throbber
Arum. Rev. Pharmacal. Toxicol. 1995. 35:195—21 I
`Copyright 0 1995 by Annual Review; Inc. All rights reserved
`
`‘D QENVNIEVCE Further
`Qutck links to onllne content
`
`Annu.Rev.Pharrnacol.Toxicol.1995.35:195-211.DownloadedfromwwwannualreviewsorgbyJohnsHopkins
`
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`TAMOXIFEN: Toxicities and
`
`Drug Resistance During the
`Treatment and Prevention of
`
`Breast Cancer
`
`V. Craig Jordan
`Robert H. Lurie Cancer Center, Northwestern University Medical School, Chicago,
`Illinois 60611
`
`KEY WORDS:
`
`endometrial cancer. rat liver cancer. antiestrogens. raloxifene, ICI 182,780
`
`ABSTRACT
`
`Tamoxifen, a nonsteroidal antiestrogen, is the endocrine therapy of choice for
`all stages of breast cancer. There are six million women-years of experience
`with tamoxifen, and the drug has produced survival advantages in node-posi-
`tive and node-negative patients who have had 2—5 years of adjuvant tamoxifen
`therapy. A low incidence of side effects has been reported with tamoxifen,
`resulting in the proposal to use the antiestrogen as a preventive agent for breast
`cancer. Three separate clinical trials are currently under way—in the United
`States, Italy, and the United Kingdom. Current concerns about tamoxifen are
`the development of rat liver tumors during long-term treatment and an in-
`creased incidence of endometrial carcinomas observed in patients. Another
`concern is the development of drug resistance to long-term tamoxifen therapy.
`There is increased interest in both determining the mechanism of drug resis-
`tance and evaluating new antiestrogens that may be more beneficial as a
`preventive, as an adjuvant therapy, or for the treatment of advanced breast
`cancer.
`
`INTRODUCTION
`
`Tamoxifen is a nonsteroidal antiestrogen (1—4) that exhibits antitumor prop-
`erties in laboratory animals (5—9). Although many compounds were investi-
`
`195
`
`0362-1642/95/0415-0195$05.00
`
`AstraZeneca Exhibit 2017 p. 1
`InnoPharma Licensing LLC V. AstraZeneca AB
`IPR2017-00905
`
`

`

`196
`
`JORDAN
`
`gated in the 1960s and 70s (10), only tamoxifen was successfully developed
`in the laboratory for the treatment of breast cancer (10-12). The initial focus
`of anti-breast cancer drug development in the 1970s was as a palliative therapy
`for advanced breast cancer. However, adjuvant therapy and the exploration of
`long-term tamoxifen therapy (13, 14) has revolutionized breast cancer treat-
`ment. An overview analysis of randomized clinical trials of adjuvant tamoxifen
`therapy demonstrates that
`long-term (i.e. more than one year) adjuvant
`tamoxifen is an appropriate strategy to control the recurrence of both node-
`positive and node-negative breast cancer (15). Tamoxifen confers a survival
`advantage to those women with breast cancer who are treated for more than
`two years. Tamoxifen is the hormonal treatment of choice for all stages of
`breast cancer.
`The use of tamoxifen as a treatment for breast cancer has over six million
`
`women-years of experience. The drug has a low reported incidence of serious
`side effects (16), and five years of therapy is commonplace (17).
`The success of tamoxifen as a treatment for breast cancer has fueled interest
`
`in the drug as a preventive agent. Estrogen is known to promote the develop-
`ment of breast cancer, so it is only natural that an antiestrogenic drug that has
`been extensively clinically tested would be the leading candidate for evalua-
`tion. The pharmacology of tamoxifen is complex (18, 19), but the scientific
`rationale for testing tamoxifen has merit. An important component of the drug’s
`pharmacology is the target-site specificity of tamoxifen; the drug can act as
`an antitumor agent (probably as an antiestrogen) in the breast but can also be
`estrogenic at physiological sites to prevent bone loss and decrease circulating
`cholesterol.
`
`Animal studies demonstrate that tamoxifen prevents mammary carcinogen-
`esis (5, 7, 20—22), and clinical studies show that adjuvant tamoxifen therapy
`decreases the incidence of second primary breast cancers by 40% (15). Post-
`menopausal bone density is maintained by tamoxifen treatment (23-25), which
`could ultimately lead to the prevention of osteoporosis. Tamoxifen also de-
`creases low-density lipoprotein cholesterol levels in postmenopausal women
`(26-28). This positive property of tamoxifen may be responsible for the de-
`crease in hospital visits for the treatment of any cardiac condition (29) and the
`significant decrease in fatal myocardial infarction for women treated for five
`years with tamoxifen (30, 31). These data provide a scientific basis for pla-
`cebo-controlled clinical trials to test tamoxifen’s ability to prevent breast
`cancer.
`
`PREVENTION STUDIES
`
`Unlike the laboratory models of mammary tumorigenesis, where all animals
`develop tumors and the efficacy of tamoxifen is readily demonstrated, targeting
`
`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`AstraZeneca Exhibit 2017 p. 2
`
`

