throbber
Critical Reviews in Oncology/Hematology, 1993; 14: 173-188
`© 1993 Elsevier Scientific Publishers Ireland Ltd. All rights reserved. 1040-8428/93/$24.00
`
`173
`
`ONCHEM 00068
`
`Tamoxifen resistance in breast cancer
`
`Valerie J. Wiebe, C. Kent Osborne, Suzanne A.W. Fuqua and Michael W. DeGregorio
`Department of Medicine, Division of Oncology, The University of Texas Health Science Center at San Antonio, Texas, USA
`(Accepted 22 January 1993)
`
`
`
`Contents
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`,
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`174
`
`I
`
`II.
`
`III.
`
`Introduction .
`
`Mechanisms of anti-estrogen action .
`
`. .
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`175
`
`.
`
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`. .
`.
`.
`.
`. . .
`.
`. .
`.
`.
`Potential mechanisms of acquired tamoxifen resistance .
`. ..
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`. .
`.
`A. Altered levels of estrogen receptor .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`. .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1.
`Clinical clues .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`. .
`.
`.
`.
`.
`.
`. ..
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`B. Altered estrogen receptor .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`_
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1.
`ER variants .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.
`Tissue specific transcription factors .
`.
`.
`. .
`.
`.
`.
`.
`.
`Enhanced biologic mechanisms for circumvention of tamoxifen cytotoxicity .
`.
`.
`1. Growth factors
`2.
`Antiestrogen binding sites (AEBS) .
`.
`.
`.
`. .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`D. Decreased intracellular drug . . . .
`.
`.
`. .
`.
`. .
`.
`.
`. . .
`.
`. .
`.
`. . _ _
`. .
`. . . .
`.
`.
`. . .
`.
`. .
`.
`. .
`.
`.
`_
`,
`E.
`Tamoxifen metabolites and the development of resistance .
`.
`.
`.
`, ,
`.
`.
`.
`, . .
`,
`.
`. .
`. ..
`].
`Estrogenic metabolites .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`,
`.
`.
`.
`. .
`.
`.
`.
`.
`.
`. .
`. .
`.
`. ..
`2,
`Tamoxifen isomers .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`. .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`. ..
`F. Other contributing factors to anti-estrogen failure ,
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`. ..
`G. Circumvention
`
`
`C.
`
`[75
`176
`176
`176
`176
`177
`177
`I77
`179
`[79
`18]
`182
`182
`183
`183
`
`185
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`Summary .
`
`.
`
`_
`
`.
`
`.
`
`_
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`,
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`,
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`, .
`
`.
`
`185
`
`References
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`Tamoxifen (TAM) resistance is the underlying cause
`of treatment failure in many breast cancer patients
`receiving TAM. The mechanism(s) involved in TAM
`resistance are poorly understood. A variety of mech-
`anisms have been proposed but only limited evidence
`exists to substantiate them. Studies have now shown
`
`that in many patients TAM resistance is not related to
`the down regulation or loss of estrogen receptors (ER).
`Variant ER have been identified, but their significance
`clinically remains to be proven. Since breast cancer cells
`secrete several estrogen-regulated growth factors and
`growth inhibitors that may have autocrine or paracrine
`
`Correspondence to: Dr. Valerie Wiebe, Department of Medicine,
`Division of Oncology, The University of Texas Health Science Center.
`7703 Floyd Curl Drive, San Antonio, Texas, 78284-7884, USA.
`
`activity, altered growth factor production is another
`possible mechanism for TAM resistance. Tissue-specific
`transcription activating factors that may alter how the
`signal induced by TAM binding to the receptor is inter-
`preted by the cell also require further investigation. An
`increase in antiestrogen binding sites (AEBS), which
`could effectively partition TAM and reduce its concen-
`tration at the ER has also been proposed as a potential
`mechanism. Pharmacologic mechanisms, such as a shift
`in metabolism toward the accumulation of estrogenic
`metabolites, are supported by recent data demonstrating
`metabolite E and bisphenol
`in tumors from TAM-
`resistant patients. Furthermore, a decrease in tumor
`TAM accumulation and an altered metabolite profile
`have been reported in TAM-resistant breast
`tumors
`
`Astrazeneca Ex. 2019 p. 1
`Mylan Pharms. Inc. V. Astrazeneca AB IPR2016-01326
`
`

`
`174
`
`grown in nude mice. These and other studies suggest
`that TAM resistance may be multifactorial in nature,
`but definitive identification of mechanisms that are op-
`erative in clinical TAM resistance requires further study.
