throbber
Current Pharmaceulical Design, 1998, 4. 71 -92
`
`71
`
`Selective Estrogen Receptor Modulators (SERMs)
`
`T. A. Grese‘k and J. A. Dodge
`
`Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, USA
`
`
`
`I7B-estradiol and estrone, have
`Abstract: Naturally occurring estrogens, such as
`traditionally been thought to play a central role in the development and maintenance 01
`the female reproductive system and secondary sexual characteristics. In recent years, their
`beneficial effects on the skeleton,
`the cardiovascular system, and the central nervous
`system, as well as the cancer risks associated with long term exposure have also been
`recognized. The widespread use of “ antiestrogcns " such as tamoxifen for the prevention
`and treatment of breast cancer has revealed that such compounds, while functioning as estrogen antagonists in
`mammary tissue, actually mimic the effects of estrogen in other tissues. The search for more selective agents
`has led to the development of raloxifenc. a Selective Estrogen Receptor Modulator. which functions as an
`estrogen antagonist
`in the breast and uterus and as an estrogen agonist
`in the skeleton and cardiovascular
`system. Recent progress in the development of SERMs is the subject of this review, with an emphasis on
`structure activity relationships and on their effects in non—traditional target tissues.
`
`Introduction
`
`The central role played by endogenous estrogens, such as
`l7B~estradioL l, and estrone, 2,
`in the development and
`maintenance of the female sex organs, mammary glands, and
`ollrcr sexual characteristics has long been recognized [1,2].
`Recently,
`their involvement
`in the gr0wth and function of a
`number of other tissues, such as the skeleton, the cardiovascular
`system, and the central nervous system,
`in both males and
`females has been recognized [3,4l.
`
`The primary site of estrogen biosynrhesis in the adult female
`is the ovary. After the menopause,
`the ovarian production of
`estrogens declines dramatically producing a wide range of
`primary and secondary physiological effects [5,6]. The decline
`in levels of circulating estrogens has also been linked to a
`number of pathological conditions
`including osteoporosis
`[7,8], coronary artery disease [9,10], depression [I 1,12]. and
`Alzheimer‘s disease [12,13]. Estrogen replacement
`therapy
`(ERT) has proven effective in reducing the frequency and severity
`of these pathologies, but
`the increased risk of endometrial
`cancer observed with ERT has necessitated the development of
`therapeutic regimens in which the uterine effects of estrogen are
`opposed by progestin treatment (hormone replacement
`therapy
`or HRT)
`[14,15]. Sideeeffects of progestrn treatment, such as
`resumption of menses, central nervous system disturbances, and
`the possibility of attenuated cardiovascular benefits. have
`unfortunately resulted in decreased patient compliance [16,17].
`
`Table 1.
`
`Classification of Estrogen Receptor Modulators
`
`Furthermore, recent studies which confirnr the increased risk of
`breast and endometrial cancer associated with long term ERT or
`HRT have led to the search for treatment alternatives [18,19|.
`
`importance of estrogen in the development and
`The
`maintenance of the female reproductive system has led to the
`pharmaceutical development of a variety of steroidal and non—
`sterordal compounds which interact with the estrogen receptor
`(ER) as contraceptives and for the treatment of breast cancer,
`uterrne dysfunction, and other reproductive disorders. Several
`reviews of ERemodulators, with a particular emphasis on their
`utility in the treatment of breast cancer, have been recently
`published
`[20,21]. Early synthetic
`estrogens
`such
`as
`diethylstilbestrol (DES), 3, and hexestrol, 4, were once widely
`utilized as estrogen substitutes, but due to concerns similar to
`those encountered with the natural hormones and other side
`
`their utility has diminished. The discovery that
`effects
`compounds such as
`\tlERAZS. 5, antagonize the action of
`estrogen in breast
`tissue led to intensive pharmaceutical
`research, culminating in the development of tamoxifen, 6.
`which has found great utility in the treatment of brezist cancer
`[22]. Early concerns
`that
`the
`longeterm use of
`these
`"antiestrogens" would lead to increased risks of osteoporosis
`and cardiovascular disease have been dispelled by
`the
`paradoxical
`finding that some compounds (i.e.
`tamoxifen and
`raloxifene, 7) actually mimic the effects of estrogen in skeletal
`and cardiovascular trssues. although others tic.
`ICI 182780,
`8b) do not
`['23]. Findings such as
`these have led to a
`
`Classification
`
`Uterine Stimulation
`
`Butte/Cardiovascular
`
`Example
`
`Pure Antiestrogens
`
`Estrogen Agonists
`
`Partial Agonists (Ist Generation SERMs)
`
`2nd Generation SERMS
`
`agonist
`
`agonist
`
`agonrsr‘
`
`17Beestr'adiol, 1
`
`tamoxifen. 6
`
`r'aloxrfcne, 7
`
`antagonist/neutral
`
`1C1 182780.81)
`
`Hill-6125998 $15 f)()+.00
`
`it? 1998 Bentham Screrrce Publishers B.V.
`
`AstraZeneea Exhibit 2023 p. 1
`lnnoPharma Licensing LLC V. AstraZeneea AB lPR2017-00905
`
`

`

`72 Current Phammceutical Design. I998, Vol. 4, No. I
`
`Grese and Dodge
`
`Me OH
`
`
`
`
`
`HO
`
`l7B-estradiol, 1
`
`estrone, 2
`
`DES. 3
`
`ON NEtz
`
`
`
`
`
`MER—25. 5
`
`hexestrol, 4
`
`tamoxifen, 6
`
`
`
`raloxifene, 7
`
`Fig. (1). Representative estrogen receptor modulators.
`
`reclassification of estrogen receptor ligands (Table l) [24] on
`the basis of
`their effects in various traditional and non—
`
`traditional target tissues. An explosion of research to understand
`the molecular basis for this specificity [25] and a race to develop
`these “designer estrogens" or Selective Estrogen Receptor
`Modulators (SERMS) as pharmaceutical products has also taken
`place [26]. The prototypical 2nd generation SERM, raloxifene,
`7,
`is currently undergoing clinical evaluation for the prevention
`and
`treatment of postmenopausal
`osteoporosis
`[’27],
`Nevertheless,
`it should be noted these distinctions may be
`somewhat arbitrary, since there is likely to be a continuum of
`activities from full agonist
`to full antagonist and the relative
`activity of an individual compound may be different for each
`tissue or animal species examined.
`
`In this review, we will discuss the known pharmacology of
`various structural classes of estrogen receptor modulators.
`particularly with respect
`to their effects in non—traditional
`tissues. We will describe the structure—activity relationships of
`these compounds, where such data is available, concentrating
`upon how elements of structure contribute to their tissue—specific
`actions. Finally, we will provide a brief overview of the current
`theories which have been developed to account
`for
`tissue
`specificity of ER-modulators.
`
`OH
`
`HO
`
`"(crime
`
`to 164384, 8a x = IO, R = CON(Me)n—Bu
`ICI [82780. Sb x = 9, R = SO(CHZ)3CF2CF3
`
`Steroidal ER Modulators and the Estrogen
`Pharmacophore
`
`Natural and synthetic steroidal estrogens have shown great
`utility and significant therapeutic benefits in the replacement of
`endogenous hormones in postmenopausal women [7—10,]4-18].
`Although most studies have focused on the efficacy of ERT or
`HRT in the prevention of osteoporosis, cardiovascular disease,
`and disorders of the urogenital
`tract [28], recent reports have
`also described benefits in the central nervous system, including
`improvements
`in cognitive
`function,
`and palliation of
`Alzheimer's disease and postmenopausal depression [1 1-
`1329.30].
`
`ERT and/or HRT have been demonstrated to provide a variety
`of cardiovascular benefits, resulting in a 40—50% reduction in the
`relative risk of coronary disease and atherosclerosis [31,32].
`The effects of estrogens on cardiovascular risk factors include
`raising serum levels of high—density lipoprotein (HDL)
`cholesterol and apolipoprotein A-l, and lowering levels of low—
`density lipoprotein (LDL)
`cholesterol,
`lipoprotein (a),
`endothelin—l, and apolipoprotein B [33,34]. Estrogen has also
`been demonstrated to have direct and indirect effects on blood
`
`vessel walls
`
`including increased nitric oxide synthesis,
`
`AstraZeneca Exhibit 2023 p. 2
`
`

`

`Estrogen Receptor Modulators
`
`Current Pharmaceutical Design, I 998. Vol. 4, No. I 73
`
`inhibition of vascular smooth muscle cell proliferation, and
`increased vasodilation [35]. Currently it
`is
`felt
`that
`the
`combination of these effects on serum lipids and on vascular
`tone are responsible for the overall cardioprotective effects of
`estrogen therapy.
`
`function
`In the prevention of osteoporosis, estrogens
`primarily as antiresorptive agents, leading to decreased turnover
`of both cortical and cancellous bone [36,37]. As with other
`antiresorptive agents,
`this benefit
`is partially offset by a
`subsequent decrease in bone formation, however the overall
`result of ERT or HRT is a substantial increase in bone mineral
`density and a decrease in fracture incidence [38,39]. Although
`ERs have been detected in both osteoblasts and osteoclasts, it is
`currently unclear if the effects of estrogens on bone metabolism
`are direct or indirect [40].
`
`Steroidal Estrogens
`
`Early efforts to identify selective estrogens focused on
`changes in the parent steroid to elicit tissue specific biological
`responses. For example,
`the estrogen metabolites estriol, 9, and
`l7ot-estradiol, 10, were found to be time—dependent mixed
`agonist—antagonists of estrogen, which
`stimulate
`early
`uterotrophic responses but have little effect on true uterine
`
`hypertrophy and hyperplasia unless administered chronically at
`high doses [4l,42]. Estriol causes significantly less uterine
`hyperplasia than l7B—estradiol and inhibits the development of
`breast cancer in rodents [43]. In addition,
`l70t—estradiol has been
`shown to exert
`a neuroprotective
`effect
`in
`a human
`neuroblastoma cell
`line
`(SK—N-SH)
`[44]. The estrogen
`metabolite, 2—methoxyestradiol, 11, has been implicated in the
`angiogenesis of vascular tissue and a number of analogs have
`been reported which potently inhibit
`tubulin polymerization
`[45,46]. The estrogen analog l70t-ethynylestradiol
`(EEZ), 12,
`has been extensively studied for its bone, uterine, and lipid
`effects due,
`in large part.
`to an enhanced oral activity profile
`relative to l7B-estradiol.
`
`Improvements in tissue selectivity have been observed with
`a family of D~ring halogenated estrones (such as 13) which have
`demonstrated potent
`lipid lowering yet diminished uterine
`hypertrophy relative to estrone [47]. Other attempts to attenuate
`the estrogentc activity of steroids via opening of the steroid
`nucleus, such as 9,ll—seco steroids, 14, have met with limited
`success [48,49].
`
`Recently, the components of Premarin® (the most prescribed
`form of ERT) have been evaluated for their lipid lowering effects.
`These conjugated equine estrogens contain sulfate esters of two
`distinct estrogen structural classes; (1) ring B saturated steroids
`
`
` McO
`
`14
`
`OH
`
` HO
`
`'l7B—dihydroquilinen, l7
`
`Me 9”
`
`
`
`l7ot—estradiol, 10
`
`
`
`MeO
`
`
`
`HO
`
`
`
`cquilin,15
`
`equilinen, 16
`
`
`
`I70L-dihydroequilinen, 18
`Fig. (2). Steroidal estrogen receptor modulators.
`
`
`
`tibolonc, l9
`
`AstraZeneca Exhibit 2023 p. 3
`
`