`

`TAMOXIFEN AND BREAST CANCER
`
`197
`
`the appropriate population at risk for breast cancer is difficult. Numerous risk
`factors have been identified, and these have been reviewed elsewhere (32).
`However, because the incidence of breast cancer is small in the general pop-
`ulation, women volunteers with a high-risk profile must be recruited. It is
`essential to design a double-blind, placebo-controlled trial, but the large num-
`bers of volunteers required and the long time period necessary to obtain a
`statistically significant result mandates data management, compliance moni-
`toring, and an enormous clinical trials effort.
`Three clinical trials are currently recruiting and following volunteers to test
`tamoxifen’s ability to prevent breast cancer. The first trial was begun at the
`Royal Marsden Hospital in 1986 (33, 34). The initial goal was to recruit 2000
`women as a Vanguard study and monitor the progress of the volunteers.
`Healthy women aged 30—70 are eligible provided they have a family history
`of breast cancer on the maternal side, with at least one first-degree relative
`(sister, mother, daughter) under the age of 45 years who has developed breast
`cancer or bilateral breast cancer or with a first-degree relative and at least one
`other maternal relative affected. The women are randomly assigned to receive
`20 mg of tamoxifen or a placebo daily for at least eight years. At five years
`(by June 1993) compliance for the 2012 women assigned to the tamoxifen arm
`of the study was greater than 70% (35).
`There is a significant increase in hot flashes (34% vs 20%), vaginal discharge
`(16% vs 4%), and menstrual irregularities (14% vs 9%) for tamoxifen- vs
`placebo-treated women. Safety monitoring shows no obvious effects on radial
`bone mineral density, but fibrogen, antithrombin III, and cholesterol levels
`decrease out to five years in the tamoxifen-treated women.
`Most importantly, the Marsden study demonstrates an increased incidence
`of uterine fibroids and benign ovarian cysts as a result of tamoxifen treatment.
`An in-depth study (36) of the postmenopausal women demonstrated that ta-
`moxifen causes potentially malignant changes in the endometrium, but trans-
`vaginal ultrasonography can be used to identify those women who should be
`monitored. These findings resulted in approval by the Department of Health
`(July 1993) to recruit an additional 15,000 women volunteers from sites around
`the United Kingdom. Recruitment of additional volunteers has also been con-
`ducted for more than a year in Australia.
`The second prevention trial began recruiting volunteers from throughout
`North America in May 1992. The study, funded by the National Cancer
`Institute, will recruit 16,000 women who will be randomly assigned to be
`treated with tamoxifen (20 mg daily) or a placebo for five years. Those eligible
`for entry into the study include any woman over the age of 60 or women
`between the ages of 35 and 59 years whose five-year risk of developing breast
`cancer, as predicted by the Gail model (37), equals that of a 60-year-old
`woman. Any woman over 35 years of age, with a diagnosis of lobular carci-
`
`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`AstraZeneca Exhibit 2017 p. 3
`
`