`
`I. Introduction
`
`Tamoxifen (TAM) is a nonsteroidal antiestrogen that
`was originally synthesized in 1966 as an antifertility drug
`[1]. However, in the 1970s TAM was noted to have ac-
`tivity in the treatment of metastatic breast cancer, and
`clinical trials began in the United States in 1974. In 1978
`TAM was primarily used to treat postmenopausal
`women with estrogen receptor positive, metastatic
`breast cancer. However, its clinical role has expanded to
`include all stages of the disease (Stage I and II) and both
`pre- and postmenopausal patients [2]. Response rates
`and duration of response in studies comparing TAM
`and oopherectomy in premenopausal patients are
`similar. Response rates to TAM increase with higher
`tumor ER levels. Overall, TAM prolongs both the
`
`disease-free and overall survival of women following
`primary surgery [3], and it induces tumor regression in
`about half of women with advanced estrogen receptor
`positive, metastatic breast cancer [4]. TAM has demon-
`strated efficacy in the prevention of contralateral breast
`cancer and it is also currently being evaluated for use as
`a chemopreventative agent in healthy women at high
`risk of breast cancer.
`
`Although approximately 50% of estrogen receptor-
`positive (ER+) tumors will
`respond to TAM, only
`60-75% of patients with metastatic breast cancer have
`estrogen receptor-positive tumors. Therefore only 35%
`of metastatic breast cancer patients actually benefit
`from TAM therapy [5]. In addition, all patients who
`initially respond to therapy will eventually develop
`acquired TAM resistance following prolonged adminis-
`tration.
`
`The development of acquired TAM resistance, where
`cell populations initially sensitive to TAM become
`insensitive, differs from innate resistance where cell
`populations are insensitive to TAM from the onset of
`
`Potential Mechanisms of Tamoxifen Inhibitory Activity
`
`TAM
`
`Mammary Cell
`
`Nucleus
`
`Cell Replication
`
`Autocrine
`
`Paracrine
`
`Shows the mechanism of estrogen (E2) binding to the estrogen receptor (ER) and the growth inhibitory effects of tamoxifen (TAM). TAM
`Fig. 1.
`competitively blocks the binding of E2 to the ER (1), it also binds to the antiestrogen binding sites (AEBS) (2). TAM blocks cells in GO/G1 (3)
`inhibiting cell replication. TAM may also decrease concentrations of TGF-oz, a growth factor that is stimulatory (4) and may increase levels of
`TGF-B, an inhibitory growth factor (5).
`
`Astrazeneca Ex. 2019 p. 2
`
`

`
`drug exposure. Unfortunately, the cellular and molecu-
`lar mechanisms underlying the development of acquired
`resistance to antiestrogens remains unclear. However, a
`variety of potential mechanisms have been suggested,
`and several reports concerning potential mechanisms for
`acquired TAM resistance have recently been published.
`
`II. Mechanism of antiestrogen action
`
`The mechanism(s) by which TAM inhibits tumor cell
`growth are believed to be mediated is primarily through
`interaction with ER (Fig. 1). Competitive antagonism of
`estrogen at
`the ER by TAM slows the growth of
`estrogen-dependent cancer cells by blocking them in the
`G0/Gl phase of the cell cycle [6]. Binding of TAM to the
`receptor is believed to form a complex that, when bound
`to estrogen—response elements, fails to trigger transcrip-
`tion of target genes. The resulting blockade is believed
`to be predominantly cytostatic in nature and may be
`reversed by the addition of estradiol. Whether TAM in-
`duces apoptosis or cell death is not yet clear.
