S. Schwartz, R. L. Smith, and R. J.
`Stubbs, J. Med. Chem., 1991, 34, 2489.
`55 D. L. J. Clive, K. S. K. Murthy, R. George, and M. J. Poznansky,
`J. Chem. SOC., Perkin Trans. I , 1990, 2099.
`56 N. Serizawa, K. Nakagawa, K. Hamano, Y. Tsujita, A. Terahard,
`and H. Kuwano, J. Antibiot. (Japan), 1983, 36, 604.
`57 N. Serizawa, S. Serizawa, K. Nakagawa, K. Furuya, T. Okazaki,
`and A. Terahara, J. Antibiot. (Japan), 1983, 36, 887.
`58 T. Matsuoka, S. Miyakoshi, K. Tanzawa, K. Nakahara, M.
`Hosobuchi, and N. Serizawa, Eur. J. Biochem., 1989, 184, 707.
`59 A. Endo, H. Yamashita, H. Naoki, T. Iwashita, and Y. Mizukawa,
`J. Antibiot. (Japan), 1985, 38, 328.
`60 H. Joshua, M. S. Schwartz, and K. E. Wilson, J. Antibiot. (Japan),
`1991, 44, 366.
`61 G. E. Stokker, W. F. Hoffman, A. W. Alberts, E. J. Cragoe, Jr.,
`A. A. Deana, J. L. Gilfillan, J. W. Huff, F. C. Novello, J. D.
`Prugh, R. L. Smith, and A. K. Willard, J. Med. Chem., 1985, 28,
`347.
`62 W. F. Hoffman, A. W. Alberts, E. J. Cragoe, Jr., A. A. Deana,
`B. E. Evans, J. L. Gilfillan, N. P. Gould, J. W. Huff, F. C.
`Novello, J. D. Prugh, K. E. Rittle, R. L. Smith, G. E. Stokker,
`and A. K. Willard, J. Med. Chem., 1986, 29, 159.
`63 G. E. Stokker, A. W. Alberts, P. S. Anderson, E. J. Cragoe, Jr.,
`A. A. Deana, J. L. Gilfillan, J. Hirshfield, W. J. Holtz, W. F.
`Hoffman, J. W. Huff, T. J. Lee, F. C. Novello, J. D. Prugh, C. S.
`Rooney, R. L. Smith, and A. K. Willard, J. Med. Chem., 1986,29,
`170.
`64 G. E. Stokker, A. W. Alberts, J. L. Gilfillan, J. W. Huff, and R. L.
`Smith, J. Med. Chem., 1986, 29, 852.
`65 J. D. Prugh, A. W. Alberts, A. A. Deana, J. L. Gilfillan, J. W.
`Huff, R. L. Smith, and M. Wiggins, J. Med. Chem., 1990,33, 758.
`66 F. G. Kathawala, in ‘Trends in Medicinal Chemistry ’88’, ed. H.
`van der Goot, G. Domany, L. Pallos, and H. Timmerman,
`Elsevier, Amsterdam, 1989, p. 709.
`67 J. Yuan, M. Y. Tsai, J. Hegland, and D. B. Hunninghake,
`Atherosclerosis, 1991, 87, 147.
`68 E. Baader, W. Bartmann, G. Beck, A. Bergmann, H. Jendralla, K.
`Kesseler, G. Wess, W. Schubert, E. Granzer, B. v. Kerekjarto, and
`R. Krause, Tetrahedron Lett., 1988, 29, 929.
`69 G. Beck, K. Kesseler, E. Baader, W. Bartmann, A. Bergmann, E.
`Granzer, H. Jendralla, B. v. Kerekjarto, R. Krause, E. Paulus, W.
`Schubert, and G. Wess, J. Med. Chem., 1990, 33, 52.
`70 G. Wess, K. Kesseler, E. Baader, W. Bartmann, G. Beck, A.
`Bergmann, H. Jendralla, K. Bock, G. Holzstein, H. Kleine, M.
`Schnierer, Tetrahedron Lett., 1990, 31, 2545.
`71 H. Jendralla, E. Baader, W. Bartmann, G. Beck, A. Bergmann, E.
`Granzer, B. v. Kerekjarto, K. Kesseler, R. Krause, W. Schubert,
`and G. Wess, J. Med. Chem., 1990, 33, 61.
`72 H. Jendralla, E. Granzer, B. v. Kerekjarto, R. Krause, U. Schacht,
`E. Baader, W. Bartmann, G. Beck, A. Bergmann, K. Kesseler, G.
`Wess, S. Granata, J. Herchen, H. Kleine, S. Schussler, and K.
`Wanger, J. Med. Chem., 1991, 34, 2962.
`73 N. Balasubramanian, P. J. Brown, J. D. Catt, W. T. Han, R. A.
`Parker, S. Y. Sit

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket