throbber

`
`PERGAMON
`
`Journal of Steroid Biochemistry and Molecular Biology 69 (1999) 51-84
`
`TheJournal of
`Steroid Biochemistry
`&
`Molecular Biology
`
`EM-652 (SCH 57068), a third generation SERM acting as pure
`antiestrogen in the mammary gland and endometrium”
`
`Fernand Labrie*, Claude Labrie, Alain Bélanger, Jacques Simard, Sylvain Gauthier,
`Van Luu-The, Yves Mérand, Vincent Giguere, Bernard Candas, Shouqi Luo,
`Céline Martel, Shankar Mohan Singh, Marc Fournier, Agnes Coquet, Virgile Richard,
`Ronald Charbonneau, Gilles Charpenet, André Tremblay, Gilles Tremblay,
`Lionel Cusan, Raymonde Veilleux
`
`Oncology and Molecular Endocrinology Research Center, Centre Hospitalier Universitaire de Québec (CHUQ), Pavilion CHUL, Department of
`Medicine, Laval University, Québec, G1V 4G2, Canada
`
`
`
`Abstract
`
`Breast cancer is the most frequent cancer in women while it is the second cause of cancer death. Estrogens are well recognized
`to play the predominantrole in breast cancer development and growth and much efforts have been devoted to the blockade of
`estrogen formation and action. The most widely used therapy of breast cancer which has shown benefits at all stages of the
`disease is the use of the antiestrogen Tamoxifen. This compound, however, possesses mixed agonist and antagonist activity and
`major efforts have been devoted to the development of compounds having pure antiestrogenic activity in the mammary gland
`and endometrium. Such a compound would avoid the problem of stimulation of the endometrium and the risk of endometrial
`carcinoma. We have thus synthesized an orally active non-steroidal antiestrogen, EM-652 (SCH 57068) and the prodrug EM-800
`(SCH57050) which are the most potent of the known antiestrogens. EM-652 is the compound having the highest affinity for the
`estrogen receptor,
`including estradiol. It has higher affinity for the ER than ICI 182780, hydroxytamoxifen,
`raloxifene,
`droloxifene and hydroxytoremifene. EM-652 has the most potent inhibitory activity on both ERa and ERf comparedto any of
`the other antiestrogens tested. An important aspect of EM-652 is that it inhibits both the AF1 and AF2 functions of both ERa
`and ERf while the inhibitory action of hydroxytamoxifen is limited to AF2,
`the ligand-dependent function of the estrogen
`receptors. AF1 activity is constitutive, ligand-independent and is responsible for mediation of the activity of growth factors and
`of the ras oncogene and MAP-kinase pathway. EM-652 inhibits Ras-induced transcriptional activity of ERa and ERf and
`blocks SRC-1-stimulated activity of the two receptors. EM-652 was also found to block the recruitment of SRC-1 at AF1 of
`ER§,this ligand-independent activation of AF1 being closely related to phosphorylation of the steroid receptors by protein
`kinase. Most importantly, the antiestrogen hydroxytamoxifen has no inhibitory effect on the SRC-1-induced ERP activity while
`the pure antiestrogen EM-652 completely abolishes this effect, thus strengthening the need to use pure antiestrogens in breast
`cancer therapy in order to control all known aspects of ER-regulated gene expression. In fact, the absence of blockade of AF2
`by hydroxytamoxifen could explain why the benefits of tamoxifen observed up to 5 years become negative at longer time
`intervals and why resistance develops to tamoxifen. EM-800,
`the prodrug of EM-652, has been shown to prevent
`the
`development of dimethylbenz(a)anthracene (DMBA)-induced mammary carcinomain the rat, a well-recognized model of human
`breast cancer. It is of interest that the addition of dehydroepiandrosterone, a precursor of androgens,
`to EM-800,
`led to
`complete inhibition of tumor development in this model. Not only the development, but also the growth of established DMBA-
`induced mammary carcinoma was inhibited by treatment with EM-800. An inhibitory effect was also observed when
`medroxyprogesterone was added to treatment with EM-800. Uterine size was reduced to castration levels in the groups of
`animals treated with EM-800. An almost complete disappearance of estrogen receptors was observed in the uterus, vaginum and
`
`* Proceedings of Xth International Congress on Hormonal Steroids, Quebec, Canada, 17-21 June 1998.
`* Corresponding author. Tel.: + (418) 654-2704; fax: + (418) 654-2735.
`E-mail address: fernand.labrie@crchul.ulaval.ca (F. Labrie)
`
`0960-0760/99/$ - see front matter © 1999 Published by Elsevier Science Ltd. All rights reserved.
`PII: $0960-0760(99)00065-5
`
`AstraZeneca Exhibit 2034 p. 1
`InnoPharma Licensing LLC v. AstraZeneca AB IPR2017-00904
`
`