`

`74 Current Pharmaceutical Design. 1998‘, V01. 4, No. I
`
`Crete and Dodge
`
`including traditional sex steroid hormones such as estrone, 17B—
`estradiol, and l70t—estradiol, and (2) ring B unsaturated estrogens
`such as equilin (Eq), 15, equilenin (Eqn), 16,
`I7B«
`dihydroequilenin (l7B—DHEqn), 17,
`l7B—dihydroequilin (17B-
`DHEq),
`I70t—dihydroequilenin (I70t—DHEqn), 18, and l70t—
`dihydroequilin (I70t—DHEq). In I991. Bhavnani and co—workers
`examined these individual steroids,
`in their unconjugated form.
`to determine their relative binding affinities for the estrogen
`receptor and their in vivo effects on uterine hypertrophy in the
`immature rat
`[50].
`In this
`study,
`the majority of equine
`components mimicked estrogen in their ability to increase
`uterine weight relative to vehicle treated animals. The notable
`exception to this uterotrophic response was l70t—DHEqn which
`did not cause a significant effect at the dose examined (2 mg/kg).
`More recently,
`the sulfate ester conjugate of l7oc—DHEqn has
`been shown to
`lower
`serum cholesterol
`and increase
`
`hippocampal dendritic spine density in rats. and improve arterial
`vasomotor function in macaques [SI].
`
`Work from our own laboratories on the relative effects of
`
`conjugated equine estrogens on bone versus uterus has shown
`that
`l70t-DHEqn is a partial estrogen agonist [52]. In this study.
`uterotrophic effects were observed after 4 days of oral dosing for
`Eq. 15, Eqn. 16.
`l7B—DHEqn, 17, and l7OL-DHEqn. 18.
`Increases in uterine wet weight relative to ovariectomized (OVX)
`controls ranged from 263% for Eq to 100% for
`l7ot-DHEqn.
`Serum cholesterol levels were lowered with similar potencies for
`all equine estrogens [52]. Bone mineral density measurements
`indicated that
`l70t-DHEqn effectively prevented osteopenia in a
`dose-dependent
`fashion after 5—weeks of oral administration
`(59.9% of ovariectomy-induced bone loss was prevented at
`l
`mg/kg.
`ll9 % at 10 mg/kg).
`In addition, an average uterine
`weight gain of IOO.4% relative to OVX controls was observed at
`the l mg/kg dose [52]. These data demonstrate that
`l70taDHEqn
`is a full estrogen agonist on bone, but a partial agonist on the
`uterus in the OVX rat and further highlight
`the structural
`significance of both the stereochemistry at the l7—position and
`unsaturation in the Bvring.
`
`is a unique steroid that possesses
`Tibolone (OD-l4). 19,
`estrogenic, progestenic and androgenic properties. At doses of
`less
`than 2.5 mg/day, OD—l4 appears
`to reduce skeletal
`remodeling without producing concomitant
`endometrial
`stimulation [53]. However, because of its estrogenic activity,
`endometrial hypertrophy over
`the
`long term remains
`a
`possibility.
`
`Pure Antiestrogens
`
`While estrogen agonists, partial agonists, and SERMs can
`mimic the pharmacology of
`the natural hormone, pure
`antiestrogens (e. g., 8a,b) represent a class of therapeutic
`agents which are devoid of estrogen agonism regardless of the
`target
`tissue. Initially introduced by Wakeling in 1988,
`these
`compounds demonstrate an absence of estrogenic activity on the
`rat uterus, vagina, and hypothalamic-pituitary axis as well as
`effectively antagonizing the stimulatory effects of estrogen
`[54]. In non—reproductive tract tissue, pure antiestrogens behave
`like estrogen antagonists as well. For example,
`[(31 164,384.
`88, and ICI 182,790. 8b, exhibited no capacity for lowering
`serum cholesterol or sparing bone loss in the OVX rat model
`[55]. Recent data suggests that
`[Cl
`l82,780 has significantly
`complex effects on rat skeletal tissue [56]. For example.
`loss of
`
`cancellous bone is observed in intact rats after administration of
`the compound whereas no bone loss is observed in OVX rats.
`
`Estradiol Pharmacophore
`
`Recently. Katzenellenbogen, et. al. have combined literature
`ER binding affinity data for a large number of steroidal estrogen
`analogs with molecular modeling and receptor sequence analysis
`to develop a detailed picture of the estradiol pharmacophore
`[57]. Their study recognizes the important contributions of the
`two hydroxy groups of estradiol to receptor binding, with the 3-
`hydroxy acting primarily as
`a hydrogen bond donor and
`contributing approximately 1.9 kcal/mol
`to the binding free
`energy. while the
`I7B~hydr0xy functions primarily as
`a
`hydrogen bond acceptor and contributes approximately 0.6
`kcal/mol
`[58]. The preferred distance between the hydroxy
`functionalities appears to be somewhat flexible, possibly due to
`the inclusion of water molecules in the binding cavity [57].
`Large, preformed. hydrophobic pockets apparently exist within
`the ligand binding domain which are able to accommodate large
`substituents at the IIB— and 7ot-positions [59]. Smaller pockets
`appear to exist at the lofl and INS—positions. while the 160.-
`position and the aromatic A—ring are relatively intolerant of
`substitution [57]. These properties of the ER ligand binding
`cavity, which were primarily determined empirically, appear to
`be supported by the recently reported X-ray crystal structure of
`the ER ligand binding domain complexed with estradiol [60].
`
`Triphenylethylenes
`
`The most thoroughly investigated class of non—steroidal ER
`modulators
`are
`the
`triphenylethylenes
`tTPE’s),
`such as
`tamoxifen. 6. and clomiphene, 20. A common structural motif
`which is
`incorporated in many classes of
`tnolecules with
`estrogen antagonist activity involves the attachment of
`a
`sidechain containing a hydrogen bond acceptor to an ER binding
`core unit. This theme is illustrated for the triphenylethylenes via
`the progression from DES to MER—QS and tamoxifen. Originally
`investigated for contraceptive activity.
`the strong estrogen
`antagonist activity observed with many of these compounds in
`mammary tissue, has led to their development for treatment of
`breast cancer [20d]. The success of tamoxifen in this arena, has
`led to the investigation and development of a wide variety of
`analogs. To date, SAR work in this series has been Confined
`primarily to the investigation of antagonist effects in mammary
`and uterine tissue [20]. Recently, reports of estrogen agonist
`effects of some of
`these compounds in the skeletal and
`cardiovascular system have begun to appear [26].
`In general,
`although they partially antagonize the effects of estrogen on the
`uterus, the members of this structural class tend to induce some
`
`level of uterine stimulation in the absence of endogenous
`estrogen, therefore they have been classified as partial agonists
`or first generation SERMs [6i].
`
`to be utilized clinically was
`the first TPE’s
`One of
`clomiphene, 20. Although it was originally developed as a
`contraceptive, clomiphene has been mainly utilized for
`the
`induction of ovulation in anovulatory women [62]. Its effects on
`uterine tissue are complex. and are at least partly complicated by
`its availability as
`a mixture of double—bond
`isomers
`(zuclomiphene and enclomiphene), but
`it appears to cause
`significant stimulation of uterine epithelia in the rat [63,64].
`
`AstraZeneca Exhibit 2023 p. 4
`
`

