throbber
HMG-CoA Reductase Inhibitors: An Exciting
`Development in the Treatment of
`Hyperlipoproteinemia
`
`F. G. Kathawala
`Preclinical Research Department, Sandoz Research Institute, Route 10, East Hanover, New Jersey 07936
`
`I. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`II. Design Aspect for HMG-CoA Reductase Inhibitors at Sandoz Research Institute
`Leading to Fluvastatin (XU 62-320) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`III. General Chemistry Approach. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`A. Synthesis of Synthon 1 and 2, Fig. 6 (Scheme 1 and Scheme 2) . . . . . . . . . . . . . . .
`B. Choice of R and Synthesis of Intermediates 3, Fig. 6, and 4, Fig. 7. . . . . . . . . . . .
`C. Synthesis of Indole Intermediates. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`D. Synthesis of Indene Intermediates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`E. Synthesis of Naphthalene Intermediates....................................
`F. Synthesis of Imidazole Intermediates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`G. Synthesis of Pyrazole Derivatives. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`H. Synthesis of HMG-CoA Reductase Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`IV. Biological Results and Discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`A. Results in in vitro HMG-CoA Reductase Microsomal Assay and in in vivo
`Cholesterol Biosynthesis Assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`B. SAR of Fluvastatin (XU 62-320) Analog. . . . . . . . . .. . . . .. . . . .. . . . . .. . . . . . . . . . .
`C. SAR of Indene Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`D. SAR of Naphthalene Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`E. SAR of Pyrazole Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`F. SAR of Imidazole Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`V. Effects of Fluvastatin (XU 62-320) on Plasma Lipoprotein Levels . . . . . . . . . . . . . . . . .
`VI. Toxicological, Drug Metabolism and Pharmacokinetic Studies of Fluvastatin
`(XU 62-320).................................................................
`VII. Human Studies with Fluvastatin (XU 62-320)...................................
`VIII. Overview of Published Literature on HMG-CoA Reductase Inhibitors . . . . . . . . . . . .
`IX. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`References.......................................................................
`
`121
`
`122
`124
`125
`126
`128
`128
`128
`129
`129
`129
`133
`
`133
`133
`137
`137
`138
`138
`139
`
`140
`140
`140
`143
`145
`
`I. INTRODUCTION
`
`Coronary heart disease (CHO) continues to be one of the major health
`problems in all the developed countries of the world. A considerable body
`of clinical and epidemiological data has emerged over the years linking ele(cid:173)
`vated blood levels of total cholesterol, 1ow Qensity 1ipoprotein ~holesterol
`(LDL-C), and Yery 1ow Density 1ipoprotein ~holesterol (VLDL-C) as im(cid:173)
`portant risk factors for the development of coronary heart disease. 1
`For the treatment of elevated LDL-C and VLDL-C, a judicious diet, low in
`cholesterol and fat with saturated fatty acids replaced by polyunsaturated
`fatty acids, is the recommended choice. However, for patients nonresponsive
`
`Medicinal Research Reviews, Vol. 11, No. 2, 121-146 (1991)
`© 1991 John Wiley & Sons, Inc.
`
`CCC 0198-63251911020121-26$04.00
`
`NCI Exhibit 2027
`Page 1 of 26
`
`