`

`52
`
`F. Labrie et al. | Journal of Steroid Biochemistry and Molecular Biology 69 (1999) 51-84
`
`tumors in nude mice treated with EM-800. EM-652 was the most potent antiestrogen to inhibit the growth of human breast
`cancer ZR-75-1, MCF-7 and T-47D cells in vitro when compared with ICI 182780, ICI 164384, hydroxytamoxifen, and
`droloxifene. Moreover, EM-652 and EM-800 have no stimulatory effect on the basal levels of cell proliferation in the absence of
`E2 while hydroxytamoxifen and droloxifene had a stimulatory effect on the basal growth of T-47D and ZR-75-1 cells. EM-652
`was also the most potent inhibitor of the percentage of cycling cancer cells. When human breast cancer ZR-75-1 xenografts were
`grown in nude mice, EM-800 led to a complete inhibition of the stimulatory effect of estrogens in ovariectomized mice while
`tamoxifen was less potent and even stimulated the growth of the tumors in the absence of estrogens,
`thus illustrating the
`stimulatory effect of tamoxifen on breast cancer growth. When incubated with human Ishikawa endometrial carcinoma cells,
`EM-800 had no stimulatory effect on alkaline phosphatase activity, an estrogen-sensitive parameter. Raloxifene, droloxifene,
`hydroxytoremifene and hydroxytamoxifen, on the other hand,all stimulated to various extent, the activity of this enzyme. The
`stimulatory effect of all
`four compounds was blocked by EM-800,
`thus confirming their estrogenic activity in human
`endometrial
`tissue. When administered to ovariectomized animals, EM-800 prevents bone loss,
`the effect on bone mineral
`density, trabecular bone volume, and trabecular separation being 5—10 times more potent than raloxifene. EM-800 lowers serum
`cholesterol and triglyceride levels in the rat as well as in women. In a Phase II study performed in patients with breast cancer
`showing failure on tamoxifen, 1 patient had a complete response while 5 patients had a partial response and stable disease for at
`least three months has been observed in an additional 13 patients for a total of 19 positive responses out of 43 evaluable
`patients (44.2%). No significant secondary effect related to the drug was observed. A phase 3 international clinical
`trial
`is
`currently being performed in tamoxifen failure patients where EM-800 (SCH 57050) is compared to Arimidex. The detailed
`information obtained at the preclinical level with EM-652 or EM-800 indicates that these orally active compounds are highly
`potent and pure antiestrogens in the mammary gland and endometrium while they prevent bone loss and lower serum
`cholesterol and triglyceride levels. Preclinical and clinical data clearly suggest the interest of studying this compound in the
`neoadjuvant and adjuvant settings and, most importantly, for the prevention of breast and uterine cancer in which settings they
`should provide additional benefits on bone and lipids. © 1999 Published by Elsevier Science Ltd. All rights reserved.
`
`Keywords: Pure antiestrogen; Breast cancer; Uterine cancer; EM-652; SCH 57068; EM-800; SCH 57050; Osteoporosis; Selective estrogen receptor
`modulator (SERM); Cholesterol; Triglycerides; Prevention; Risk reduction; Adjuvant; Neoadjuvant
`
`1. Introduction
`
`1.1. Breast cancer
`
`Breast cancer is the most frequent cancer in women,
`with 176,300 new cases and 43,700 deaths predicted in
`the United States in 1999 [1]. Present
`therapies in
`breast cancer achieve meaningful clinical
`results in
`only 30-40% of patients, with response duration
`usually limited to 12-18 months [2—5]. Five-year survi-
`val in women with metastatic disease is still only 10-
`40%.
`Amongall risk factors, estrogens are well recognized
`to play the predominantrole in breast cancer develop-
`ment and growth [6-9]. However, existing surgical or
`medical ablative procedures do not result in complete
`elimination of estrogens in women [10], due to the con-
`tribution of the adrenal glands that secrete high levels
`of dehydroepiandrosterone (DHEA) and DHEA-sul-
`fate which are converted into estrogens in peripheral
`target tissues [11-13]. Considerable attention has thus
`been focused on the development of blockers of estro-
`gen biosynthesis and action [14-20].
`Since the first step in the action of estrogens in tar-
`get tissues is binding to the estrogen receptor [21,22], a
`logical approach for the treatment of estrogen-sensitive
`breast cancer is the use of antiestrogens, or compounds
`which block the interaction of estrogens with their
`specific receptor. Until very recently, no agent with
`
`pure antiestrogenic activity under in vivo conditions
`has been available.
`
`1.2. Tamoxifen
`
`the antiestrogen most widely used for
`Tamoxifen,
`the treatment of women with breast cancer has shown
`clear clinical benefit in advanced breast cancer, its effi-
`cacy being comparable to that achieved with ablative
`and additive therapies [23]. In the first clinical studies
`initiated in 1969,
`tamoxifen was found to achieve
`remissions in advanced breast carcinoma similar
`to
`those observed following estrogen therapy but with
`fewer side effects [24]. Since then, because ofits favor-
`able safety profile and clinical efficacy comparable to
`other endocrine therapies,
`including oophorectomy
`and estrogens, tamoxifen has become the treatment of
`choice for patients with advanced or metastatic breast
`cancer [25—27]. This compound, however, is known to
`possess mixed estrogenic and antiestrogenic activities
`[19,23,28] which are species-,
`tissue-, cell-, and even
`gene-specific [29,30]. In support of the clinical evidence
`for
`the estrogenic activity of tamoxifen on human
`breast cancer growth [31,32], tamoxifen and its active
`metabolite 4-OH-tamoxifen have been found to stimu-
`late the growth of human breast cancer cells in vitro
`and in vivo [29,33—40]. Tamoxifen may act as an estro-
`gen agonist more frequently than generally thought
`and this may explain some of the apparent paradoxes
`
`AstraZeneca Exhibit 2034 p. 2
`
`