`

`Estrogen Receptor Modulators
`
`Current Pharmaceutical Design, 1998’, V01. 4. No. I 75
`
`in
`Clomiphene has been reported to reduce serum cholesterol
`rats, similar to estrogen, however this may not be an ER-
`mediated effect
`[63]. Clomiphene has also been reported to
`inhibit bone resorption in vitro [65] and to protect against bone
`loss in both OVX rats [66] and in postmenopausal women [67].
`Interestingly,
`the individual
`isomers of clomiphene have been
`reported to have similar effects on bone metabolism, while the
`uterine effects are primarily induced by zuclomiphene [68].
`Recently a clomiphene analog, MDL—lO3.323, 2], with
`antiproliferative activity in breast cancer assays has been
`reported to protect against bone loss in OVX rats with minimal
`uterine stimulation [69].
`
`Consistent with the importance of the hydroxyl moieties of
`estradiol
`for receptor binding, tamoxifen, 6, binds only weakly
`to the ER, however evidence suggests
`that
`the primary
`biologically active species may be its 4—hydroxy metabolite
`[70,71]. Estrogen antagonist effects in mammary tissue have
`been demonstrated in a variety of cell
`lines and animal models
`[20d]. Tissue specific estrogen agonist effects have been
`demonstrated in the OVX rat model of estrogen deficiency, where
`
`tamoxifen reduced serum cholesterol by 50% at doses of [ll—10
`mg/kg and protected against bone loss with an ED50 of 0.l
`mg/kg [6l,72,‘73].
`In vitro effects on bone resorption and
`osteoclast viability have also been demonstrated [64,74].
`In a
`primate model.
`tamoxifen was shown to significantly inhibit
`the progression of coronary artery atherosclerosis [75].
`
`Due to the widespread use of tamoxifen in the treatment of
`breast cancer, a large body of clinical evidence with respect to
`its effects in other tissues has also accumulated [22].
`ln the
`cardiovascular
`system,
`tamoxifen
`has been shown to
`significantly reduce risk factors of disease including LDL
`cholesterol,
`ltpoprotein (a), and fibrinogen in postmenopausal
`women with little or no effect on triglycerides or HDL
`cholesterol [76.77]. A corresponding decrease in mortality due
`to cardiovascular disease has also been reported [78]. Clinical
`effects on the skeleton have included the preservation of bone
`mineral density at the lumbar spine, femoral neck, and forearm
`in postmenopausal women |77,79] as well as an estrogen—like
`reduction in serum markers of bone turnover
`[79b—d,80].
`Interestingly.
`in premenopausal women decreases in bone
`
`O M
`
`DL- I 03323, 21
`
`/\/\/ NELE
`
`O/\/ NMCE
`
`NEI
`
`O/\/
`
`2
`
`tamoxifen, 6
`
`clomtt‘ene. 20
`
`NMe
`
`O/\/
`
`2
`
`O/\/ NO
`
`NlVlew
`
`O/\/
`
`/
`
`1
`
`idoxifene. 23
`
`/
`
`Cl
`
`toremifene, 24
`
`ON NMef’t'
`
`OH
`droloxn'ene, 22
`
`('HO)2P(())O
`
`Fig. (3). Triphenylcthylene estrogen receptor modulators.
`
`TAT/59. 25
`
`TM], 26
`
`AstraZeneca Exhibit 2023 p. 5
`
`