`

`122
`
`KATHA WALA
`
`to dietary intervention, the development of effective and safe therapeutic
`agents for the treatment of hyperlipoproteinemia remains an important need.
`This need has gained considerable support as a result of two important events:
`(1) the results of the Lipid Research Clinic's Coronary Primary Prevention
`Trial (LRC-CPPT), a multicenter, randomized, double-blind study involving
`3806 asymptomatic middle-aged men in the United States with type II hy(cid:173)
`perlipoproteinemia, that demonstrated that a statistically significant reduction
`of 19% in the rate of fatal plus nonfatal coronary heart disease was associated
`with a 9% decrease in blood cholesterol levels, 2 and (2) the recommendation
`to treat individuals with blood cholesterol above the 75th percentile, which
`emerged from the consensus panel of the December, 1984 NIH Consensus
`Development Conference on the lowering of blood cholesterol to prevent
`coronary heart disease. 3
`In recent years, to achieve this goal of finding effective and safe therapeutic
`agents to lower LDL-cholesterol, great interest has focused on potent inhib(cid:173)
`itors of the enzyme [3-Hydroxy-[3-Methyl-Glutaryl-CoA reductase (HMG-CoA
`reductase, EC 1.1.1.34), which controls a key step in the endogenous synthesis
`of cholesterol. Several studies, both in animals and humans, have been re(cid:173)
`ported with HMG-CoA reductase inhibitors: compactin (Mevastatin), CS-514
`(Pravastatin, Mevalotin®, Pravachol®), mevinolin (Lovastatin, Mevacor®) and
`Synvinolin (Simvastatin, Zocor®), 4 which are structurally very closely related
`to one another. In order to assess fully the potential of HMG-CoA reductase
`inhibitors as an effective therapeutic intervention for the treatment of hyper(cid:173)
`lipoproteinemia, it is thus desirable to study in humans a variety of these
`inhibitors derived from different structural prototypes which can be distin(cid:173)
`guished in their overall biological profile from one another. This conceptual
`framework formed the basis for initiating efforts at the Sandoz Research In(cid:173)
`stitute to develop and study a variety of HMG-CoA reductase inhibitors with
`chemical structures different in several respects from compactin, pravastatin
`(a hydroxy analog of compactin), lovastatin (a methyl analog of compactin),
`and simvastatin (a dimethyl analog of compactin), and has led to fluvastatin
`(XU 62-320), the first totally synthetic HMG-CoA reductase inhibitor currently
`in Phase III human clinical trials (Fig. 1).
`
`II. DESIGN ASPECT FOR HMG-CoA REDUCTASE INHIBiTORS AT
`SANDOZ RESEARCH INSTITUTE LEADING TO FLUVASTATIN
`(XU 62-320)
`
`Investigations by Akira Endo with compactin4 have to be largely credited
`for the resurgence of the research on cholesterol biosynthesis and the renewed
`interest in HMG-CoA reductase inhibitors, a field now almost three decades
`
`F. G. Kathawala obtained his M.Sc. from the University of Bombay, India, and his Ph.D.
`in 1961 from Technische Hochschule Braunschweig, West Germany (Prof. H. H. Inlwffen), in
`Synthetic Organic Chemistry. After a few years of postdoctoral work at Harvard (Prof. R. B.
`Woodward), Wisconsin (Prof. H. Muxfeldt), and Gottingen (Prof. F. Cramer), he joined Sandoz
`in East Hanover, New Jersey, as a Senior Scientist, in 1969. Currently, he is the Director of
`Medicinal Chemistry in the area of Lipoprotein Metabolism/Atherosclerosis. His research in(cid:173)
`terests in Medicinal Chemistn; are focused towards the discoven; of agents affecting lipoprotein
`metabolism/atherosclerosis.
`
`NCI Exhibit 2027
`Page 2 of 26
`
`

`

`HMG-CoA REDUCT ASE INHIBITORS
`
`123
`
`II
`
`Mevastatin ( Compactin )
`
`Lovastatin ( Mcvinolin )
`HO ycooNa
`.. '>r Oil
`
`~g II, Oil
`
`Simvastatin ( Synvinolin )
`F
`
`Oil OH O
`
`Pravastatin ( Eptastatin )
`
`Fluvastatin (XU 62-320)
`
`Figure 1
`
`old. While all intensive studies hitherto conducted have been with closely
`related metabolites, such as compactin, mevinolin, and CS-514 (pravastatin),
`derived from fungal broths, efforts at the Sandoz Research Institute towards
`the development of new HMG-CoA reductase inhibitors have been based on
`synthesis, guided by the following assumptions:
`(a) There are two regions at the active site of the enzyme: one with high
`specific recognition of a 5-carbon unit (C-1 to C-5 as shown below) of the ~­
`OH-~-Methyl-glutaryl portion, and the other of CoA moiety present in HMG(cid:173)
`CoA
`(Fig. 2).
`
`2
`4
`CoAS~Oll
`~~
`~;
`
`/\OH
`
`HMG-CoA
`
`Figure 2
`(b) Compactin (R = H, Fig. 3), a known inhibitor of the enzyme, may be
`regarded as a transition state analog, when in the open dihydroxy acid form.
`
`6
`4
`2
`7~011
`,,H H OH H OH o
`CH3
`
`Figure 3
`
`Compactln ( R= II )
`
`NCI Exhibit 2027
`Page 3 of 26
`
`

`

`124
`
`KATHA WALA
`
`The 5-carbon unit of the side chain present in compactin (Fig. 3) probably
`occupies the same region as the 5-carbon unit in HMG-CoA (Fig. 2); the bicyclic
`A-B-ring system, with its substituents in compactin (Fig. 3), possibly sits in the
`same region or very close to the same region the CoA portion of the substrate
`HMG-CoA occupies at the active site of the enzyme. However, it is difficult to
`see any similarity in structure between the bicyclic-ring system of compactin
`and CoA, when one examines the structure of CoA shown in Fig. 4.
`
`Figure 4
`
`In light of (a) and (b) above, one hoped that it might possible to prepare
`interesting synthetic inhibitors of HMG-CoA reductase with a very general
`structure as shown in Fig. 5, with the 5-carbon unit (C-1 to C-5) preferably
`possessing the absolute configurations of C-3-0H and C-5-0H as present in
`compactin.
`Choice of R and R1 in Fig. 5 has depended on:
`
`R
`
`7
`
`6
`
`5 4
`
`2
`
`3
`
`I OH
`
`~ R1
`
`0
`
`Figure 5
`
`Rl=H,CH3
`
`(a) Consideration of the elements of structure of CoA.
`(b) Considerations of the overall shape and assumptions of the importance
`of substituents on Ring A-B of compactin (Fig. 3), first with molecular models
`and later with computer modelling.
`(c) Exploiting the knowledge gained in structure activity relationships with
`our own Sandoz Research Institute compounds or being reported in literature
`by outside investigators.
`Efforts with the above considerations in mind have led to the development
`of a variety of novel HMG-CoA reductase inhibitors. Synthesis and Structure
`Activity Relationships (SAR) of some of these novel inhibitors are discussed
`below with emphasis on the Phase III candidate, fluvastatin (XU 62-320):
`[R*, S*-(E)]-( ± )-Sodium-3,5-dihydroxy-7-[3-( 4-fluorophenyl)-1-(1-methyl(cid:173)
`ethyl-lH-indol-2-yl]-hept-6-enoate (Fig. 1), a mevalonic acid analog more po(cid:173)
`tent than compactin and lovastatin.
`
`III. GENERAL CHEMISTRY APPROACH
`
`Guided by the conviction that the C-3, C-5 dihydroxy acid fragment was
`the key pharmacophore necessary for the inhibition of HMG-CoA reductase,
`
`NCI Exhibit 2027
`Page 4 of 26
`
`