`

`F. Labrie et al. | Journal of Steroid Biochemistry and Molecular Biology 69 (1999) 51-84
`
`53
`
`
`
`EM-652 R=H
`EM-800 R=COC(CHs)3
`
`X=Y=Z=H, R)=R2=CH3
`Tamoxifen
`Droloxifene X=H, Y=OH, Z=H, Ri=Ro=CHyg
`Toremifene X=Y=H, Z=Cl, R=R2=CH3
`Idoxifene
`X=l, Y=Z=H, Ry,R2=C4Hg
`
`OH
`
`HX
`
`HO
`IC} 164,384
`EM-139
`IC1 182,780
`
`CHa),
`n=
`X=H,
`X=Cl, n=10, RECON(CH;)C,H9
`X=H
`
`
`
`Raloxifene
`
`Fig. 1. Antiestrogens—molecular structures.
`
`[5,14,16,18,19,62,63], especially ICI 164,384, EM-139,
`and ICI 182,780 (Fig. 1).
`These compounds have been shown to possess pure
`and potent antiestrogenic activity in most well recog-
`
`of endocrine treatments such as response to second en-
`docrine therapy and withdrawal responses [27].
`are
`Additionally, while
`benefits
`of
`tamoxifen
`observed on breast cancer in up to 40% of patients,
`the long-term use of this compound has recently been
`recognized as being associated with a_
`significant
`increase in the incidence of endometrial carcinoma
`[41-55], an effect which is likely caused by the intrinsic
`estrogenic activity of the compound and_possibly
`because of its genotoxic action on the DNA,by form-
`ing DNA adducts. The close analogs of tamoxifen,
`namely toremifene,
`Idoxifene and droloxifene, also
`possess estrogenic effects analogous to those of tamox-
`ifen [56,57 data not shown].
`
`2. Need for an orally active pure antiestrogen in the
`mammary gland and endometrium
`
`Since clinical data suggest that long-term (5 years)
`tamoxifen adjuvant therapy is preferable to the short-
`term (2 years) use of the antiestrogen [58,59] and stu-
`dies are in progress on the long-term use of tamoxifen
`as a chemo-preventive in breast cancer [54,60,61],
`it
`has become important to develop a pure antiestrogen
`to avoid the negative effects of the partial estrogenic
`activity of Tamoxifen and thus make available a com-
`pound having activities limited to the desired thera-
`peutic action. The first class of pure antiestrogens
`obtained were
`7a-substituted estradiol
`derivatives
`
`EM 652
`
`Fig. 2. Structure of EM-652 (SCH 57068).
`
`AstraZeneca Exhibit 2034 p. 3
`
`