`

`76 Current Pharmaceutical Design, [998, Vol. 4, No. l
`
`Grese and Dodge
`
`mineral density have been observed with tamoxifen treatment
`[79a,e].
`
`the
`recently been approved for
`Toremifene, 24, has
`treatment of breast cancer and has demonstrated clinical effects
`
`Notwithstanding the positive effects described above for
`tamoxifen, there continues to be considerable concern about the
`increased risk of endometrial cancer which has been associated
`
`with tamoxifen use [81,82]. Estimations of the magnitude of
`this risk vary, however the average value of about a five—fold
`increase is similar to that observed with ERT [83]. Significant
`stimulation of uterine endometrial tissue is also observed in the
`
`[61,84] and DNA adduct formation has been
`OVX rat model
`Observed in both rats and humans [85,86]. Tamoxifen has also
`been shown‘to induce liver cancer in rats [87].
`
`The growing concern over the potential cancer causing or
`cancer promoting effects of tamoxifen has recently led to the
`development of a number of tamoxifen analogs. The hypothesis
`that metabolic hydroxylation of tamoxifen at
`the 4-position is
`in some way responsible for
`these effects has
`led to the
`investigation of agents in which this metabolic pathway is
`blocked [20d]. Examples of this strategy include droloxifene,
`22, and idoxifene, 23. which have been reported to show
`decreased
`levels
`of DNA adduct
`formation
`and
`
`In toremifene, 24, chlorination
`hepatocarcinogenicity [88,89].
`of the aliphatic substituent of tamoxifen also appears to reduce
`DNA—adduct formation [90]. Alternative strategies for modifying
`the metabolic fate and/or tissue distribution observed with
`
`tamoxifen are represented by TAT—59, 25. and tamoxifen
`methiodide or TMI, 26. To date these compounds have been
`most extensively evaluated for the treatment of breast cancer
`[90,91], however recently reports on their effects in non—
`traditional target tissues have begun to appear.
`
`the 3-position of the
`in droloxifene, 22, hydroxylation at
`TPE core leads to an altered metabolic profile and decreased
`estrogen agonist activity relative to tamoxifen [92,93].
`ln OVX
`rats. droloxifene has been reported to reduce serum cholesterol
`40-46% and to protect against
`loss of bone mineral density,
`similar to tamoxifen but with reduced uterine stimulation [94]. in
`a head—to-head comparison with tamoxifen, droloxifene was
`found to be at least 10-fold less potent in terms of its effects on
`serum cholesterol and bone density, even though it has a tenfold
`higher binding affinity to the ER [95]. Estrogenic effects in the
`skeleton have also been observed by histornorphometry at both
`cancellous and cortical bone sites [96]. Recently, droloxifene
`has been observed to induce apoptosis of both MCF—7 cells and
`osteoclasts in culture, while estrogen has similar effects on
`osteoclasts but
`is mitogenic to MCF—7 cells [97]. This tissue-
`specific difference has led to the hypothesis that a common
`mechanism may account
`for both the estrogen agonist and
`antagonist activities of droloxifene. Although droloxifene has
`been evaluated Clinically for efficacy in breast cancer treatment
`[91a], its effects on other estrogen target tissues in humans have
`not yet been reported.
`
`idoxifene, 23, was designed to reduce both metabolic
`oxidation and N—demethylation, via iodination of the 4‘7
`position and replacement of
`the dimethylamino group of
`tamoxifen with a pyrrolidine ring, respectively [98]. As with
`droloxifene.
`idoxifene has been evaluated clinically for breast
`cancer treatment [91b]. and a preliminary report describing its
`effects on serum cholesterol and bone density in the OVX rat has
`also appeared [99].
`It has also been reported to be less
`uterotrOphic than tamoxifen [100].
`
`on serum cholesterol and bone mineral density which are similar
`to those of tamoxifen in postmenopausal breast cancer patients
`[101,102]. Although it has been reported to be less uterotrophic
`in the rat
`[103],
`its estrogenic effects on the uterus
`in
`postmenopausal women have been reported to be comparable to
`those of tamoxifen [104].
`
`Tamoxifen methiodide, 26, was designed to inhibit crossing
`of the blood-brain barrier,
`in order to avoid possible estrogen
`antagonist effects
`in the central nervous
`system [91d].
`Interestingly,
`this compound has recently been reported to
`selectively stimulate creatinine kinase activity in bone cells but
`not uterine cell
`lines, while tamoxifen and estrogen stimulate
`this activity in both [105]. Similar effects have been described
`in vivo, although correlation of these effects with bone density
`and uterine stimulation have not yet been reported [106].
`
`Two new TPE‘s which contain carboxylic acid functionality
`in place of the amine side chain moiety have also been reported.
`GW5638, 27. has been described as a bone—selective estrogen
`agonist. and has demonstrated decreased uterine stimulation,
`relative to tamoxifen,
`in OVX rats [107,108].
`Interestingly.
`amide analogs of 27 showed an increased tendency toward
`uterine stimulation both in vivo and in 1’fll‘0 [107].
`in OVX rats,
`27 was observed to maintain bone mineral density at both the
`lumbar spine and the proximal tibia with an efficacy similar to
`that of 17B-estradiol or
`tamoxifen at doses of 1—10 mg/kg
`[107,108]. it has also been shown to reduce serum cholesterol in
`OVX rats with a maximal efficacy of 20—30% [108,109]. The
`magnitude of this effect. although similar to that observed with
`17B»estradiol, appears to be somewhat muted in comparison to
`the more bioavailable l70t—ethynyl estradiol and other TPE’s,
`implying perhaps that multiple mechanisms may be involved in
`the regulation of serum lipid concentrations by these compounds
`[110]. Hydroxytamoxifen acid. 28. a tamoxifen metabolite, has
`also been reported to have bone—selective effects in the OVX rat
`i111],
`
`COZH
`
`/
`
`0A cola
`
`/
`
`\
`
`/
`
`\ \
`
`_ \Ho
`
`Gwssss, 27
`
`28
`
`Fig. (4). Acidic triphenylethylenes.
`
`Several groups have recently reported the application of
`parallel synthesis techniques for the preparation of TPE. libraries
`[112].
`it
`is expected that
`the ready availability of more
`structurally diverse members of this class,
`together with the
`development of molecular biological assays predictive of in
`FIFO tissue selectivity, will
`lead to greater understanding of the
`SAR of these compounds in multiple tissues.
`
`AstraZeneca Exhibit 2023 p. 6
`
`