`

`HMG-CoA REDUCTASE INHIBITORS
`
`125
`
`our synthetic approach towards the synthesis of compounds of generic struc(cid:173)
`ture (Fig. 5) involved:
`(a) A convergent synthesis coupling chiral Synthon 1 or racemic or chiral
`(3R, 5S) C-3, C-5-dihydroxy ester Synthon 2 with a variety of aryl or alkyl
`fragments 3 (Fig. 6), or
`(b) A linear synthesis of the C-3, C-5 dihydroxy acid derivatives wherein
`the aldehyde 4 is reacted with acetoacetate 5 (Fig. 7) to provide a hydroxyketo
`ester intermediate, which, with subsequent steps, gives the desired final
`products of Fig. 5.
`
`R~X
`X = pt(Ph) 3z(cid:173)
`y = P=O(OR 3J2
`
`Rt = Si(t-Bu)Phz
`
`2
`
`3
`Rz "R3 = AlkJI, z· =Cl, Br
`
`R 7 6 5 0
`~
`H
`
`4
`
`0
`0
`4 u 2 u
`~JOR2
`
`5
`
`Figure 6
`
`Figure 7
`
`A. Synthesis of Synthon 1 and 2, Fig. 6 (Scheme 1 and Scheme 2)
`
`Synthon 1 has been synthesized starting from 0-glucose via the key lithium
`aluminum hydride reductive opening of the epoxide as depicted in Scheme
`1. 5 The desired axial alcohol could be separated from the equatorial isomer
`by preparation of the silyl derivatives. The protected axial alcohol on PCC
`oxidation gave the desired lactol aldehyde.
`
`R = Sl(t-llu)Phz
`
`SCHEME 1
`
`NCI Exhibit 2027
`Page 5 of 26
`
`

`

`126
`
`KATHA WALA
`
`Ph3CO~OCH3
`
`cmo
`lt -Butylacetate
`
`LDA
`
`1 . MeOH I AcCJ
`HO~OH
`2. DBMS I NaBH4
`
`cm 0
`
`0
`
`1. Et3 B /Nalll14
`
`0
`H~Ot-Bu
`2. TllDPSCI
`IMIDAZOLE
`OR OR 0
`
`R = S I ( I • ll u ) Ph 2
`
`3 . TFA I HzO I CHzClz
`
`4 • PCC I CllzCl2
`
`SCHEME 2
`
`OH
`
`{1
`
`HO
`
`OH
`
`112/Ra:"I
`
`EtOll
`
`Oft
`
`OR
`
`H000H
`
`l\llDAZOLE
`
`TBDPSCl/D\IF ROD OH
`J:r
`
`OR
`
`OR
`
`~COOCH3
`
`CHO
`
`RO
`
`t.TFA/\-leOH
`
`2.PCCICll2Cl2
`
`RO
`
`iPCC
`c11 2c1 2
`
`\ICPBA
`
`:"allC03
`Cll2Clz
`
`OR
`
`.)io
`
`(R = TllDPS)
`
`SCHEME 3
`
`Synthesis of chiral Synthon 2 has been accomplished starting from S-malic
`acid in excellent yields via an eight-step reaction as illustrated in Scheme 2. 6
`On the other hand, an efficient route was developed for the preparation
`of racemic Synthon 2 starting from 1,3,5-trihydydroxy benzene through a
`five-step reaction sequence shown in Scheme 3. 7
`
`B. Choice of Rand Synthesis of Intermediates 3, Fig. 6, and 4, Fig. 7
`
`Our initial efforts at the synthesis, and the biological results of C-3, C-5-
`dihydroxy acid derivatives (Fig. 5) wherein choice of R was based on elements
`of substructures of coenzyme A (Fig. 4) or the decalin ring structure of com(cid:173)
`pactin (Fig. 3) were not promising. 8 This led us to question the importance
`and the necessity of the complex stereochemistry and the substituents present
`in the decalin ring of compactin and turn our attention towards the prepa(cid:173)
`ration of C-3, C-5-dihydroxy acid derivatives (Fig. 5) wherein R was a naph(cid:173)
`thalene ring. During these ongoing efforts, we were being encouraged and
`helped by two important publications9 describing mevalonolactone deriva(cid:173)
`tives of the general structure 6 and 7 as inhibitors of HMG-CoA reductase
`(Fig. 8).
`Further exploration of R in Fig. 5 led to the first interesting indolyl derivative
`(Fig. 9) comparable to compactin in its inhibitory activity against HMG-CoA
`reductase. 10<a>
`An extensive and rapid analog program allowed the choice of XU 62-320
`
`NCI Exhibit 2027
`Page 6 of 26
`
`