`

`54
`
`Table 1
`
`F. Labrie et al. | Journal of Steroid Biochemistry and Molecular Biology 69 (1999) 51-84
`
`Comparison of the estrogen receptor affinity of a series of antiestrogens and related compoundswith estradiol (E2) and diethylstilbestrol (DES)
`in human breast cancer and normal humanuterine cytosol*
`
`Breast Cancer
`Uterus
`
`
`ethanol
`DMF
`ethanol
`DMF
`
`
`Compounds
`K; (nM) (max)
`RBA
`K; (max)
`RBA
`K; (max)
`RBA
`K; (max)
`RBA
`
`
`0.181
`0.113
`100
`0.138
`FE,
`-
`-
`110
`0.126
`DES
`0.069
`0.076
`291
`0.047
`(S)-6(EM-652)
`-
`-
`6.62
`2.09
`(R)-6(EM-651)
`-
`-
`2.32
`4.71
`(S)-1(EM-800)
`-
`-
`< 0.04
`> 270
`(R)-1(EM-776)
`10.3
`1.76
`1.53
`3.00
`4.60
`ICI 164,384
`27.2
`0.668
`-
`0.755
`1.81
`7.63
`ICI 182,780
`-
`-
`43.8
`0.346
`-
`-
`43.8
`0.249
`(Z)-4-OH-Tamoxifen
`
`
`
`
`
`
`
`
`11.9 0.92 - - 34.4 0.92 -Tamoxifen -
`
`100
`-
`150
`-
`-
`-
`
`746
`15.1
`
`0.120
`0.128
`0.042
`1.89
`11.14
`-
`
`2.33
`
`-
`
`100
`93.5
`284
`6.34
`1.32
`
`-
`
`5.15
`
`100
`-
`264
`-
`-
`-
`
`* Incubations were performed at room temperature for 3 h using 100 pL of cytosol, 100 pL of PHJE2 (5 nM Ep,final) and 100 pL of the indi-
`cated unlabelled compoundsleading to final concentrations of 3.3% ethanol or 2.5% dimethylformamide (DMF). The apparent inhibition con-
`stant (Kj) and relative binding affinity (RBA) values were calculated as described [73,223]. The apparent inhibition constant K; values were
`calculated according to the following equation: K;=ICs9/(1+S/K) where S represents the concentration of labelled E, and Kis the Kp value of
`E> (0.14 nM) for the estrogen receptor. RBA values were calculated as follows: RBA =ICsg of Ep/ICso of tested compound x100 [56].
`
`cancer and normal human uterine cytosol as described
`[56,73]. As measured by competition studies in human
`breast cancer tissue, the affinity of EM-652 (K;=0.047
`+0.003 nM, RBA =291, relative to 17f-estradiol set at
`100) studied in the presence of ethanol was 2.9 higher
`
`100
`
`oa
`
`a
`
`40
`
`80
`
`60
`
`©O-O E2
`@-@ EM-652
`
`[--L] Ici 182780
`
`20 mm ici 164384
`
`ZA DROLOXIFENE
`
`AA TOREMIFENE
`
`
`
`-11
`
`-10
`
`9
`
`“8
`
`7
`
`-6
`
`“5
`
`including human
`nized in vitro and in vivo systems,
`breast cancer cells
`[14,16,18,19,64,65]. The 7a-alkyl
`estradiol derivative ICI 164384, however, has been
`found to possess some estrogenic agonistic activity in
`guinea pig uterine cells
`[66,67]. Furthermore, both
`OH-tamoxifen and ICI-164384 can stimulate CAT ac-
`tivity in MCF-7 cells transfected with a pS2-tkCAT
`fusion gene [68]. Moreover, such 7a-alkyl estradiol de-
`rivatives are difficult to synthesize and their bioavail-
`ability by the oral route is very low, thus necessitating
`parenteral administration. We therefore concentrated
`our efforts on the synthesis of non-steroidal com-
`pounds having oral activity in order to overcome this
`difficulty.
`In order to facilitate large-scale purification, EM-
`800 (SCH 57050), the bipivalate derivative of EM-652
`was synthesized. EM-800 is rapidly transformed into
`EM-652 in intact cells and following in vivo adminis-
`tration. The other derivative currently used in our stu-
`dies is EM-652.HC1] (SCH 57068.HCl). In an effort to
`develop an orally active agent, EM-652 was
`syn-
`thesized (Fig. 2). As will be discussed later, the active
`compound EM-652 derived from EM-800 or EM-
`652.HCI behaves as a highly potent and pure antiestro-
`gen in human breast and uterine cancer cells in vitro
`as well as in vivo in nude mice [56,69—72].
`
`3. Binding characteristics to the estrogen receptors «
`and B
`
`The estrogen receptor affinity of EM-652, the active
`drug of EM-800, was first measured in human breast
`
`[3H]E2BOUND(%)
`
`CONCENTRATION(LOG M)
`
`ICI
`Fig. 3. Effect of increasing concentrations of EM-652, E2,
`182780, Droloxifene, ICI 164384, and Toremifene on [°H] 178-estra-
`diol binding to the rat uterine estrogen receptor. The incubation was
`performed with 5 nM [PH] 17f-estradiol (E,) for 2 h at room tem-
`perature in the presence or absence of the indicated concentrations
`of unlabeled compounds[224].
`
`AstraZeneca Exhibit 2034 p. 4
`
`