`

`198
`
`JORDAN
`
`noma in situ (LCIS) treated with biopsy alone, is eligible for entry. In the
`absence of LCIS, the risk factors for entry vary with age, so a 35-year-old
`woman must have a relative risk of at least 5, while a 45-year-old woman’s
`relative risk must be 1.8 to be eligible for entry.
`Seven thousand women were recruited in the first year. Pretrial concerns
`that younger women at risk would not volunteer for the trial are not substan-
`tiated by the population distribution. About one third of the volunteers are
`35—50 years old, with relative risks ranging on average between 10 for the
`youngest participants to 4 for the 50-year-olds.
`By December 1993, 11,000 women had been recruited to the prevention
`trial. Administrative concerns about the development of uterine carcinoma
`during tamoxifen treatment halted recruitment to National Surgical Adjuvant
`Breast Project (NSABP) trials for several months during 1994, but recruitment
`was reinitiated in June 1994 after the Food and Drug Administration had
`reviewed the concerns.
`
`The final tamoxifen prevention trial was initiated in Italy (38) by the Euro-
`pean Institute of Oncology and the Milan Cancer Institute. Up to 20,000
`volunteers who are over the age of 45 but who have already had a hysterectomy
`will be recruited. The aim of these restrictions is to avoid the complications
`of both pregnancy and endometrial cancer. Volunteers are being randomly
`assigned to tamoxifen (20 mg daily) or a placebo for five years. There were
`more than 3000 volunteers recruited by July 1993.
`For the first time, the clinical trials community is evaluating a therapy to
`prevent breast cancer. Although the majority of women recruited to the trials
`will not develop breast cancer, they will experience symptoms and side effects
`from tamoxifen. The evaluation of the toxicity of tamoxifen in the trials is
`extremely important, not only to determine the therapeutic value of the inter-
`vention but also to assess whether compliance can be maintained by the study
`population. Extra attention is being paid to chronic toxicities.
`
`TOXICITIES OF TAMOXIFEN
`
`Considerable concern has been expressed about the potential toxic effects of
`tamoxifen that could become critical in any evaluation of the drug given to
`women without breast cancer. These concerns are listed in Table 1 and have
`
`been the subject of a recent commentary (39). Ocular problems and the small
`increase in thromboembolic disease has been adequately reviewed (16), but
`the potential of tamoxifen to be carcinogenic is a serious risk.
`In the laboratory, tamoxifen can stimulate the growth of human endometrial
`carcinoma but can block the growth of a breast tumor transplanted in the same
`immune-deficient mouse (40). This possibility was demonstrated in patients
`when a 40% decrease in second primary breast cancers but a threefold increase
`
`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`AstraZeneca Exhibit 2017 p. 4
`
`

`

`TAMOXIFEN AND BREAST CANCER
`
`199
`
`Table 1 Potential long-term toxicities from tamoxifen therapy
`
`Ocular problems
`Thromboembolic disorders
`Endometrial cancers
`Liver cancer
`
`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`in endometrial cancer was observed in an adjuvant clinical trial of tamoxifen
`(41). Seventeen endometrial cancers were reported for the 1000 patients taking
`tamoxifen after eight years of evaluation. The rate of detection is two per
`thousand per year (42). A similar rate is reported by the NSABP (43). However,
`the NSABP study reports 6 deaths out of the 23 patients who developed
`endometrial carcinoma over the 6 years of evaluation for the 2639 women
`(43). The causes of death and the association with the duration of tamoxifen
`treatment are shown in Table 2. It is clear that women must be monitored for
`
`the development of endometrial carcinoma, but perhaps most importantly,
`patients must be screened before therapy to ensure that preexisting endometrial
`carcinoma is not present.
`Investigators are currently interested in determining whether tamoxifen pro-
`duces a higher-grade, more aggressive endometrial carcinoma. Initial reports
`from the Yale—New Haven tumor registry suggested that women were “at risk
`for high-grade endometrial cancers that have a poor prognosis" (44). Current
`comparisons of histological grade and stage in patients treated with tamoxifen
`(42, 43, 45) demonstrate the same proportions noted for the general population.
`
`Table 2 Patients randomly assigned to the tamoxifen arms of NSABP
`BM who died of EC“
`
`Patient
`
`Age
`
`Time on
`tamoxifen
`(months)
`
`Diagnosis of EC
`after tamoxifen
`(months)
`
`Cause of
`death
`
`1
`
`2
`3
`4
`5
`6
`
`66
`
`68
`63
`58
`54
`68
`
`—
`
`EC
`
`Never took
`tamoxifen
`CV diseaseVD
`0
`5
`EC
`0
`9
`EC
`73
`22
`EC
`23
`42
`Pulmonary
`0
`65
`embolism
`
`“Endometrial cancer
`“Cardiovascular disease
`
`AstraZeneca Exhibit 2017 p. 5
`
`

