Purpose: Cross-resistance is the primary issue facing the evaluation of new antiestrogens to treat metastatic breast cancer because they may be tested, initially, in populations of patients that have failed long-term adjuvant tamoxifen (Tam) therapy.

Experimental Design: We have tested the benzothiophene derivatives, arzoxifene (Arzox; LY353381) and LY117018 in two models of Tam-stimulated tumor growth derived from either MCF-7 (M. M. Gottardis and V. C. Jordan, Cancer Res., 48: 5183–5187, 1988) or T47D (J. MacGregor Schafer et al., Clin. Cancer Res., 6: 4373–4380, 2000) breast cancer cells.

Results: Using the MCF-7:Tam model, we found that both Arzox and LY117018 (1.5 mg/day) resulted in tumor growth and, therefore, were partially cross-resistant with Tam. Next, using the T47D:17β-estradiol (E2) model, we compared the antiestrogenic/antitumor properties of Arzox and LY117018 and determined that neither Arzox nor LY117018 caused T47D:E2 tumor growth after 21 weeks. In addition, we determined that long-term treatment does not result in failure and subsequent development of transplantable Arzox- or LY117018-stimulated tumors. To establish whether Arzox and LY117018 are cross-resistant in T47D:Tam tumors, mice were treated with Arzox or LY117018 (1.5 mg/day), and, again, we found that neither resulted in the growth of transplantable tumors. Lastly, we showed that Arzox and LY117018 were only partially able to compete with postmenopausal E2 (0.3 cm silastic capsule) in T47D:Tam tumors. However, when T47D:E2 tumors were treated for 7 days instead of 5 days, both Arzox and LY117018 were more effective.

Conclusions: Arzox is not cross-resistant with Tam in the T47D athymic mouse model but does exhibit cross-resistance in the MCF-7 model.

Tam4 is the prototype of a class of drugs called SERMs that have antiestrogenic effects in the breast but exhibit E2-like effects in bone and blood. Tam is the adjuvant endocrine treatment of choice for node-positive and node-negative ER-positive breast cancer (1) and is currently prescribed for the prevention of breast cancer in high-risk women (2). In addition, up to five years of adjuvant Tam treatment of patients with ER-positive disease confers a long-term survival benefit that continues for at least an additional 5 years after Tam treatment ceases (3). Unfortunately, >5 years of Tam treatment can lead to the clonal selection of metastatic breast cancers that are resistant to Tam (2). In fact, these tumors are stimulated by Tam as evidenced by the reduction in growth of tumors after Tam treatment is stopped (4, 5, 6). Most of these Tam-stimulated tumors retain expression of the ER (7) and, therefore, retain the ability to respond to second-line endocrine therapies such as the pure antiestrogen ICI182,780 (8) or an aromatase inhibitor such as anastrozole (9). A pure antiestrogen has no estrogenic properties because it causes destruction of the ER (10). Aromatase inhibitors are effective as second-line therapies because they block the synthesis of a woman’s endogenous E2(9), which can promote tumor growth after Tam treatment fails.

A goal of current efforts in medicinal chemistry is to design better SERMs. The ideal agent would: (a) not produce premature drug resistance; (b) not be cross-resistant with Tam; and (c) have fewer side effects such as the increase in endometrial cancer incidence observed with Tam. Clearly, a new SERM that could be used for >5 years and exhibit a better toxicological profile than Tam would provide great benefit as both a treatment and preventive agent.

Any new SERM would be directly compared with Tam and must not only be equally good but show a significant advantage over Tam. An agent under investigation would first be tested in a population of women with recurrent breast cancer that have failed Tam during adjuvant therapy; therefore, there is a high likelihood that these cancers would be resistant to Tam. Thus, any new agent should first be tested for cross-resistance to Tam in the laboratory. These data are extremely important, because they will be part of the clinical drug development decision-making paradigm that could influence the advancement of a drug for use as an adjuvant therapy in Tam-naive women.