`The antiestrogenic activity of TAM has also been
`evaluated in several species, and the biological effects of
`the drug appear to be dependent both on the species
`studied and the target
`tissue examined. In rats and
`humans, TAM has similar biological activity. In both
`species TAM has partial estrogen agonist effects on
`uterine tissues, but it is primarily considered an estrogen
`antagonist [7]. TAM’s weak estrogenic like effects have
`also been noted in postmenopausal patients in whom
`estrogenic effects were noted on gonadotrophin levels.
`plasma proteins and vaginal epithelium [7—9]. Whether
`the difference in antiestrogenic action is related to spe-
`cies specific or tissue specific metabolism of TAM. or to
`the presence of specific transcription factors that alter
`signal interpretation by the cell following interaction of
`the antiestrogen with the estrogen receptor is unknown.
`However, many other factors may also play a role in the
`cellular response to TAM.
`Several studies have now shown that cellular inhibi-
`
`tion by TAM may involve a complex series of events.
`Modulation of breast cancer cell growth by the differen-
`tial stimulation or inhibition of growth factor produc-
`tion from cells may also be involved in antiestrogen
`action. Recent evidence now suggests that estrogens
`may stimulate cell growth in part by inducing cells to
`synthesize growth factors and/or receptors. TAM on the
`other hand may act by inhibiting the estrogen-induced
`production of growth factors, while at the same time
`stimulating the production of growth inhibitory factors.
`At least one pathway may involve the stimulation of
`transforming growth factor-beta (TGF-B) production
`
`175
`
`by TAM. TGF-B has both growth inhibitory and stimu-
`latory effects. In stromal cells such as fibroblasts or
`endothelial cells, it stimulates cell growth. However, in
`most types of epithelial tumor cells,
`including breast
`tumors, it acts as a growth inhibitor [10]. Although the
`exact mechanism of TGF-B growth inhibitory effects are
`poorly understood, it does appear to inhibit tumor cell
`growth independent of the ER. A number of other
`growth factors are produced by breast cancer cells, and
`their expression is modified by estrogens and antiestro-
`gens. These include TGF-alpha, IGF-II, PDGF, and
`members of the EGF family [11]. However, the exact
`role that each of these growth factors plays in the induc-
`tion of cell growth by estrogen and the inhibition of cell
`growth by TAM remains to be elucidated.
`TAM has also been noted to bind to sites that are
`
`independent of the ER. These high affinity binding sites
`(Kd = 1 nM) are referred to as antiestrogen binding
`sites (AEBS) [12]. AEBS have been identified in several
`tissues with the highest concentrations noted in liver,
`uterus, ovaries, brain, and kidneys [l 3]. AEBS appear to
`be distinct from the ER and are only observed following
`prior treatment with estradiol [12].
`The affinity of antiestrogens for AEBS does not close-
`ly correlate with the biological potency of antiestrogens,
`suggesting that AEBS do not directly mediate antiestro-
`gen action [14,15]. Many studies have attempted to cor-
`relate binding of AEBS to other cellular events related
`to antiestrogen actions including protein kinase C in-
`hibition [16], calmodulin inhibition [17] and interac-
`tions with a variety of receptors, including histamine
`[18], dopamine [19] and muscarinic receptors [20]. In-
`deed much interest has been placed on the study of
`AEBS over the years; however, their true function and
`role in the antitumor efficacy of TAM remains to be
`established.
`
`III. Potential mechanisms
`resistance
`
`of
`
`acquired
`
`tamoxifen
`
`A variety of mechanisms has been implicated in the
`development of acquired resistance to TAM. However,
`little definitive data are available to support many of the
`proposed mechanisms. At one time it was assumed that
`drugs were responsible for inducing some biochemical
`modification in cells that resulted in acquired resistance
`to that drug. However, for many types of drug resistance
`the drug does not play a direct role in the development
`of resistance, but instead provides a strong selective
`pressure in favor of drug-resistant subclones. Drug-
`resistant subclones resulting from spontaneous muta-
`tions differ genetically from the original population. A
`
`Astrazeneca Ex. 2019 p. 3
`
`

`
`176
`
`deletion or modification of a specific enzyme, or altera-
`tion in some other cellular property in the genetically
`altered cell population may be responsible for the
`altered sensitivity of cells to the drug.