`

`200
`
`JORDAN
`
`Nevertheless, tamoxifen does not retard the development of endometrial car-
`cinoma, and clonal selection may result in premature changes in pathology.
`These findings contrast with the effects of estrogens that only promote growth.
`In contrast, the concern about hepatocellular carcinoma is based upon lab-
`oratory studies alone. Numerous reports show that large doses of tamoxifen
`produce liver tumors in rats (46-48). Tamoxifen produces DNA adducts in rat
`liver (48—50) and protein adducts in vitro (51). It is hypothesized that tamoxifen
`can become metabolically activated through selective hydroxylation to form
`an unstable alkylating species (52). Nevertheless, even though DNA adducts
`can be formed by human liver microsomes in vitro (53), no practical demon-
`stration of DNA adduct formation has occured in humans. Indeed, there have
`
`been no reports of hepatocellularcarcinoma in women taking 20 mg tamoxifen
`daily and only two reports of hepatocellular carcinoma in women who took
`40 mg daily (41). Part of the problem is that the tumor is so rare—5 per 100,000
`of the population—that even a tenfold rise would be difficult to detect.
`The striking differences observed for the toxicology for tamoxifen in the rat
`may be species and dose related. Tamoxifen is used therapeutically at a dose
`of 250 ug/kg in both humans and rats (14). In contrast, rats are given 12 mg/kg
`of tamoxifen for half their lifetime to produce livertumors. This regimen (48)
`is approximately 40 times the human therapeutic dose given for about 8 times
`as long as the relatiVe human duration, which is five years or 6% of a woman’s
`life. The excessive doses of tamoxifen given to the rat could be overwhelming
`the capacity of the liver to such a degree that the rat must invoke unique
`metabolic routes to cope with the overdosing schedule. The equivalent exper-
`iment in humans would be a woman taking 800 mg tamoxifen daily for 40
`years.
`
`The relationship of liver carcinogenesis and tamoxifen is undoubtedly an
`important area of toxicology, not only to protect the treatment population but
`also to determine the relevance of certain animal models to human disease
`processes.
`
`Overall, the broad use of tamoxifen, both as a treatment and as a preventive
`agent in clinical trials has necessitated increased vigilance by the clinical
`community to identify untoward side effects as rapidly as possible. Tamoxifen
`is an effective therapy for breast cancer, and it has become an important
`cornerstone of treatment strategies. However, drug resistance is the inevitable
`result of any long-term therapy. The discovery of the processes of growth
`deregulation will provide additional therapeutic opportunities for the future.
`
`DRUG RESISTANCE
`
`The potential molecular mechanisms of resistance to antiestrogen therapy have
`recently been surveyed (54), but several new developments deserve comment.
`
`Annu.ReV.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`AstraZeneca Exhibit 2017 p. 6
`
`

`

`TAMOXIFEN AND BREAST CANCER
`
`201
`
`LOSS OF
`
`!_ RECEPTOR
`
`
`
`
`H UTATED
`"Begum;
`
`AmET'
`Ehi'hglbUCTION
`'
`H"
`
`
`lie—AL.—
`METABOLISM
`'—'
`
`
`
`
`
`TAMOXIFEN
`
`METABOLITES
`
`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`INCREASED
`ESTRADI 0L
`
`Figure I The potential mechanisms of drug resistance against tamoxifen in human breast cancers.
`AEBP = antiestrogen binding protein. ER = estrogen receptor.
`
`Three major areas of concern are increased estrogen levels observed in pre-
`menopausal women during tamoxifen therapy, local tumor metabolism to
`unstable compounds that stimulate tumor growth, and the isolation of mutant
`estrogen receptors from tumors stimulated to grow by tamoxifen. These pos-
`sibilities are illustrated in Figure 1.
`
`High Estrogen Levels
`
`Tamoxifen causes an elevation in circulating estrogen levels in premenopausal
`patients (55—58), and a high estrogen environment could reverse the antitumor
`actions of tamoxifen. Tamoxifen is most effective in vitro in a low—estrogen
`environment; however, in vivo, tamoxifen is converted to a range of anti-
`estrogens with high affinity for the receptor. The reversal of the actions of
`tamoxifen as an antitumor agent is complicated. Studies in athymic mice
`demonstrate that high levels of circulating estradiol (>1600 pg/ml) will par-
`tially reverse the growth inhibitory effects of low levels (40 ng/ml) of circu-
`lating tamoxifen (59). Clinical experience demonstrates that tamoxifen alone
`is effective in the control of Stage I and IV breast cancer in premenopausal
`patients (15, 60—62). Nevertheless, if patients with Stage IV disease initially
`respond and then fail treatment, there is a 30% probability of a second response
`to oophorectomy (63).
`
`AstraZeneca Exhibit 2017 p. 7
`
`