`

`F. Labrie et al. | Journal of Steroid Biochemistry and Molecular Biology 69 (1999) 51-84
`
`55
`
`Binding properties of estrogen receptors
`
`
`
`Foldinduction
`
`Bound/Free
`
`2 —
`
`0.5
`
`0.4
`
`oO @
`
`o NS
`
`
`
`Bound(fmol)
`
`3
`2 (nM)
`
`4
`
` 3 m 0 2 3
`
`0
`
`
`100=.200 300 0 50 100 150
`
`12
`-10
`8
`6
`4
`0
`E2 concentration (logM)
`Bound (fmol)
`
`
`
`
`
`Fig. 4. Dose-response and binding properties of mERa« and mER. (A) Cos-1 cells were transfected with 500 ng mER® (open circles) or mMER«
`(closed circles) expression vectors and 1 wg vitA,-ERE-TKLuc and then incubated for 12 h with increasing concentrations of E, as indicated. (B)
`Specific binding of [2,4,6,7-3H]-17f-estradiol (PHJE2) to mERf# was determined using receptors generated from rabbit reticulocyte lysates.
`Binding was determined over a concentration range of 0.01-3 nM [HIJE,in the absence or presence of a 200-fold excess of unlabeled E>. Thesat-
`uration plot is shown in the inset, and results were plotted by the method of Scatchard. Each point was determined in triplicate in each exper-
`iment, and the above results are representative of at least two separate experiments. (C) Specific binding to mER« using the conditions described
`in panel B [65].
`
`(RBA=6.62). Similar
`itself
`than that of estradiol
`results were obtained on the human uterine estrogen
`receptor (Table 1). It can be seen in the same table
`that ICI 182,780 has about 10 times lower affinity than
`EM-652 to displace [HJE. from the human estrogen
`receptor while (Z)-4-OH-Tamoxifen is about 6 times
`less potent under the experimental conditions used.
`The new antiestrogen EM-652 thus shows the highest
`affinity for the human estrogen receptor ofall the com-
`pounds tested [56] (Table 1).
`It can be seen in Fig. 3 that EM-652 is 7- to 8-fold
`more potent
`than E2 and ICI 182780 in displacing
`CHJE from the rat uterine estrogen receptor (ICso
`values of 0.52, 4.13, and 3.59 nM for EM-652, E>, and
`ICI 182780, respectively). ICI 164384 and Droloxifene
`are 21-fold less potent than EM-652 while Toremifene
`is 400 times less potent than EM-652.
`Over the past decade, all the studies on the elucida-
`tion of the molecular events underlying the mode of
`ER action as well as the antiestrogen-designed therapy
`have focused on the ERa identified and cloned several
`years ago [21,74,75]. Recently, a second estrogen recep-
`tor, designated ER, has been described and shown to
`share commonstructural and functional characteristics
`with ERe [65,76,77]. Based on amino acid sequence
`comparison, ERf shares with ERa the same modular
`
`[78].
`(A-F)
`six domains
`composed of
`structure
`Domain C, which contains the two zinc fingers respon-
`sible for DNA binding, is the most conserved followed
`by domain E, responsible for ligand binding, homodi-
`merization and nuclear localization. Domain E also
`contains a ligand-dependentactivation function (AF-2)
`involved in trans-activation by the ERs. A second acti-
`vation function, AF-1, resides in the A/B domain and
`acts in a ligand-independent manner [79-81].
`Both ERs recognize a specific estrogen response el-
`ement (ERE) composed of two AGGTCA motif half-
`sites configured as a palindrome spaced by three
`nucleotides [65]. ERa has also been shown to interact
`with a number of coregulators via the AF-2 domain,
`and these protein-protein interactions promote tran-
`scriptional
`regulation
`of
`target
`genes
`[82-85].
`Following cloning of mouse ERf [65], comparison
`could be made of the activity of ER« and ERf and
`measurement could be made of the affinity of the two
`ERsfor various ligands, especially, antiestrogens.
`Wefirst tested the activity of both receptors in
`the presence of increasing E, concentrations using
`the
`vitA2-ERE-TKLuc
`reporter
`in Cos-1
`cells.
`Comparison of the dose-response curves of Fig. 4A
`shows a shift of approximately 4-fold of the E, con-
`centration required to achieve half of the maximal
`
`AstraZeneca Exhibit 2034 p. 5
`
`