New SERMs are being sought to improve the efficacy of breast cancer treatment and decrease side effects. The benzothiophene antiestrogens were first described in 1980 (11). The compound LY117018 (Fig. 1) demonstrated significantly lower uterotropic activity compared with Tam and, in fact, blocked increases in Tam-stimulated rat uterine weight (12, 13). A successor compound LY156758, known as keoxifene (Ref. 14; Fig. 1), has antitumor activity in laboratory models (15, 16, 17). The drug was not developed as a breast cancer therapy; the drug, renamed Ral, was subsequently developed clinically for the prevention (18) and treatment (19) of postmenopausal osteoporosis, with the additional effect of reducing the incidence of primary breast cancer (20). Although Ral is being evaluated as a preventive for breast cancer, it is not being promoted as a treatment for breast cancer. Bioavailability is too low at 2% of the administered dose (21) so there are concerns about effective continuous antitumor activity. As a result, the new agent, Arzox (22) has been designed to improve bioavailability and provide sustained antiestrogenic blockade (Fig. 1) so that it can be used to treat breast cancer.

The athymic mouse model that is routinely used to predict the potential clinical success of a new antiestrogen in a breast cancer clinical trial, is the ER-positive MCF-7 tumor xenograft that grows in response to E2(23, 24), Tam blocks E2-stimulated growth (25). The model has also been used to study the development of drug resistance to antiestrogens (26, 27, 28).

MCF-7 cells express wild-type p53 (29), which represents only 50% of breast cancers (30). Therefore, for these studies, we also use a novel human breast tumor line derived from T47D cells (31), which express a mutant p53 that is nonfunctional (32). After 10 weeks of Tam treatment, T47D tumors became Tam-stimulated (T47D:Tam) and remained E2-responsive. This occurs at a faster rate than MCF-7 tumors, which suggests that the differences between these tumor lines directly impact the development of Tam-stimulated growth (31).

Athymic Mouse Model.

The MCF-7 and T47D tumors used in these parallel experiments were developed as described previously (31). Ovariectomized 4–5-week-old athymic mice (Harlan Sprague Dawley, Madison, WI) were given bilateral transplants s.c. in the axillary mammary fat pads with 1-mm3 pieces of MCF-7 or T47D tumor using a trochar. The Animal Care and Use Committee of Northwestern University approved all of the procedures involving animals.

Hormone and Drug Treatments.

Mice were divided into groups of 10 and were treated with E2 (Sigma Chemical Co., St. Louis, MO), antiestrogens, or combinations. Silastic E2 capsules (0.3 cm or 1 cm in length) made as described previously (33) were implanted s.c. and were replaced every 8–10 weeks. The 0.3-cm E2 capsules produced a mean 83.8 pg/ml of serum E2, whereas 1.0-cm E2 capsules produced a mean 379.5 pg/ml of serum E2(34). Each was designed to represent the low or high E2 levels observed in post- or premenopausal women, respectively. Tam (Sigma Chemical Co.), LY117018, or Arzox (generous gifts of Eli Lilly, Indianapolis, IN; Fig. 1) were first dissolved in ethanol and suspended in a solution of 90% carboxymethylcellulose (1% carboxymethylcellulose in double-distilled water) and 10% polyethylene glycol 400/Tween 80 (99.5% polyethylene glycol 400 and 0.5% Tween 80). Ethanol was evaporated under nitrogen before use. Tam, LY117018, or Arzox was administered p.o. by gavage at a dose of either 0.5 or 1.5 mg per mouse per day, 5 days/week unless otherwise indicated.

Tumor Measurements.

Tumor measurements were performed weekly using Vernier calipers. The cross-sectional area was calculated using the formula: length × width/4 × π. Mean tumor cross-sectional area was determined by measuring tumors at all of the implantation sites.

Statistical Analysis.

Comparisons in mean tumor cross- sectional area between the animal groups were analyzed by ANOVA at each week followed by unpaired Student’s t test. The two-tailed P of the last week of each experiment was reported using StatMost 2.5 (Datamost Corp., Salt Lake City, UT).