`

`202
`
`JORDAN
`
`Local Metabolism
`
`The pharmacokinetics and metabolism of tamoxifen have been extensively
`studied in patients (64—66), and there is no evidence that high levels of estro-
`
`genic metabolites are produced during long-term therapy. However, the tumor
`cells or the stromal component could locally metaboliZe tamoxifen to potent
`estrogens that could stimulate tumor growth. In the laboratory long-term tam-
`oxifen treatment will eventually cause the growth of MCF-7 breast cancer cells
`transplanted into athymic mice (67, 68). The tumors are estrogenoreceptor
`positive and grow in either athymic rats or athymic mice in response to either
`estradiol or tamoxifen (68, 69). Pure antiestrogens will block tamoxifen-stim-
`ulated growth; therefore, tamoxifen must be converted to.estrogens locally to
`stimulate growth through the estrogen receptor (70).
`Tamoxifen is metabolized to 4-hydroxytamoxifen in the mouse (71). This
`metabolite is a potent antiestrogen (72) that is known to have antitumor activity
`in the athymic mouse model (73). However, the potent antiestrogenic Z isomer
`is unstable (74, 75) and can be converted to the weakly antiestrogenic E isomer
`(76—78). If the isomerization occurs locally, the net antiestrogenicity of tam-
`oxifen will decrease, but this would not in itself account for increased tumor
`
`growth because an estrogenic stimulus is required. Minute amounts of Metab-
`olite E (tamoxifen without the dimethylaminoethane side chain) have been
`detected in human tumors during tamoxifen therapy (79). Fortunately, this
`metabolite of tamoxifen is too weakly estrogenic to promote tumor growth
`alone (64, 78). Nevertheless, the metabolite is unstable and can isomerize to
`a potent estrogen (78). If large quantities of this estrogenic metabolite can
`accumulate in the tumors, they could be the stimulus for tamoxifen-stimulated
`tumor growth. The hypothesis (80, 81) that tamoxifen-stimulated growth de-
`pends upon the simultaneous isomerization of metabolites to weak antiestro-
`gens and potent estrogens is illustrated in Figure 2.
`We recently addressed the question of metabolite isomerization as the mech-
`anism of tamoxifen-stimulated growth by determining the ability of tamoxifen
`derivatives, which cannot isomerize, to cause tumor growth. We found that a
`fixed-ring version of tamoxifen (Figure 2) can adequately support and develop
`ligand-stimulated tumor growth (82). Osborne and coworkers (83) have con-
`firmed our finding but also report that the related compound toremifene (see
`section on new agents) stimulates tumor growth, as does a tamoxifen derivative
`lacking the ether oxygen in the alkylaminoethoxy side chain. This latter com-
`pound cannot form Metabolite B, so the hypothesis is untenable. At present it
`is unclear how tamoxifen-stimulated tumor growth occurs within the cell, but
`one possibility is that the development of mutated estrogen receptors could
`alter the pharmacology of the antagonist to an agonist.
`
`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`AstraZeneca Exhibit 2017 p. 8
`
`