`Recent evidence derived from in vitro and in vivo
`
`studies suggests that TAM can stimulate cells to grow
`following prolonged exposure [2l—23]. Whether TAM
`is selecting a subclone of TAM-stimulated cells, whether
`the cells are altering TAM in such a way as to generate
`a stimulatory signal, or whether TAM is capable of
`inducing a genetic mutation that results in altered sensi-
`tivity to the drug remains to be established. One study
`has now shown that TAM can produce DNA adducts in
`the liver of rats suggesting that it may have genotoxic
`activity that could theoretically lead to mutations [24].
`Although the mechanisms underlying TAM resistance
`remain vague, a variety of recent studies suggest that
`multiple mechanisms may contribute to TAM resis-
`tance. Studies examining (A) altered level of ERs, (B) a
`decrease in ER affinity, (C) enhancement of cellular
`mechanisms
`for bypassing TAM cytotoxicity,
`(D)
`decreased cellular TAM concentration, (E) increased
`concentration of antagonizing metabolites and (F) a
`variety of other pathways, have contributed to our
`understanding of the possible mechanisms underlying
`TAM resistance.
`
`III-A. Altered levels of estrogen receptor
`
`III-A.1. Clinical clues
`
`ER expression is regulated to meet the demands of the
`cell. In the presence of high concentrations of estradiol,
`down regulation of receptors is believed to occur. The
`absence or loss of estrogen receptors could explain the
`development of hormonal independence or TAM resis-
`tance, particularly since ER-negative tumors rarely re-
`spond to TAM. However, clinical studies suggest that
`resistance to TAM is not always caused by selection of
`a hormone independent and/or ER-negative clone of
`tumor cells. Sequential biopsy studies have shown that
`apparent loss of ERs is common when the second biopsy
`is performed while the patient is taking TAM or within
`two months of stopping TAM presumably, due to recep-
`tor occupancy by the drug causing a false-negative
`ligand binding assay [25]. When the second biopsy is
`performed after two months, tumors frequently remain
`ER-positive, suggesting that there is no selection of a
`truly ER—negative clone. In a recent study of tumors
`from patients with TAM-resistance, an immunohisto-
`chemical technique was used to detect both bound and
`free receptors, ER was found in seven out of 13 tumors
`examined [26]. Maintenance of ER and/or PgR levels
`
`and responses to secondary hormonal therapies are not
`uncommon in patients with acquired TAM resistance
`[4,25]. Furthermore, in vitro studies suggest that follow-
`ing the selection of antiestrogen-resistance cells, many
`resistant cell
`lines may remain sensitive to estrogens
`[27,28].
`Thus, ER loss may play a role in acquired TAM resis-
`tance in some patients, but it cannot account for the
`resistance noted in the majority of patients. In addition,
`clinical evidence suggests that patients that
`initially
`respond to TAM, but who later develop resistance,
`frequently respond to secondary hormonal treatment,
`suggesting that the development of resistance to anti-
`estrogens does not confer resistance to other hormonal
`agents [4].
`
`III-B. Altered estrogen receptor
`
`Protein structure modifications leading to altered affi-
`nity of the ER for TAM is a plausible resistance mecha-
`nism. Site—specific mutations,
`including nonsense or
`frameshift mutations in the structural gene coding for
`the ER may potentially result in various types of func-
`tionally abnormal
`receptors. These mutations may
`render the ER entirely nonfunctional; thus, the tumor
`would appear clinically as if it were ER-negative. Alter-
`natively, if mutations result in amino acid substitutions
`in important domains of the receptor, then the result
`may be the generation of ER species which are func-
`tionally active, but which exhibit altered specilicities for
`estrogens and antiestrogens.