`

`56
`
`F. Labrie et al. | Journal of Steroid Biochemistry and Molecular Biology 69 (1999) 51-84
`
`125
`
`100
`
`
`
`oi“Ioai
`
` °o
`repressionofE2response(%) R
`
`A
`
`B
`
`ERo
`
`1C1182,780
`
`1C1182,780
`
`-
`
`E2 -11-10 -9 -8
`
`-7
`
`-
`
`E2 -11-10 -9
`
`-8
`
`-7
`
`antagonist concentration (logM)
`
`Fig. 5. Dose-response of antagonists on ERand ER/-mediated
`transactivation. Comparison of the dose-responses of the antagonists
`in the presence of 10 nM E, on the transcriptional activity of ERa
`(A) and ERS (B) using the vitA,ERETKLucreporter in COS-1 cells.
`Results represent the mean --SEM ofthree separate experiments and.
`are expressed as percentages of the maximal induction by EF, alone
`(set arbitrarily at 100%); filled bars) for the two ERs. The untreated
`ERa and ERf basal levels are also shown [105].
`
`level of induction between the two receptors, the ERe
`being moresensitive to Ep.
`The above-indicated results already suggested that
`ERf may have loweraffinity for E, than mERa«. To
`verify if the difference in E, responsiveness was due to
`a difference in ligand binding, we performed a binding
`analysis on both mERf# and mER«. PHJE was used
`to conduct binding studies with mERf, and results
`were plotted by the method of Scatchard. As shown in
`Fig. 4B,
`this analysis yielded an average dissociation
`constant (Kg) of 0.5 nM for E, when performed on
`ERf prepared from rabbit reticulocyte lysates. This
`value is comparable to that obtained for the rat ERP,
`which was reported to be 0.6 nM [76]. On the other
`hand, we obtained an average Kg of 0.2 nM for mER«
`(Fig. 4C), which is well within the range of previously
`published determinations for the cloned humanrecep-
`tor [86]. Therefore,
`this slightly reduced affinity of
`mERf for E, may provide an explanation for the shift
`in E, responsiveness indicated by the dose-response
`curves (Fig. 4A).
`To further evaluate the potency of various antiestro-
`gens, we compared their dose-dependent inhibition of
`E,-induced
`ERa«
`and
` ERf
`activity
`using
`vitA,ERETKLuc in COS-1 cells (Fig. 5). When com-
`pared to ICI182,780, EM-652 was highly effective,
`achieving a complete blockade of the E,-induced effect
`of ERa (Fig. 5A) and ERf (Fig. 5B) at concentrations
`of 10° M and above. Comparison of the apparent
`ICs values showed that under the conditions used,
`EM-652 was more potent in repressing ERwa activity
`(Csg=2 nM) than ICI182,780 UCsg=20 nM). Both
`
`ERoa, ERB
`
`HN-| [ara]sd-2]~~ COOH
`
`Ligand-
`dependent
`+
`
`coactivators
`
`Ligand-
`independent
`Constitutive
`
`But activated by
`dopamine
`growth factors
`PKAactivators
`
`Cyclic AMP
`Ras-MAPkinase pathway
`
`Fig. 6. Schematic representation of the activation functions 1 and 2
`of ERa and ERS. AF-1 is ligand-independent but is activated by
`dopamine, growth factors, cyclic AMP, MAP kinase, PKA activators
`and RAS. AF-2 on the other hand, is activated by estrogenic com-
`pounds. EM-652 blocks both AF-1 and AF-2 completely while OH-
`Tamoxifen blocks AF-2 only.
`
`antiestrogens were more effective to inhibit ERf than
`ERe« function with ICs) values of 0.4 nM and 8 nM
`for EM-652 and ICI182,780, respectively. In addition,
`lower concentrations of EM-652 in the 107'°-107!’ M
`range contributed already to a 25-30% reduction in
`the E, response of both ERs, and even when added at
`1073 M, EM-652 already showed a 20-25% repres-
`sion (data not shown).
`
`4. EM-652 inhibits both AF-1 and AF-2 functions of
`ERo and ERB
`
`As mentioned above, the two ERs share many func-
`tional characteristics based on their well conserved
`modular structure (Fig. 6). AF-2 is responsible for es-
`trogen-dependent activation through recruitment of
`coactivator proteins including members of the steroid
`receptor coactivator (SRC) family [85,87—93]. On the
`other hand, AF-1 activity is constitutive and ligand-
`independent [79-81].
`In addition to the classical hormoneactivation path-
`way, a numberof steroid receptors including ERx and
`B have been shown to be activated by non steroidal
`agents (Fig. 6) including dopamine, growth factors and
`PKAactivators [65,94—98].
`
`4.1. EM-652 inhibits RAS-induced transcriptional
`activity of ERa and ERB
`
`Potential phosphorylation of serine 118 in human
`ERe« [96,99,100] and serine 60 in mouse ERP [65]
`through activation of the Ras-MAPK pathway has
`been shown to further maximize the E, response of
`both estrogen receptors. To investigate whether EM-
`652 could efficiently block this effect, we used the wild-
`type H-Ras and its dominant active form H-RasY”” in
`
`AstraZeneca Exhibit 2034 p. 6
`
`