`

`HMG-CoA REDUCT ASE INHIBITORS
`
`127
`
`HO
`.... ,.
`H
`
`HO~
`
`6
`
`7
`
`F
`
`01-1011 0
`
`o(cid:173)
`N a+
`
`X= CH=CH , (Cll2) m
`
`01-1 OH 0
`Z= A~o~a+
`
`Either one or two of the
`atoms a,b,c, = Heteroatom
`
`Figure 8
`
`Figure 9
`
`Figure 10
`
`(Fig. 1) as a candidate for extensive biological testing. Currently, fluvastatin
`(XU 62-320) is in clinical Phase III trials.
`With the discovery of XU 62-320, the stage was set for a large number of
`variations of R in Fig. 5. Extensive work at the Sandoz Research Institute has
`led to many novel HMG-CoA reductase inhibitors, some of which are dis(cid:173)
`cussed in this paper as shown in Fig. 10, 10 and Figs. 12-14.21
`23
`-
`Synthesis of the many interesting fragments 3 (Fig. 6) and 4 (Fig. 7) needed
`for synthesis of final HMG-CoA-R inhibitors are described in Schemes 4-12
`below. 10 Since the appearance of Merck & Co., Inc. and Sandoz patents and
`publications, s, 9,IO(a) extensive efforts have followed in many laboratories
`worldwide with semi-synthetic and totally synthetic HMG-CoA reductase
`inhibitors. A brief overview of these reported activities is presented in Section
`VIII. It is no wonder that in such a feverish pursuit of finding patentable
`HMG-CoA reductase inhibitors, review of patent and published literature
`presents overlapping activities in the laboratories of competing pharmaceut(cid:173)
`ical research companies.
`
`NCI Exhibit 2027
`Page 7 of 26
`
`

`

`128
`
`KATHA WALA
`
`R1 0 NH
`
`I
`Rz
`
`+
`
`EtOH
`
`CHO
`
`1. Mc2N r
`
`2. NaOH
`
`SCHEME 4
`
`R,-rfrP
`R 1
`.-.... Rnf:'.~
`~~~~~ ~CHO
`:-iallll,i l R2
`
`N
`I
`Rz
`
`D:\1F
`
`I
`
`~R3-v
`J .SOCl2
`....._ ____ Ri-f:{_J(~r·•
`N
`c11 2011
`I
`R2
`
`SCHEMES
`
`C. Synthesis of Indole Intermediates
`
`Scheme 4 describes the preparation of cx,[3-unsaturated aldehydes readily
`obtained from a variety of 3-phenyl substituted indoles using dimethylamino(cid:173)
`acrolein and phosphorous oxychloride, while the triphenyl phosphonium
`salts of indolyl derivatives are prepared via the 2-formyl and 2-hydroxymethyl
`indoles using standard procedures (Scheme 5). 10<al
`
`D. Synthesis of Indene Intermediates
`
`A variety of indenyl-cx, [3-unsaturated aldehydes and phosphonates have
`been synthesized via a six-step reaction sequence as depicted in Schemes 6
`and 7. The synthesis of these derivatives involves the preparation of the
`desired indenes from the respective indanones followed by either formylation
`at C-2 and subsequent alkylations at C-1 or vice versa, and then processing
`the formyl group through standard reaction sequences to the desired inter(cid:173)
`mediates.10<b) .
`
`E. Synthesis of Naphthalene Intermediates
`
`For the preparation of naphthalene derivatives, a novel photochemical route 11
`was exploited to give the key hydroxy aldehyde, which on dehydration pro(cid:173)
`vides the ene aldehyde. Dehydrogenation of the ene aldehyde and chain
`
`NCI Exhibit 2027
`Page 8 of 26
`
`

`

`HMG-CoA REDUCT ASE INHIBITORS
`
`129
`
`l. R2 MgX, llOAc
`
`2. Ph'.'\ (Me )CHO
`
`POCl3
`
`l. DIBAL
`
`R3
`
`R2
`
`R1
`
`R1
`
`co 2 Me
`
`SCHEME 6
`
`l'\all
`
`-----.-
`
`l NaBH4
`
`2 SOCl2
`
`3 P(OMe)3
`
`SCHEME 7
`
`y oco
`
`~CHO
`
`88 % by G.C.
`
`l Ph '.'\(:\-le)CllO
`
`POCl3
`
`II
`
`Horisll l C6 H5 CH3
`
`Reflux
`
`0
`
`II
`
`l . DDQ, Toluene
`
`2. n·llu3SnCJl=CHOEt
`n -lluLI, TllF, -60°
`
`3. p-TsOH ,THF,1120
`
`16 hrs, r. t.
`SCHEMES
`
`H
`
`NCI Exhibit 2027
`Page 9 of 26
`
`