`
`III—B.1. ER variants
`Much is known about the structure and function of
`
`the ER [29]. The ER contains discrete domains involved
`in hormone binding, DNA binding, and subsequent
`activation of estrogen-responsive genes. Human ERs
`have now been shown to contain five distinct functional
`
`domains A/B, C, D, E and F [30]. Although there is
`some overlap between domains, regions E and D appear
`to primarily involve the horrnone-binding and dimeriza-
`tion domains [30,31]. Region C is the DNA binding do-
`main, and the A/B and E regions contain the two
`transcription activating functions.
`The presence of these discrete functional domains has
`led investigators
`to examine alterations in TAM-
`resistant, or hormone-independent model
`systems.
`Many earlier studies failed to show differences in the ER
`in in vitro systems. For example, Mullick and Chambon
`[32] used two independently isolated TAM-resistant,
`ER-positive breast cancer cell lines, LY2 and T47D, to
`demonstrate that the ER was still functional in these
`
`Astrazeneca Ex. 2019 p. 4
`
`

`
`cells in spite of their hormone—insensitive growth. The
`ER was shown to be wild-type using RNase protection
`assays and by its ability to stimulate estrogen—responsive
`reporter constructs in these cells. However, neither of
`these assays rule out the possibility that mutated ER
`species may be present along with wild—type ER, and
`that
`these mutated forms may contribute to the
`tamoxifen-resistant phenotype. Direct sequence analysis
`of the ER from these cells may be required to definitive-
`ly answer this question.
`[33] have examined the
`In fact, Graham et al.
`presence of mutated ERs by cloning and sequence
`analysis in T47D cells that have been maintained in their
`laboratory. Several different ER mutations were
`detected in complementary DNAs prepared from these
`cells, including frameshift mutations within the DNA
`and the hormone binding domains of the receptor. If
`expressed, these mutated ER species could be defective
`in activity, and could contribute to the hormone-
`independent phenotype of this T47D subline. Of impor-
`tance from this study is
`that highly sophisticated
`technologies were required to identify ER mutations
`present
`in cells which were heterogenous
`for ER
`expression.
`Raam et al. [34] have utilized immunohistochemical
`procedures to demonstrate the presence of ERs deficient
`in nuclear binding in ER-positive tumors. Interestingly,
`patients with either constitutive nuclear binding, or
`those with ER which could not bind nuclei, were refrac-
`tory to hormone therapy. This encouraging result, that
`tumors may contain defective ERs, has been recently
`substantiated by studies using larger series of human
`breast tumor specimens [35,36]. Even though endocrine
`response data was not available on these later studies,
`both studies suggest that truncated forms of the ER,
`which fail to bind DNA in gel-retardation assays, may
`be present in tumors. Of note,
`is that DNA binding-
`deficient ER was most prevalent in tumors with minimal
`PgR expression, agreeing with the commonly-held doc-
`trine that PgR expression closely correlates with an in-
`tact ER response pathway. It will be interesting to apply
`the gel-retardation methodology to tumors with clinical
`response data to determine whether ERs defective in
`DNA binding may contribute to TAM failure.
`Murphy et al. [37] have identified abnormal sized ER
`mRNAs by Northern hybridization, and they have
`recently cloned these altered ERs from human breast
`tumors. Three different ER mRNAs have been iden-
`
`tified, all of which diverge from the known ER sequence
`at exon/intron borders. At the point of divergence, non-
`ER sequences have been inserted. These insertions are
`either unknown or are homologous to long interspersed
`
`177
`
`repetitive LINE-1 sequences. These three altered ERs
`are all missing the hormone binding domain of the
`receptor
`in addition to containing unique non-ER
`segments. One of the mutated ERs, designated clone 4,
`is widely expressed in breast tumor samples.(Dozlaw,
`unpublished results). Although clone 4 ER was devoid
`of transcriptional activity in in vitro assays, its presence
`may be functionally significant in tumors. Experiments
`are currently underway to examine its distribution in
`breast tumors where clinical information is available
`
`(Murphy and Fuqua, unpublished observations).