`

`Estrogen Receptor Modulators
`
`Benzothiophenes
`
`In order to avoid the problems associated with double bond
`isomerization of the TPE‘s, a variety of cyclic frameworks have
`been investigated for their ER modulating properties Out of
`these
`structure—activity
`studies,
`raloxifene,
`7,
`a
`benzothiophene—containing compound with a unique profile of
`biological activity emerged [26h,c].
`.
`,
`Ralox1fene has been shown to bind the estrogen receptor
`with high affinity and to function as
`a potent estrogen
`antagonist
`in mammary tumor cells and in rat models of
`mammary cancer [113,114].
`In contrast,
`in the cardiovascular
`
`Current Phannaceutical Design, 1998, Vol. 4, No. l 77
`
`system, raloxifene functions primarily as an estrogen agonist.
`ln cell culture, raloxifene has demonstrated estrogen—like effects
`on vascular smooth muscle cells and on the inhibition of LDL
`oxidation [115,116]. in the OVX rat model, raloxifene has been
`shown to reduce serum cholesterol by 50—75% after l—5 weeks of
`daily dosing [117,118]. Most importantly,
`in postmenopausal
`women treated daily with raloxifene, significant reductions in
`total serum cholesterol and LDL cholesterol have been observed
`after both ei ht we k
`nd tw
`e
`'
`'
`.
`g
`B S a
`O y a” “treatment [27]
`Similarly,
`the effects of raloxifene on the skeleton seem to
`parallel
`those observed with estrogen.
`In vitch studies have
`shown similar effects of
`raloxifene and l7B—estradiol on
`
`Table 2.
`
`ER Binding and Inhibition of MCF-7 Cell Proliferation by 2-Aryl Raloxifene Analogs [127a]
`
` MCF-7 lnhib. [C50 (nM)C
`
` 4-0Hitame
`
`raloxifene, 7
`
`0.2
`
`estradiol
`
`0-36
`0.34
`
`
`
`6-0H
`
`470H
`
`1
`
`1
`'
`
`7a
`
`7b 7
`7c
`7d
`7t
`
`7f
`7g
`7h
`7i
`
`77j
`
`7
`
`7
`'
`
`1
`
`,7
`7
`
`7
`
`77
`
`7
`
`7
`
`7
`
`77
`7
`
`7
`
`none
`
`some 7
`76770H 7
`6-OMe
`77none
`
`none
`
`470Me
`40M:
`4—OH 7
`4—OH
`
`6701—1
`"my 7
`67Cl
`67—OH
`
`G—Me
`
`7
`
`7
`
`7
`
`7
`
`7
`
`.
`
`7
`
`none
`4-Cl
`4—UH
`47Me
`
`4.014
`
`7
`
`_
`77
`
`7
`
`7
`
`7
`
`7
`
`7
`
`7
`
`<0.002
`
`<0.002
`0.073
`0.008
`0.003
`
`7
`
`300 7
`
`30077
`100077
`2507 7
`3577
`
`7
`
`0.062 7
`0.046
`0.006
`0.07
`
`NAd
`
`7
`
`2.5
`I
`1000
`750 7
`
`300
`
`77
`
`7
`
`77
`
`7
`
`7
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`77
`
`
`
`
`77v
`;
`son 7
`xenon
`
`7 7w
`:
`6»OH
`7
`3«F,470H
`
`
`
`b-Ol—l 7x 3,5»di(Mc),4iOH
`h
`
`7
`
`7
`
`7
`
`7
`
`7
`
`7
`
`7
`
`.
`
`7k
`7717 7
`77r7n
`7n
`77 7o 7
`
`7
`
`7
`
`7
`
`7p
`7q
`7i
`
`78
`7t
`7711
`
`7
`
`7
`
`7
`
`7
`
`7
`
`76701-1
`7 777730171 7
`7747(7)}1
`5-011 7
`son 7
`
`6—O7H
`5-F,6»OH
`57M9,6!0H
`
`5,7-di(Me),6»O7H
`6iOH7
`son
`
`7
`
`7
`
`%
`
`.
`'
`
`7
`
`7
`
`4—1:
`4—OH
`40H 7
`4.014
`2—OH
`
`3—01-17
`40H
`7 40H
`
`4-OH
`'27Me77477017‘17
`737Me.470H
`
`‘
`‘
`
`:
`
`'
`
`77
`
`7
`
`0.19
`7 0.02
`<0002
`0.10
`0.057
`
`0.16
`0,098
`0.07
`
`0.005
`0.41
`0.13
`
`2.773
`730077
`1907
`10077
`to
`
`73,2 77
`377
`My
`
`5007
`2777
`71
`
`'
`
`7
`
`7
`
`7
`
`.
`
`7
`
`7-
`'
`
`7
`
`7
`
`7
`
`7
`
`7
`
`7
`
`77
`
`.
`
`7
`
`0.12
`0.20
`
`77237
`707.7377
`
`7
`
`77
`
`7
`
`7
`
`7
`
`
`
`(Dose quuired to give 50% inhibition
`Average of at least 2 determinations. Values are i 10%.
`“REA 2 relative binding affinity by competition with 3H71717i-estradiol.
`of a ttiaximally effective (IO'H) dose of 17B-estradiol. Average of atleast
`3 determinations, Values are i 10% dNA : not active at
`the doses tested,
`‘74,
`Hydroxytamoxifen, the primary biologically active metabolite of tamoxifen.
`1’ ND : not determined
`
`AstraZeneca Exhibit 2023 p. 7
`
`