`

`130
`
`KATHA WALA
`
`extension of the formyl group then leads to the desired cx,(3-unsaturated
`aldehydes10(c) (Scheme 8).
`
`F. Synthesis of Imidazole Intermediates
`Highly substituted imidazole derivatives with the desired functional group
`at the desired C- or hetero- atom are not well described in the literature.
`Synthesis of the required imidazole intermediates was best accomplished
`starting from the respective glycine derivatives as shown in Scheme 9. The
`key step in the synthetic pathway involves oxidation of the methyl group
`with potassium persulfate to give the 5-formyl imidazole derivatives, which
`through standard reaction sequences give the needed a,(3-unsaturated alde(cid:173)
`hydes or the phosphonates. ID(d)
`
`J.R2COCI
`2. Ac20 I AcOH, Pyridine, .1
`
`3. R3NH2, pTsOH, MgS04, Toluene, .1
`
`4. PCl5 I CHCl3
`
`RJ, R2, R3 =ALKYL OR SUBSTITUTED ARYL
`
`SCHEME9
`
`G. Synthesis of Pyrazole Derivatives
`
`A number of pyrazole intermediates have been prepared via procedures
`dependent on whether one needs the 1,5 (Scheme 10), the 1,3 (Scheme 11),
`or the 3,4 (Scheme 12) disubstituted pyrazole intermediates. 2,3-disubstituted
`pyrazole derivatives are obtained through the reaction of the appropriate
`diketoesters with aryl-hydrazines, requiring separation from the concomitant
`formation of the corresponding 1,3 isomer (Scheme 10). 10
`(e)
`1,3-disubstituted pyrazoles can be best synthesized from the imide chloride
`on reaction with the acetoacetate derivatives (Scheme 11), while the ring
`closure of arylhydrazones give the desired 3,4 diaryl pyrazole intermediates
`(Scheme 12).
`
`H. Synthesis of HMG-CoA-R Inhibitors
`
`All of the intermediates of the many different prototypes described above
`in Schemes 4-12 could be converted to the final HMG-CoA reductase inhib-
`
`NCI Exhibit 2027
`Page 10 of 26
`
`

`

`HMG-CoA REDUCTASE INHIBITORS
`
`131
`
`F
`
`F
`
`PhNHNH2
`
`I. LIAIH4 I
`2.PCC t
`
`F
`
`F
`
`CHO
`
`J. Ph3P=CHCOOEt
`
`2.LIAIH4
`J.Mno2
`SCHEME JO
`
`F 2
`
`0
`
`HN
`I
`A-NH
`
`v
`
`PCl5 ...
`
`F
`
`2 _1. __ '_"'_'"-~_._n_12_c_oo--1E~1
`
`N,, Cl
`
`NaOEt
`
`F
`
`COO Et
`
`er~H 2.H+
`
`SCHEME 11
`
`F
`
`~6
`~ 0
`
`J. l-Pr-NHNH2 I EtOH I AcOH (80°)
`
`I.NBS
`
`SCHEME 12
`
`N-N
`
`)-
`
`NCI Exhibit 2027
`Page 11 of 26
`
`

`

`132
`
`KATHA WALA
`
`0
`0
`~OR
`
`' CHO Nall/ n-BuLI R1
`
`COOR
`
`l 1 • Et 311 , TllF
`
`2 . NaBll4, -78°
`Et
`
`COOR
`
`COON a
`
`SCHEME 13
`
`itors either using the linear route involving the "dianion chemistry," or the
`coupling of the respective phosphonates or phosphonium salts with the chiral
`Synthons 1 and 2 (Fig. 6) or with the racemic Synthon 3 (Fig. 6).
`1. Linear Route. Synthesis using the linear route is illustrated in Scheme 13
`for the preparation of the indolyl HMG-CoA reductase inhibitors. The key
`step involves the reduction of the hydroxyketoester using trialkylbor(cid:173)
`ane/THF/MeOH with sodium borohydride at - 78° (Ref. 12) to give the mixture
`of desired erythro and threo isomers in the ratio of 95-98:5-2%, respectively.
`In some cases, the boronic esters can be crystallized, which on methanolysis
`and subsequent hydrolysis with sodium hydroxide provide the desired so-.
`dium salts. Nonstereoselective reduction of hydroxyketoester with borane t(cid:173)
`butylamine complex has been used to prepare a mixture of cis and trans
`lactones separable on flash chromatography. 10<aJ
`
`R1~
`
`N
`R2
`
`CH2P+Ph3
`er
`
`1. AcOH /THF I H20
`
`SCHEME 14
`
`Rt= Si( t-Bu )Ph2
`
`NCI Exhibit 2027
`Page 12 of 26
`
`