`The progression of steroid sensitive cells to steroid in-
`sensitivity was evaluated by Darbe and King [38]. In
`their study they demonstrate that clones of steroid-
`responsive cells can give rise to a population of
`unresponsive cells in a series of phenotypic changes
`which are brought about solely by long-term withdraw]
`of the hormone. In a further study they report that
`transfection of a steroid inducible gene into unrespon-
`sive cells (S1 15) results in that gene being fully inducible
`by steroids. Therefore, the machinery for steroid respon-
`siveness, including receptors, appears to be intact. They
`suggest that the process appears to be independent of
`the loss of steroid receptor function [39]. This was fur-
`ther substantiated by Clarke et al. who examined the
`progression of human breast cancer cells from hormone-
`dependent to hormone-independent growth in vitro and
`in vivo. They report that an ovarian—independent, but
`honnone-responsive phenotype may occur early in the
`natural progression to hormone—independence, but that
`altered hormone receptor expression may be a late event
`in the acquisition of this phenotype [40].
`ER variants have also been isolated from a variety of
`ER-positive,
`and
`supposedly ER-negative
`breast
`tumors, using sensitive RNA-directed polymerase chain
`reaction (PCR) methodologies [41,42]. Tumors which
`are ER-negative, but PgR-positive often express high
`levels of a variant ER lacking exon 5 of the hormone
`binding domain of the receptor. This deletion results in
`the production of a variant ER truncated within the hor-
`mone binding domain, and as such, is unable to bind es-
`trogen. However, the receptor appears to bind DNA
`and is constitutive for activation of estrogen-responsive
`genes. When the exon 5 ER variant is coexpressed with
`wild-type receptor
`in MCF—7 cells, TAM-resistant
`growth is conferred to these cells. Thus, overexpression
`of the variant, even in the presence of wild-type recep-
`tor, may contribute to TAM resistance. Furthermore,
`ER-positive tumors which express wild-type ER often
`coexpress the exon 5 ER deletion variant. Tumors with
`this variant may escape the normal growth dependence
`of estrogens, and subclones may be selected for under
`
`Astrazeneca Ex. 2019 p. 5
`
`

`
`[78
`
`conditions where TAM is present which would inhibit
`only the wild-type receptor present in these cells. The
`heterogenous expression of variant ERs within a tumor
`may contribute to the emergence of TAM resistance
`during treatment. Thus, the selection of TAM-resistant
`ER variants may be clinically significant with TAM ac-
`tually providing the selective pressure for their eventual
`outgrowth. These are testable hypotheses, and we anx-
`iously await the answers.
`
`III-B.2. Tissue specific transcription factors
`TAM resistance may also involve alterations in tissue-
`specific transcription activating factors that might not
`alter TAM or estrogen binding, but could alter how the
`signal induced by TAM binding to the receptor is inter-
`preted by the cell (antiestrogenic versus estrogertic).
`Transcriptional activation functions (TAF) have been
`associated with certain regions of the estrogen receptor
`(regions E and A/B). At least two TAFS (TAF-1 and
`TAF-2) have been associated with antiestrogen action.
`TAF-2, from region E,
`is
`located in the hormone-
`binding domain and is reported to be stimulated by es-
`trogens and inhibited by antiestrogens.
`In contrast,
`TAF-1 is believed to have constitutive activity in the ab-
`sence of estrogen binding in some cells [43]. TAF—1’s
`role in stimulating transcription may therefore be de-
`pendent on the cell type, promoter and the presence of
`other TAFS. Mutations in either TAF-1 or TAF-2 do-
`
`mains could potentially modify the cellular response to
`TAM leading to an estrogenic rather
`than an an-
`tiestrogenic effect. To date there is little evidence in sup-
`port of this mechanism. In preliminary sequence studies
`performed in our laboratory, no mutations in either of
`the two transcription—activating domains or
`in the
`hormone-binding domain of the ER in a TAM-resistant
`MCF-7 in vivo tumor model were identified (unpublish-
`ed observations).