`

`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`TAMOXIFEN AND BREAST CANCER
`
`203
`
`/
`ON'K
`
`NC
`
`POTENT
`ANTIESTFIOGEN
`
`Isousmzs
`____,'
`
`wEAK
`ANTIESTROGEN
`
`H0
`4-HYDROXYTAMOXIFEN
`N/
`0N \
`
`umon
`METABOLIC
`
`H0
`
`u<
`
`
`
`TAMOXIFEN
`
`BLOCK
`
`BLOCK
`
`BREAST
`RESPONSE TUMOR
`I GROWTH
`
`FIXED RING
`TAMOXIFEN
`
`ROUTE
`
`vsnv umon
`METABOLIC
`ROUTE
`
`I
`'
`:V
`
`0"
`
`WEAK
`
`EsTROGEN
`
`\
`
`¢——-—-—
`__._..
`lSOIIERflE
`H0
`
`POTENT
`ESTROGEN
`
`METABOLITE E
`
`Figure 2 A proposed scheme for the metabolism of tamoxifen in breast tumors that could cause
`tamoxifen-stimulated growth. Tamoxifen could be converted to the potent antiestrogen
`4-hydroxytamoxifen and the weak estrogen referred to as Metabolite E. The key event in the
`hypothesis is the ability of the metabolites in the tumor cells to isomerize to a weak antiestrogen and
`a potent estrogen. The fixed-ring derivative of tamoxifen that cannot isomerize is biologically active
`at promoting growth of tamoxifen-dependent breast tumors. This observation makes the proposed
`scheme unlikely to be the major mechanism for tamoxifen-stimulated growth.
`
`Mutated Estrogen Receptors
`
`There is considerable interest in determining the biological relevance of mutant
`steroid hormone receptors. Laboratory models have demonstrated that specific
`mutations of the androgen (84) and progesterone receptors (85) can change
`the biological properties of antiandrogens and antiprogestins to full agonist
`molecules. Therefore, mutations in the estrogen receptor that change the phar-
`macology of an antiestrogen to an estrogen could explain tamoxifen-stimulated
`growth in tumors.
`The screening of clinical tumor material and cell lines has resulted in the
`identification of several mutations of the estrogen receptor (86-88), but the
`
`AstraZeneca Exhibit 2017 p. 9
`
`

`

`204
`
`JORDAN
`
`biological relevance of these findings is unclear. However, the impact of point
`mutations in the estrogen receptor on the pharmacology of antiestrogens can
`be examined under laboratory conditions. If MDA-MB-231 breast cancer cells
`(receptor negative) are transfected with either a wild-type estrogen receptor
`(ER) gene or an ER gene with a glycine-to-valine mutation at amino acid 400,
`the resulting transfected clones will respond to estrogen by decreasing growth
`(89). This laboratory model then becomes a test of the estrogenicity of any
`ligand receptor complex under controlled conditions. Pure antiestrogens pre-
`vent the inhibitory effect of estradiol in both wild-type and mutant cDNA
`(HEO) transfectants (89).
`In contrast, the antiestrogens that are normally partial agonists in assays
`involving wild-type receptors only express estrogenic activity in HEO trans-
`fected cells (90, 91). We have proposed a model that describes the changes in
`the folding of the mutant receptor around the antiestrogen that produces an
`estrogenic coupling (91). However, the HBO mutant is known to be a labora-
`tory cloning artifact, and until recently, no single~point mutations of the re-
`ceptor had been observed in nature.
`Tamoxifen-stimulated MCF—7 breast tumors that grow in athymic mice
`appear to have normal estrogen receptors. However, a screen of mRNAs for
`estrogen receptors in tamoxifen-stimulated tumors, using first reverse trans-
`criptase and polymerase chain reaction followed by single-stranded conforma-
`tional polymorphism, revealed a tumor line containing an estrogen receptor
`with a mutation (92). The error is a single-point mutation in the codon, which
`converts an aspartate to a tyrosine at AA351 in the steroid-binding domain
`(93). Preliminary studies with the cDNA for the novel mutant receptor
`demonstrate high activity for the protein when MDA-MB-231 cells are trans-
`fected with the gene and, most importantly, a conversion of the pharmacology
`of nonsteroidal antiestrogens to potent estrogenic activity (94). Although the
`development of drug resistance to tamoxifen is clearly not caused by mutation
`of the estrogen receptor alone, this mutation may be one of the many mecha’
`nisms that come into play during long-term tamoxifen treatment.
`A drug development program is well under way by the pharmaceutical
`industry to find either new first-line antiestrogens, which are less toxic than
`tamoxifen, or new second-line antiestrogenic agents to be used after tamoxifen
`treatment failure.
`
`CONCLUSIONS AND FUTURE DEVELOPMENTS
`
`The enormous success of tamoxifen as a first-line endocrine therapy for all
`stages of breast cancer has encouraged a search for alternative antiestrogens
`that might ultimately replace, or at least compliment, tamoxifen. Extensive
`clinical testing of a number of tamoxifen derivatives is under way. Toremifene
`
`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`AstraZeneca Exhibit 2017 p. 10
`
`