We used the Tam-stimulated MCF-7 tumor model (MCF-7:Tam) to determine whether differences in tumor characteristics play a role in cross-resistance to Tam. The MCF-7:Tam tumors were developed as described previously (31). Fifty athymic mice bitransplanted with MCF-7:Tam tumors were treated as follows: 0.3 cm E2 capsule, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg LY117018, or control (Fig. 2). In addition to the above-described treatments, mice were treated with 1.5 mg Arzox+0.3 cm E2 capsule or 1.5 mg LY117018+0.3 cm E2 capsule, but these data were omitted from the figure for clarity. As expected, after 10 weeks, the E2 and Tam groups grew maximally. The growth of all of the groups compared with control was significant (P < 0.0001). However, the growth of E2versus Tam was not significant (P = 0.25). The growth of the 1.5 mg Arzox (P = 0.019) and 1.5 mg LY117018 (P = 0.016) tumor groups were significantly less than that of the E2 group. However, the growth of the 1.5 mg Arzox and LY117018 tumor groups compared with Tam, although less, only approached significance (P = 0.06). This demonstrates that Arzox and LY117018 are partially cross-resistant with Tam in the MCF-7 model. The growth of the 1.5 mg Arzox+E2 and LY117018+E2 tumor groups was not significantly different from the E2 or Tam groups (P > 0.09; data not shown).

We compared these findings with the T47D breast cancer tumor model (31). Previously, we showed that the T47D:E2 tumors grew robustly with pre- and postmenopausal levels of E2, and that Tam was able to block this growth (31). We tested Arzox and LY117018 in this model. T47D:E2 tumors were bitransplanted into 50 athymic mice and treated with 1 cm E2 capsule, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg LY117018, or control (Fig. 3). After 21 weeks, the E2-treated tumors grew rapidly, whereas the LY117018 and control groups did not grow at all. Tam produced stimulated tumors compared with either 1.5 mg Arzox (P = 0.004) or of LY117018 (P < 0.0001). The Tam group began to grow as expected at about 15 weeks demonstrating the acquisition of Tam-stimulated growth. The Arzox group grew minimally with two tumors measuring ∼0.5 cm2; these tumors were harvested and retransplanted but did not grow at all after 10 weeks of treatment (data not shown). This demonstrates that these tumors did not experience the phenotypic switch that the Tam-treated tumors experience and, therefore, did not become Arzox-stimulated.

We used a different experimental strategy in an attempt to produce a line of Arzox-stimulated tumors. We transplanted T47D:E2 tumors and treated all of the mice with 1 cm E2 capsules to establish a consistent tumor take. After 8 weeks, once the tumors had reached an average size of 0.3 cm2, the 1 cm E2 capsules were removed. The mice were subsequently treated for 4 weeks as follows: 1 cm E2 capsule, 0.5 mg Tam, 1.5 mg Arzox, or control (Fig. 4). By the end of week 12, the E2 group had grown significantly more (P < 0.05) than the 0.5 mg Tam group, 1.5 mg Arzox group, or control. Interestingly, both the 0.5 mg Tam and 1.5 mg Arzox treatments reduced tumor size compared with control, although Arzox was more effective. After an additional 10 weeks of treatment, the E2 group grew maximally (P < 0.001) compared with 0.5 mg Tam group, 1.5 mg Arzox, and control which did not grow (Fig. 5). We have shown previously, that low-dose (0.5 mg) Tam does not result in the development of Tam-stimulated tumor growth (31), and 1.5 mg Tam daily is necessary to produce Tam-stimulated tumors (Fig. 3).

Cross-resistance of the benzothiophenes and Tam was investigated using a Tam-stimulated T47D tumor line (T47D:Tam) whose growth is stimulated by both E2 and Tam (1.5 mg daily; Ref. 31). The T47D:Tam tumors were bitransplanted and were treated with 1 cm E2 capsule, 0.5 mg Tam, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg LY117018, or control (Fig. 6). After 18 weeks, as expected, both the E2 and the Tam groups grew maximally and were not significantly different from each other (P > 0.05) but were significantly different from control (P < 0.001), whereas the 0.5 mg Tam treatment group grew at about 50% of the 1.5 mg Tam rate. Neither the Arzox nor the LY117018 groups differed significantly from control (P > 0.05). This shows that both Arzox and LY117018 are not cross-resistant with Tam in the T47D:Tam tumor model. This is an important observation, but the real question is whether, when combined with E2 in the T47D:Tam tumor model, Arzox and LY117018 continue to act as antiestrogens.