`

`F. Labrie et al. | Journal of Steroid Biochemistry and Molecular Biology 69 (1999) 51-84
`
`37
`
`SHAN -
`a RXCGq03#RD
`foldactivation 3
`nNa 0 -12-11-10 -9 -8 -7
`
` ao%ofmaximalresponse
`
`A
`
`Es
`EMS52
`iCliaa,yeg
`
`ae
`my
`:
`input st GST-nEReDEr GST-mERSEF
`al
`Babe Se &
`-
`- ee
`= Oe a eo
`~
`mec
`ache
`pe
`ag
`aes wl
`
`
`
`
`
`
`
`
`
`
`Control
`
`a O
`
`B
`
`E2 +
`EM-652 cl
`
`E2
`
`E2+ E2+
`EM-652 ICI
`
`1e1182,780
`
`1C1182,780
`
`= oi
`
`_— Na
`
`= QooO
`
`~ou
`
`0 -12-11-10 -9 -8 -7
`
`antagonist concentration (logM)
`
`Fig. 7. EM-652 blocks the Ras-induced ERo and ER#transcriptional
`activity.
`(A) COS-1
`cells were
`cotransfected with
`1
`pg
`vitA,ZERETKLuc and 500 ng pCMX-ER« in the presence or absence
`of 1 yg Ha-Ras or Ha-Ras‘!” expression plasmids. The cells were
`then grown in the presence or absence of 10 nM E> or 100 nM of
`EM-652 or ICI 182,780 (ICI). The basal activity of ERa in the
`absence of estradiol was set arbitrarily at 1.0. (B), same as in (A),
`except that ERf expression vector was used. (C). Dose responses of
`EM-652(filled squares) and ICI 182,780 (open squares) in the pre-
`sence of 10 nM E, on ER« activity in COS-1 cells transfected with
`vitA,ERETKLuc reporter and Ha-Ras’'* expression plasmid. The
`maximal induction by E, alone was set arbitrarily at 100%.
`(D)
`same as in (C) except that ERB expression vector was used [105].
`
`
`
`Fig. 8. EM-652 blocks the estrogen and SRC-1-stimulated AF2 ac-
`tivity of ERa and ER§. (A) GST pull-down experiments. The puri-
`fied fusion proteins were incubated with labeled SRC-1 in the
`absence (lanes 3 and 7) or presence of 5 nM Ep (lanes 4-6 and 8-10)
`in addition to a 100-fold excess of EM-652 (lanes 5 and 9) and ICI
`182,780 (lanes 6 and 10). The input lane (lane 1) represents 20% of
`the total amount of labeled SRC-1 used in each binding reaction. An
`equivalent amount of protein was used in the sample containing only
`GST (lane 2).
`(B) COS-1 cells were cotransfected with 1 pg
`vitA,ZERETKLuc and 500 ng pCMX-ER« in the presence or absence
`of 1 ug SRC-1 expression plasmid. Cells were incubated with or
`without 10 nM E, or 100 nM antagonist as indicated. Results are
`expressed as fold response over basallevels set arbitrarily at 1.0. (C),
`same as in (B), except that ERf expression vector was used. (D) and
`(E), same as in (B) and (C) respectively, except that pS2Luc reporter
`and HeLacells were used in transfections. (F), dose response of EM-
`
`
`
`x B
`
`aan
`
`ofeaxienalreeperpeeas oe4
`
`cS
`
`0 18-110 «<8 «7?
`BAR 1I1G oe
`sthagenial canbead logit}
`
`652 (filled squares) and ICI 182,780 (open squares) in the presence of
`10 nM E, on ERzoactivity in COS-1 cells
`transfected with
`vitA,ERETKLuc reporter and SRC-1 expression plasmid. The maxi-
`mal induction by E, alone wasset arbitrarily at 100%. (G), same as
`in (F) except that ERf expression vector was used [105].
`
`AstraZeneca Exhibit 2034 p. 7
`
`