`

`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`TAMOXIFEN AND BREAST CANCER
`
`205
`
`ON NMag
`
`ONNMlz
`
`Q]
`Q] “ a]
`Tamoxifen
`
`Q
`Q] “ g]
`Toreiititene
`
`NNMng
`
`0
`
`0H
`
`"°
`
`a
`
`3] @395
`
`HO
`
`%(CH2)93°(CH21:0F25Fa
`
`Droloxifene
`
`ICI 182,780
`
`Figure 3 The formulae of new antiestrogens for breast cancer therapy.
`
`(95, 96) and droloxifene (97) are completing phase III trials against tamoxifen
`in postmenopausal women (Figure 3). Idoxifene (98) is entering phase II trials
`in the United Kingdom.
`The pure antiestrogen ICI 182.780 (Figure 3) is in clinical trials in the United
`Kingdom (99) and offers the advantage that it could be used as a second-line
`therapy if long-term adjuvant treatment results in tamoxifen-stimulated tumor
`growth. This principle has been demonstrated in the laboratory (70). The pure
`antiestrogens have a complete inhibitory effect on estrogen action in the
`primate uterus (100), but most importantly, the mode of action appears to be
`different than the nonsteroidal antiestrogens. Pure antiestrogens cause the loss
`of estrogen receptor from tumors and estrogen target tissues (101—103); thus
`the tissue becomes refractory to additional estrogenic stimulation.
`Finally, new antiestrogens could be targeted for novel applications. The
`nonsteroidal antiestrogen keoxifene (now renamed raloxifene) (Figure 4) pre-
`serves bone density in the ovariectomized rat (104, 105), and large doses will
`prevent the development of rat mammary tumors (21). The compound also
`decreases circulating cholesterol in the rat (105) and has only a weak agonist
`
`AstraZeneca Exhibit 2017 p. 11
`
`

`

`Annu.Rev.Pharmacol.Toxicol.l995.35:l95-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`206
`
`JORDAN
`
`
`
`4-IHYDROXYTAMOXIFEN
`HIGH AFFINITY FOR en
`
`\
`
`0H ® RALOXIFENE
`IMPROVED BONE DENSITY
`TARGETED
`
`ESTROGEN/ANTIESTROGEN
`
`//I4
`
`{C H2}B SO(('.‘1‘|2):I C|=2CFa
`
`ICI 182, 780
`PURE ANTIESTROGEN
`
`Figure 4 The new antiestrogen strategies to be developed based on the knowledge about the high
`binding affinity of 4-hydroxytamoxifen for the estrogen receptor. Each of the new agents has high
`binding to the estrogen receptor. but unlike the pure antiestrogen 1C1 182,780, the antiestrogen
`raloxifene has target site—specific effects and will be used to treat osteoporosis.
`
`effect in the rat and mouse uterus (105). One could also predict that there is
`a low probability of rat liver carcinogenesis.
`Raloxifene is being developed as a treatment for osteoporosis in post-
`menopausal women. The large numbers of postmenopausal women who would
`be treated with raloxifene to prevent osteoporosis might also be protected from
`coronary heart disease and breast cancer and have with a low probability of
`developing endometrial carcinoma and hepatocellular carcinoma. A future
`decrease in the incidence of breast cancer may occur as a positive side effect
`from the prevention of osteoporosis by an antiestrogen with targeted estrogenic
`properties.
`
`Any Annual Review chapter, as well as any article cited in an Annual Review chapter,
`may be purchased from the Annual Reviews Preprints and Reprints service.
`
`1-800-347-8007; 415-259-5017; email: arpr@class.org
`
`Literature Cited
`
`1. Harper MJK, Walpole AL. 1966. Con-
`trasting endocrine activities of cis and
`trans isomers in a series of substituted
`triphenylethylenes. Nature 212:8?
`
`2. Harper MJK, Walpole AL. 1967. A new
`derivative of triphenylethylene: effect
`on implantation and mode of action in
`rats. J. Reprod. Fem'l. 131101—19
`
`AstraZeneca Exhibit 2017 p. 12
`
`