`

`HMG-CoA REDUCT ASE INHIBITORS
`
`133
`
`LDA ff HF
`
`OHC~Ot-Bu
`
`OR OR 0
`
`~ YOt-Bu
`
`OR OR 0
`
`F~ 1. n- Bu4NF
`
`2. NaOH
`
`ON a
`
`OH
`
`R =SI (t-Bu ) Ph2
`
`SCHEME 15
`
`2. Convergent Route. For illustrative purposes, a convergent route for the
`preparation of chiral indolyl HMG-CoA reductase inhibitors using the silyl
`protected Synthon 1 is depicted in Scheme 14. The crucial step in this reaction
`pathway is the oxidation of lactol with RuC12(PPh3)J/NMM0. 10<rJ
`Scheme 15 shows the use of silyl-protected aldehyde Synthon 2 (derived
`from malic acid) for the synthesis of indenyl HMG-CoA reductase inhibitors. 13
`
`IV. BIOLOGICAL RESULTS AND DISCUSSION
`
`A. Results in in vitro HMG-CoA Reductase Microsomal Assay and in in
`vivo Cholesterol Biosynthesis Assay
`
`All initial studies to assess the inhibitory potency of various compounds
`against HMG-CoA reductase were conducted with rat liver microsomal sus(cid:173)
`pensions, freshly prepared from male Sprague-Dawley rats, using an assay
`for HMG-CoA reductase activity as described in Ref. 14. The potency of each
`compound is expressed as IC50 (in µmoles, the concentration which inhibits
`to the extent of 50% conversion of the substrate HMG-CoA to mevalonate)
`and for structure activity relationship compared either to compactin = 1 or
`to XU 62-320 = 1. Tables I-XII summarize the most salient features of structure
`activity relationships for a few of the varied structural prototypes as HMG(cid:173)
`CoA reductase inhibitors being currently studied at the Sandoz Research
`Institute. In Tables X-XIII, the Relative Potency column is derived from the
`ICso values of each compound vs. compactin in the in vitro rat microsomal
`HMG-CoA reductase assay.
`
`B. SAR of Fluvastatin (XU 62-320) Analogs
`
`Table I compares the in vitro inhibitory activity against HMG-CoA reductase
`of XU 62-320 with compactin and lovastatin and as their corresponding sodium
`salts. XU 62-320 is 146- and 52-fold more active than compactin and Lovastatin,
`respectively. As compared to the respective sodium salts of compactin and
`Lovastatin, XU 62-320 is 22- and 10-fold more potent in inhibiting HMG-CoA
`reductase. It is important to note that current clinical studies are being con(cid:173)
`ducted with XU 62-320, which is a dihydroxy acid sodium salt. In contrast,
`
`NCI Exhibit 2027
`Page 13 of 26
`
`

`

`134
`
`KATHA WALA
`
`Table I
`Comparison of Microsomal HMG-CoA Reductase Inhibitory Activity
`
`Compound
`
`ICso (µM)
`
`Relative Potency•
`
`XU 62-320
`Compactin
`Lovas ta tin
`Na Salt Compactin
`Na Salt Lovastatin
`
`0.0069
`1.011
`0.3S2
`0.154
`0.068
`
`146.1
`1.0
`2.8
`6.S
`14.8
`
`*As compared to Compactin = 1
`
`compactin used in clinical studies and Lovastatin (Mevacor®), now marketed,
`both exist as the lactone forms (Fig. 1).
`Features of the side chain are very important for maximal inhibitory activity
`as shown in Table II. Erythro configuration, as well as the double-bond con(cid:173)
`figuration, are very important [anti-isomer 17-fold less active and dramatic
`loss of activity for one (Z) diene isomer]. The dihydro derivative, as well as
`the ester and the lactone forms, are considerably less active. Maximal inhib(cid:173)
`itory activity resides in the 3R, SS antipode.
`The importance of the features of the side chain described in Table II for
`the indole series holds true as well for all the prototypes to be described later
`and hence, during the discussion of SAR of these prototypes, these aspects
`will not be reemphasized. HMG-CoA, the substrate for the HMG-CoA re(cid:173)
`ductase, has at C-3 a methyl group. It was important to determine if an analog
`of XU 62-320 carrying a methyl group at C-3 would be more potent. Surpris(cid:173)
`ingly, introduction of methyl group at C-3 in either of syn- or anti-configu(cid:173)
`ration was considerably less active (Table III).
`Studies of the effects of the substituents in the 3-phenyl ring of the indole
`moiety are given in Table IV. Either electron-withdrawing or electron-donat(cid:173)
`ing substituents in the 3-phenyl ring tend to decrease the potency, which is
`unaffected by the presence of alkyl groups.
`Electron-donating or electron-withdrawing substituents (not shown in Ta(cid:173)
`ble IV) or bulky alkyl groups at C-5 of the indole moiety led to decrease of
`potency. However, alkyl or alkoxy groups at C-4 and C-6 tend to maintain
`or enhance the potency slightly (Table V).
`
`Table II
`SAR of Variations in the Side Chain
`
`Compound
`
`ICso (µM)
`
`Relative Potency•
`
`F
`
`XU 62-320
`3R, SS
`I ~
`3S, SR
`Na Salt, ANTI
`OH OH 0 •
`Methyl Ester, SYN
`Trans Lactone
`o- CIS(f:) Double Bond
`Dihydro (Reduced
`Na+
`Double Bond)
`
`0.0069
`0.0024
`0.08
`0.12
`O.OS2
`0.029
`0.62
`0.114
`
`1.0
`2.8
`0.086
`O.OS7
`0.13
`0.23
`0.011
`0.06
`
`*As compared to XU 62-320 = 1.
`
`NCI Exhibit 2027
`Page 14 of 26
`
`