`

`78 Current Pharmaceutical Design, 1998, V01. 4, N0. 1
`
`Grese and Dodge.-
`
`these metabolites show significantly reduced ER binding and
`activation in vitro, conversion to the parent molecule via
`deeonjugation has been shown to occur at the tissue level [131].
`Interestingly. no significant differences
`in conversion at
`various target organs such as uterus, bone, and liver have been
`observed.
`
`Table 3.
`
`ER Binding and Inhibition of MCF—7 Cell Proliferation by
`2-Alky1, Z-Naphthyl, and 2-Heteroaryl Raloxifene Analogs
`[127a]
`
`O
`
`O\/’\
`
`N
`
`osteoclastogenesis and on creatinine kinase activity in human
`osteoblast cells
`[119,120]
`In rats.
`several
`studies have
`demonstrated a protective effect against ovariectomy—indueed
`osteopenia at doses as
`low as 0.1 mg/kg [117,121,122].
`Positive effects on bone mineral density at both cortical and
`cancellous bone sites have been reported. as have positive
`effects on bone strength [121,123].
`Interestingly, although
`raloxifene suppresses bone resorption in the rat with efficacy
`which is approximately equal
`to that of estrogen. bone
`formation appears to be suppressed to a lesser degree, resulting
`in a net gain in bone mass with raloxifene [121,123b]. These
`positive results in animal studies, have now been confirmed with
`clinical studies in postmenopausal women [27]. Significant
`positive effects on histomorphometrie parameters, bone
`markers, and on bone mineral density at both the lumbar spine
`and hip were observed after two years of raloxifene treatment
`[27,124].
`
`In terms of its pharmacology, raloxifene is distinguished
`from the TPE‘s primarily on the basis of its effects on the uterus,
`where a qualitative difference has been observed [61,125].
`In a
`direct comparison with tamoxifen, droloxifene, and idoxifene.
`raloxifene was a significantly more effective antagonist of
`estrogen action in the immature female rat uterus [61a] In this
`assay the TPE's functioned as partial agonists.
`inhibiting the
`effects of estrogen on uterine weight gain only to the level of
`their own intrinsic agonist activity. while raloxifene functioned
`essentially as a complete antagonist. Similarly.
`in OVX rats, the
`TPE's have been found to induce a larger maximal stimulation of
`uterine weight and to induce uterine eosinophilia while
`raloxifene did not
`[61]. Although raloxifene has also been
`reported to stimulate a modest
`increase in uterine wet weight,
`this increase is not dose related and is not coincident with
`
`increases in other measures of uterine hypertrophy, and has
`therefore been attributed to water
`

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