`

`58
`
`F. Labrie et al. | Journal of Steroid Biochemistry and Molecular Biology 69 (1999) 51-84
`
`our transfection experiments, as indicated in Fig. 7. As
`observed previously [65,100],
`the addition of H-Ras
`contributed to increase the activity of ERa in the pre-
`sence of E,, with an even stronger response when H-
`Ras’ ’? was used (Fig. 7A). These inductions by both
`Ras forms were completely abolished with the addition
`of EM-652 in the medium, as with ICI 182,780,
`suggesting that EM-652 is effective in blocking the
`AF-1 activity of ERa«. The same experiment was also
`conducted on ERf where H-Ras and H-Ras‘!” aug-
`mented the E, response in a similar manner (Fig. 7B).
`Again, EM-652 and ICI 182,780 abolished the Ras
`effect on ERf in the presence of E. Interestingly, we
`observed a ligand independent effect of Ras on ERf
`basal activity where a 2-—3-fold induction occurred
`with H-Ras’’? (Fig. 7B). On the other hand, noeffect
`of Ras was seen on basal levels of ER. The Ras in-
`duction of unliganded ERf was blocked by EM-652
`and ICI 182,780 (data not shown). We were also inter-
`ested to test whether EM-652 wasefficient in blocking
`ER responsiveness on a natural promoter. The pS2
`promoter has been extensively studied in respect to its
`ERe mediated regulation [101]. We previously showed
`that ERf can also modulate transactivation of a repor-
`ter gene driven by the pS2 promoter in HeLa cells,
`and that the E, response was potentiated by H-Ras
`[65]. The effects of Ras on liganded ER« and f activi-
`ties are completely abrogated by EM-652 (data not
`shown). Dose response analyses were also performed
`to further evaluate the potency of EM-652 to inhibit
`the effect of Ras on ER activities in the presence of
`E,. EM-652 was
`slightly more effective than ICI
`182,780 in blocking H-Ras*’* inductions of ER and
`ER§, especially at lower concentrations (Fig. 7C and
`D).
`
`4.2. EM-652 blocks SRC-1 induced activity of both
`ERa and ERB
`
`The co-activator SRC-1 has been shown to interact
`with and promote the transcriptional activity of a
`number of nuclear receptors including ERa [85,102].
`Morerecently, we have demonstrated that SRC-1 also
`stimulates ERf activity through a direct
`interaction
`with its ligand-binding domain (LBD) where the AF-2
`domain resides [65]. We took advantage of this effect
`of SRC-1 to study whether EM-652 could block the
`E,-activated AF-2 function of ER« and ERP.
`Wefirst generated glutathione-S-transferase (GST)
`fusion proteins with the E and F domains of mERP
`(GST-mERf# EF) and domains D-F of mER« (GST-
`mERe DEF) for use in GST-pull down experiments
`(Fig. 8A). GST-mERf EF and GST-mER« DEF were
`expressed in £. coli, purified with GST-Sepharose and
`incubated with [*°S] methionine labeled SRC-1. As
`shown in Fig. 8A,
`the LBD of mER« interacted
`
`(lane 3)
`weakly with SRC-1 in the absence of E,
`whereas addition of E, caused an increase in inter-
`action between the two proteins (lane 4). Both EM-652
`(lane 5) and ICI 182,780 (lane 6) efficiently blocked the
`ligand-dependent SRC-1 interaction, with a stronger
`effect for EM-652. A similar
`inhibition of the E,-
`dependent interaction between SRC-1 and the LBD of
`ERf was also observed whereas ICI182,780 was less
`efficient
`(see Fig. 8A)
`lanes 7-10). We also demon-
`strate that the stimulatory effect of SRC-1 on the E,
`response of both ERs in COS-1 cells was completely
`abolished with the addition of EM-652 in the medium
`as did ICI 182,780 at the concentration used (Fig. 8B,
`C). Furthermore, as observed with Ras (see above),
`SRC-1, under
`the present experimental conditions,
`enhanced the basal activity of ER# but not that of
`ER« in the absence of ligand. This ligand-independent
`effect of SRC-1 on ERf was blocked by EM-652.
`Similar results were obtained using HeLa cells trans-
`fected with a pS2Luc reporter construct (Fig. 8D, E).
`Dose response analyses were also performed to
`further evaluate the potency of EM-652 to inhibit the
`potentiating effect of SRC-1 on ER activities in the
`presence of Ex. EM-652 was very effective in blocking
`SRC-1 potentiation of ligand-dependent ER« and ER
`transcriptional activities with apparent ICsg values of
`10~'° M and 10~° M, respectively (Fig. 8F and G).
`ICI 182,780 was less potent to inhibit the SRC-1 in-
`duction of ERe« and ERfactivities with an ICsq value
`of 10~° M for both receptors.
`The present study describes the molecular action of
`EM-652, the active metabolite of EM-800, on ER tran-
`scriptional functions. We present evidence that EM-
`800, and its active metabolite EM-652, act as pure es-
`trogen antagonists on ERa and ERf transcriptional
`activities. This pure antiestrogenic profile is of primary
`Importance in endocrine-based breast cancer therapy,
`since the objective, as mentioned earlier, is to develop
`a compound having both activities, while the widely
`available antiestrogen currently available, Tamoxifen,
`acts as a mixed agonist—antagonist on ER function
`and does not inhibit the AF-1 function. Besides a rela-
`tively good clinical record in inducing remission of
`ER-positive breast cancer and in post-surgical adju-
`vant therapy, resistance to tamoxifen, a phenomenon
`likely due to its intrinsic agonist properties, does occur
`and tumor progressi

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