`

`Annu.Rev.Pharmacol.Toxicol.1995.35:195-211.Downloadedfromwww.annualreviews.orgbyJohnsHopkins
`
`
`
`
`
`
`
`Universityon06/12/14.Forpersonaluseonly.
`
`TAMOXIFEN AND BREAST CANCER
`
`207
`
`Labhsetwar AP. 1970. Role of estrogens
`in ovulation. A study using the estrogen
`antagonist ICI 46,474. Endocrinology
`87:542—51
`Jordan VC. 1975. Prolonged antiestroge-
`nic activity of 1C1 46,474 in the ovari-
`ectomized mouse. J. Reprod. Fem'l. 52:
`251-58
`Jordan VC. 1974. Antitumour activity
`of the antioestrogen ICI 46.474 (tam-
`oxifen) in the dimethylbenzanthracene
`(DMBA)-induced rat mammary carci-
`noma model. J. Steroid Biochem. 5:354
`Jordan VC. Koemer S. 1975. Tamoxifen
`(1C1 46.474) and the human carcinoma
`88 oestrogen receptor. Eur. J. Cancer
`11:205—6
`Jordan VC. 1976. Effect of tamoxifen
`(ICI 46,474) on initiation and growth of
`DMBA-induced rat mammary carcino-
`mata. Eur. J. Cancer 12:419—24
`Jordan VC, Koemer S. 1976. Tamoxifen
`as an antitumour agent: role of oestradiol
`and prolactin. J. Endocrinol. 68:305-10
`Jordan VC. Jaspan T. 1976. Tamoxifen
`as an antitumour agent: oestrogen bind-
`ing as a predictive test for tumour re-
`sponse. J. Endocrinol. 68:453—60
`Jordan VC. 1984. Biochemical pharma-
`cology of antiestrogen action. Pharma-
`col. Rev. 36:245—76
`Jordan VC. 1988. The development
`of tamoxifen for breast cancer ther-
`apy: a tribute to the late Arthur L.
`Walpole. Breast Cancer Res. Treat.
`112197—209
`Lerner LI, Jordan VC. 1990. Develop
`ment of antiestrogens and their use in
`breast cancer: Eight Cain Memorial
`.sAward Lecture. Cancer Res. 50:4177-
`9
`Jordan VC. 1978. Use of the DMBA-
`induced rat mammary carcinoma system
`for the evaluation of tamoxifen treat-
`ment as a potential adjuvant therapy.
`Rev. Endocr. Relat. Cancer Suppl. Oct:
`49—55
`Jordan VC. 1983. Laboratory studies to
`develop general principles for the adju—
`vant
`treatment of breast cancer with
`antiestrogens: problems and potential for
`future clinical applications. Breast Can-
`cer Res. Treat. 3(Suppl.):73-86
`Early Breast Cancer Trialists' Collabo-
`rative Group. 1992. Systemic treatment
`of early breast cancer by hormonal,
`cytotoxic or immune therapy. Lancet
`339:1-15
`Morrow M, Jordan VC. 1993. Risk fac-
`tors and the prevention of breast cancer
`with tamoxifen.
`In Cancer Surveys:
`Breast Cancer, ed. JT Papademitriou, I
`Fentiman. 18:211—29. Imperial Cancer
`
`10.
`
`11.
`
`12.
`
`13.
`
`14.
`
`15.
`
`16.
`
`17.
`
`18.
`
`19.
`
`20.
`
`21.
`
`22.
`
`23.
`
`24.
`
`25.
`
`26.
`
`27.
`
`28.
`
`Res. Fund. London: Cold Spring Harbor
`Lab. Press
`1994. Long Term
`Jordan VC. ed.
`Tamoxifen Treatmentfor Breast Cancer.
`Madison: Univ. Wis. Press
`Jordan VC, Murphy CS. 1990. Endo-
`crine pharmacology of antiestrogens as
`Entitumor agents. Endocr. Rev. 11:578-
`10
`Jordan VC. 1993. Fourteenth Gaddum
`Memorial Lecture: A current view of
`tamoxifen for the treatment and preven-
`tion of breast cancer. Br. J. Pharmacol.
`110:507-17
`Turcot-LeMay L. Kelley PA. 1980.
`Characterizatio

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