`
`III-C. Enhanced biologic mechanisms for circumvention
`of tamoxifen cytotoxicity
`
`The adaptation of estrogen dependent cells to survive
`in the presence of estrogen antagonists may also be re-
`lated
`to
`several
`physiologic
`and
`biochemical
`mechanisms that help the cell
`to circumvent
`the in-
`hibitory effects of TAM.
`
`III—C.I. Growth factors
`Recent research has demonstrated that breast cancer
`
`cells and stromal cells can produce and secrete a variety
`of polypeptide growth factors and growth inhibitors,
`that by autocrine and paracrine mechanisms, may con-
`
`tribute to tumor growth regulation [1l,44,45]. Expres-
`sion of several of these factors is regulated by estrogens
`and antiestrogens, suggesting the hypothesis that they
`may mediate, at least in part, the growth effects of these
`agents. As discussed earlier, one of these factors is TGF-
`6, a family of growth inhibitors that are produced by
`breast cancer cells [46,47]. A consequence of estrogen
`treatment is a reduction in the expression of TGF-6
`which might then result in enhanced growth. TAM, by
`binding to ER, blocks the effect of estrogen and induces
`the production of the growth inhibitor. Interestingly,
`TAM has also been reported to stimulate TGF-/3 pro-
`duction by breast
`tumor
`fibroblasts by unknown
`mechanisms [48]. If confirmed, these data could explain
`the infrequent response of an ER—negative tumor with
`TAM treatment.
`
`However, the role of TGF-3 in mediating the effects
`of estrogens and antiestrogens is not yet clear. High-
`passage MCF—7 breast cancer cells, that have very low
`levels of TGF-B receptors and are not
`inhibited by
`exogenous TGF-[3, are still
`inhibited by TAM [47].
`Exogenous TGF-[3 injected into nude mice in sufficient
`concentrations to cause significant
`toxicity had little
`effect on tumor growth [44,45]. Furthermore, recent
`data have shown that when a mixture of MCF—7 (ER-
`positive) and MDA MB-231
`(ER-negative) breast
`cancer cells are injected into nude mice at a ratio of
`100021, tumor growth is not inhibited by TAM, and ER-
`negative tumors result [49]. Therefore, while TAM may
`be able to stimulate the production of TGF18 from ER-
`positive cells. it does not appear to do so in sufficient
`quantities to inhibit proliferation of the ER-negative cell
`population, at least in this model.
`Thus, the effects of TGF-B on breast cancer cells in
`vivo remain speculative. If TGF-B is eventually shown
`to be important in estrogen/antiestrogen action, then it
`could play a role in TAM resistance, which could
`theoretically result from the failure of the tumor or
`stromal cells to secrete active TGF-B in response to
`TAM.
`
`Other stimulatory growth factors could also poten-
`tially play a role in TAM resistance. TGF-oz, IGF-II,
`PDGF, cathepsin D, and other growth factors have
`been shown to be produced and secreted by breast
`cancer cells [1l,44,45]. Secretion by ER-negative cells is
`constitutive, while that by ER-positive cells is regulated
`by estrogen. TAM treatment inhibits the production of
`these autocrine growth factors, perhaps contributing to
`its growth suppressive effects. The acquired ability of
`the cells to express these growth factors constitutively,
`despite TAM treatment, could theoretically result
`in
`TAM resistance. In vitro studies have, in fact, shown
`
`Astrazeneca Ex. 2019 p. 6
`
`

`
`that certain growth factors can reverse the inhibitory ef-
`fects of TAM [50,51]. At this time, however, there is no
`supportive evidence from experimental models or from
`patients to suggest that altered tumor production of
`autocrine growth factors or growth inhibitors is a cause
`of TAM resistance. This possibility, nevertheless,
`deserves additional investigation.