`

`HMG-CoA REDUCTASE INHIBITORS
`
`135
`
`Table III
`Comparative Activity of XU with the 3-Methyl Analogs
`
`Compound
`
`ICso (µM)
`
`Relative Potency*
`
`XU 62-320
`R = CH3, SYN
`R = CH3, ANTI
`
`0.0069
`0.14
`0.51
`
`1.0
`0.049
`0.013
`
`*As compared to XU 62-320 = 1
`
`Table IV
`SAR for the Substituents of the 3-Phenyl Ring
`
`R
`
`ICso (µM)
`
`Relative Potency*
`
`o-
`
`4-F
`2-Me
`2-Me, 4-F
`3-Me, 4-F
`3,5-diMe, 4-F
`3,5-diMe
`H
`4-CF3
`4-SCH3
`4-COONa
`
`0.0069
`0.14
`0.004
`0.009
`0.02
`0.005
`0.017
`0.09
`1.152
`>10.0
`
`1
`0.049
`1.7
`0.76
`0.345
`1.38
`0.40
`0.076
`0.006
`
`*As compared to XU 62-320 = 1
`
`Table V
`SAR for the Substituents of the Benzenoid Indole Ring
`
`F
`
`o·
`
`R
`
`ICso (µM)
`
`Relative Potency*
`
`H (62-320)
`4,6-diMe
`4,6-dii-Pr
`5-C6H11
`6-0CH2Ph
`
`0.0069
`0.011
`0.005
`24.0
`0.0026
`
`1.0
`0.62
`1.38
`0.0022
`2.65
`
`*As compared to XU 62-320 = 1
`
`Table VI
`SAR for the Substituents of Indolyl-Nitrogen
`
`F
`
`R
`
`ICso (µM)
`
`Relative Potency*
`
`i-Pr (62-320)
`CH3
`C2Hs
`C6H11
`CH2CH2Ph
`CH2CH(CH3)z
`
`0.0069
`0.62
`0.096
`50
`49.4
`0.245
`
`1.0
`0.011
`0.071
`0.0001
`0.0001
`O.o28
`
`*As compared to XU 62-320 = 1
`
`NCI Exhibit 2027
`Page 15 of 26
`
`

`

`136
`
`KATHA WALA
`
`Table VII
`SAR for Reversing Substituents at 1 and 3 Positions
`
`R1
`
`Ri
`
`ICso
`(µM)
`
`Relative
`Potency•
`
`~ i-Pr (62-320)
`
`:.-..
`
`N\
`
`R1
`
`o·
`Na+
`
`4-FC6H4
`i-Pr
`
`4-FC5H4, ~ 0.0069
`0.0016
`i-Pr,~
`4-FC~4, anti
`0.12
`
`1.0
`4.3
`0.057
`
`*As compared to XU 62-320 = 1
`
`Most sensitive to the activity is the substituent on the nitrogen of the indole
`moiety (Table VI). Optimal activity is provided by the isopropyl group, while
`marked loss in potency results with either bulky alkyl or phenethyl groups.
`Reversing the substituents on N-1 and C-3 of the indole moiety to give
`(Table VII) 3-isopropyl-N-p-fluorophenyl analog of XU 62-320 gives a 4-fold
`increase in potency.
`Most of the substances with a reasonable level of activity against HMG(cid:173)
`CoA reductase in in vitro microsomal assay were studied in vivo for their
`effects on inhibition of sterol biosynthesis. Results are expressed as EDso
`(mg/kg), effective concentration which inhibits to the extent of 50% incor(cid:173)
`poration of C14 acetate into sterols in rats when administered as appropriate
`doses of drug substances as compared to controls receiving vehicle alone.
`Table VIII shows that in vivo XU 62-320 is about 40- and 4.5-fold more potent
`than compactin and Lovastatin, respectively, in inhibiting endogenous cho(cid:173)
`lesterol synthesis in rats. For most substances, although not for all, the relative
`
`Table VIII
`Relative Potency for Inhibition of Cholesterol Biosynthesis
`
`Compound
`
`EDso (mg/kg)
`
`Relative Potency•
`
`XU 62-320
`Compactin
`Lovas ta tin
`(Monacolin)
`
`0.093
`3.5
`0.414
`
`37.6
`1.0
`8.4
`
`*As compared to Compactin = 1
`
`Table IX
`SAR for Cholesterol Biosynthesis Inhibition
`
`F
`
`o-
`Na+
`
`XU 62-320
`
`*As compared to XU 62-320 = 1
`
`Compound
`
`XU 62-320
`3R,5S
`3S, SR
`Na Salt, Anti
`Methyl ester, ~
`Trans Lactone
`Dihydro (Reduced
`Double Bond)
`
`ED so
`(mg/kg)
`
`0.093
`0.056
`>0.5
`1.37
`0.40
`0.33
`1.23
`
`Relative
`Potency•
`
`1.0
`1.66
`
`0.067
`0.23
`0.28
`0.075
`
`NCI Exhibit 2027
`Page 16 of 26
`
`