`
`III-C.2. Antiestrogen binding sites (AEBS)
`One theoretical mechanism by which breast cancer
`cells could potentially become resistant
`to the an-
`tiestrogenic effects of TAM is increased binding of drug
`to intracellular sites that do not participate in its anti-
`tumor actions, thereby reducing TAM concentrations at
`the ER. The fact that antiestrogen resistance can occur
`in the presence of functional ER has lead to the hypo-
`thesis that antiestrogen action is mediated independent-
`ly of ER by specific AEBS [52]. In the presence of
`antiestrogen, but not estrogens, AEBS are competitively
`reduced [l3,53—56]. However, the affinity of antiestro-
`gens for AEBS does not appear to closely correlate to
`the biopotency of specific antiestrogens [l5,57,58], and
`AEBS specific ligands with no affinity for the ER do not
`demonstrate functional antagonism [59]. Furthermore,
`AEBS remain expressed in antiestrogen-resistant tumor
`cell variants [60]. Thus,
`the role of AEBS in TAM-
`
`induced tumor growth inhibition is questionable.
`Although in theory increased binding of TAM to
`AEBS by either an increase in AEBS concentration or
`increased affinity for TAM could reduce available
`TAM, only limited evidence has been found supporting
`this theory. Pavlik et al. examined the possibility that
`AEBS may prevent the antiestrogen from interacting
`with ERs [52]. In their study, antiestrogen binding was
`compared in uterine preparations, where ER activity ex-
`ceeded AEBS, and in liver preparations, where AEBS
`binding predominated. Their results suggest that when
`AEBS activity predominates, TAM was virtually all
`bound to AEBS with little remaining available to
`associate with the ER. Furthermore, they also noted
`that the ratio of AEBS: ER was three times greater in
`antiestrogen-resistant MCF-7 cells
`(LY-2)
`than in
`antiestrogen—sensitive wild type MCF-7 cells. In addi-
`tion, examination of 128 human breast carcinomas
`
`showed that AEBS activity exceeded ER activity, sug-
`gesting that AEBS could partition TAM from ER in
`human breast tumors. They conclude that AEBS may
`provide a mechanism of antiestrogen-resistance whereby
`cells may lose their sensitivity to antiestrogens but retain
`their sensitivity to estrogens.
`However, other data do not support this theory. Two
`TAM-resistant MCF-7 clones (R3 and R27) have
`
`179
`
`unaltered levels of AEBS [61,62], and one resistant clone
`has been reported in which no AEBS could be identified
`[63].
`
`III-D. Decreased intracellular drug
`
`A decrease in intracellular drug concentration is one
`mechanism by which cells become resistant to drugs.
`Multidrug resistance is one form of resistance whereby
`a decreased intracellular drug concentration has been
`associated with an active efflux of drug from the cell.
`Drug resistance due to overexpression of P-glycoprotein
`is a well characterized form of acquired multidrug resis-
`tance (MDR) to natural product antineoplastics such as
`doxorubicin.
`In this form of resistance tumor cells
`
`develop resistance to a broad range of structurally
`unrelated drugs [64]. The mechanism underlying MDR
`is believed to involve a 170 000-Da cell membrane pro-
`tein, termed P-glycoprotein, which is overexpressed in
`MDR cells and appears to function as a drug efflux-
`pump [65]. Classical MDR is associated with the
`overexpression of the MDR-l gene that codes for the
`plasma membrane P-glycoprotein (p170). Overexpres-
`sion of the MDR-1 gene is associated with decreased
`cellular accumulation of drug due to the active energy-
`dependent efflux mechanism.
`Efforts focused on the pharmacologic modulation of
`P-glycoprotein have identified several drugs that are ef-
`fective in reverting MDR in vitro. These include calcium
`channel blockers, chlorpromazine, cyclosporin, steroids
`and calmodulin antagonists [66,67]. Interestingly. anti-
`estrogens, including both TAM and toremifene, have
`also been shown to reverse MDR in vitro. and
`toremifene is now undergoing clinical evaluation for this
`indication [68,69].
`Although no similar efflux pump has been identified
`in TAM resistance, several recent studies

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