`

`HMG-CoA REDUCTASE INHIBITORS
`
`137
`
`Table X
`SAR of Indene Derivatives
`
`R1
`
`Relative Potency*
`
`Na+
`o-
`
`OH OH 0
`
`(CH2)4
`(Racemic)
`(CH2)4
`(3R, 55)
`(Clhh
`(CH2)s
`CH2CH3
`CH3
`H,iPr
`
`R1
`
`Phenyl
`3,5-Dimethylphenyl
`iPr
`Cyclohexyl
`
`RJ
`
`4-Me
`6-Me
`7-Me
`6-0Me
`4,6-(0Meh
`
`202
`
`337
`
`38
`1.5t
`<.2
`2
`8
`
`88t
`146
`<0.5
`16.5
`
`114
`181
`24
`130
`60
`
`*As compared to Compactin = 1
`tAs its Ethyl Ester
`
`potency determined in in vitro microsomal assay against HMG-CoA reductase
`parallels the in vivo activity in rats for the inhibition of 14C-acetate into sterols.
`As an example, comparison of Tables II and IX reveals the relative potency
`of several analogs of XU 62-320 when compared in in vitro and in in vivo.
`Thus, as compared to XU 62-320, the anti-isomer is - 17- (Table II) and -
`15-fold (Table IX) less active than XU 62-320 in in vitro and in in vivo assays,
`respectively. Similarly, close parallelism prevails for the ester (less active -
`7.5-fold, in vitro vs. 4.3-fold, in vivo), trans-lactone (less active 4.2-fold, in vitro
`vs. 3.5, in vivo) and the dihydro derivative (less active 16.5-fold, in vitro vs.
`13-fold in vivo).
`
`C. SAR of Indene Derivatives
`
`The structure activity relationships for the indene derivatives can be best
`summarized as follows: Maximal activity is obtained with a spiro cyclopentyl
`group at C-1, again emphasizing the importance of the bulky group in the
`vicinity of the dihydroxy acid side chain. At C-3 the best substituent is 4-F(cid:173)
`phenyl, while the optimal substituent for the benzenoid portion of the indene
`moiety is hydrogen (see Table X).
`
`D. SAR of Naphthalene Derivatives
`
`The most interesting part of the structure activity relationships for this
`group of compounds is the difference observed in the potency of 1-(4-F-
`
`NCI Exhibit 2027
`Page 17 of 26
`
`

`

`138
`
`KATHAWALA
`
`Table XI
`SAR of Napthalene Derivatives
`
`OH OH 0
`
`o-
`
`4-F-Ph
`4-F-Ph
`4-F-Ph
`4-F-Ph
`3,5-diMe-Ph
`Ph
`i-Pr
`i-Pr
`
`H
`CH3
`Et
`i-Pr
`CH3
`CH3
`4-F-Ph
`Ph
`
`Relative
`Potency*
`
`0.10
`8
`19
`22
`56
`2
`337
`144
`
`*As compared to Compactin = 1
`
`phenyl)-3-isopropyl derivative vs. 1-isopropyl-3-(4-F-phenyl) compound (22
`times more potent vs. 337 as compared to compactin) (see Table XI).
`
`E. SAR of Pyrazole Derivatives
`
`Table XII illustrates the structure activity relationships for a few of the many
`pyrazole derivatives prepared. Here, too, the optimal substituents are the 4-
`F-phenyl and isopropyl group adjacent to the dihydroxy acid side chain. The
`dihydro and the 5-keto derivatives are substantially less potent. 1,3-diaryl(cid:173)
`substituted pyrazole derivatives show decreased inhibitory activity (not shown
`in the table) in contrast to the 1,5 and 3,4-diaryl-substituted compounds,
`which tend to have comparable potency.
`
`F. SAR of Imidazole Derivatives
`
`To emphasize the most salient features of the structure activity relationships
`for the imidazole derivatives, only a few of the derivatives prepared are
`tabulated in Table XIII. Optimal activity is obtained with 1,2-diaryl derivatives
`
`Table XII
`SAR of Pyrazole Derivatives
`
`R
`
`Relative Potency*
`
`4-F
`4-F (6,7 Dihydro)
`4-F (5 Keto)
`H
`3,5 Dimethyl
`
`4-F
`
`60
`5.9
`3.5
`5.6
`4.1
`
`30
`
`o-
`
`*As compared to Compactin = 1
`
`NCI Exhibit 2027
`Page 18 of 26
`
`

`

`HMG-CoA REDUCT ASE INHIBITORS
`
`Table XIII
`SAR of lmidazole Derivatives
`
`4-F
`(Racemic)
`4-F
`(3R, SS)
`p-CI
`p-Br
`3,5-Di-Me
`3,5-Di-Cl
`
`i-Pr
`t-Butyl
`cyclohexyl
`2-Thienyl
`1,4-Biphenylyl
`p-Dimethylamino-phenyl
`p-Nitro-phenyl
`
`F
`
`i-Pr
`4-F-Phenyl
`
`13

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