To address this question, we bitransplanted T47D:Tam tumors into athymic mice and treated as follows: 0.3 cm E2 capsule, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg Arzox+0.3 cm E2 capsule, 1.5 mg LY117018, 1.5 mg LY117018+0.3 cm E2 capsule, or control (Fig. 7). In this experiment, after 15 weeks, the growth of both postmenopausal E2 and Tam was not significantly different (P = 0.4), whereas the Arzox and the LY117018 groups were similar to the control (P > 0.05). Interestingly, when combined with postmenopausal levels of E2, both the Arzox and the LY117018 were able to significantly block growth compared with E2 and Tam alone (P < 0.01); however, they did not completely block growth. This can be explained in two ways. Firstly, Arzox and LY117018 at the doses tested are not capable of fully blocking E2-stimulated growth, i.e., higher doses are required. Secondly, mice are fed Arzox and LY117018 p.o. once/day, 5 days/week, whereas the E2 capsules are implanted resulting in dosing of postmenopausal E2 24 h/day, 7 days/week. Because the related compound, Ral, is poorly bioavailable and is cleared rapidly (21), the blood levels of Arzox and LY117018 may drop below efficacious levels and are, therefore, unable to block the growth-stimulatory effects of E2.

To address the issue of continuous drug treatment, we examined the efficacy of 5 versus 7 days of Arzox and LY117018 combined with postmenopausal levels of E2 in the T47D:E2 model. We bitransplanted T47D:E2 tumors into mice and treated them as follows: 0.3 cm E2 capsule, 1.5 mg Tam (5 days)+0.3 cm E2 capsule, 1.5 mg Arzox (5 days)+0.3 cm E2 capsule, 1.5 mg Arzox (7 days)+0.3 cm E2 capsule, 1.5 mg LY117018 (5 days)+0.3 cm E2 capsule, 1.5 mg LY117018 (7 days)+0.3 cm E2 capsule, or control (Fig. 8 A). As expected, the E2 group grew significantly more than all of the antiestrogen-treated groups (P < 0.0001).

LY117018 was less effective than Arzox at either 5 day (P < 0.001) or 7 day (P < 0.0001) dosing. Additionally, 5 day treatment with Arzox was not significantly different from 5 day treatment with Tam, and both were superior to 5 day treatment with LY117018. Treatment for 7 days with Arzox was more effective than that for 5 days (P < 0.02). Most importantly, 7 day treatment with Arzox completely blocks E2 action to the level observed in the untreated control group. The comparison of the tumor areas after 12 weeks of antiestrogen treatment is shown in Fig. 8 B.

The development of the T47D Tam-resistant breast cancer model has introduced a new dimension into the investigation of drug resistance to SERMs (31). We have found that the MCF-7 model and the T47D breast cancer model are distinctly different in their response to SERMs, and this may have important implications for the sequential use of these compounds clinically.

Arzox and LY117018 are partially cross-resistant with Tam in the MCF-7:Tam tumor model but not in the T47D:Tam tumor model. This is distinct from our findings with the triphenylethylene compound, idoxifene, which was cross-resistant to Tam in the MCF-7:Tam and T47D:Tam tumor models (35). This suggests that the differences in the estrogen-like properties and mechanism of action of SERMs in laboratory assays (36) is now demonstrated in the signal transduction pathways of two Tam-stimulated breast tumor models.

Possible differences in coregulator expression may play a role in dictating response of the tumors to different classes of antiestrogens. During the progression of MCF-7:E2 tumors from Tam-sensitive to Tam-stimulated, which occurs after ∼8 months of Tam treatment, changes occur under selection pressure that may result in the down-regulation of corepressor(s). [The acquisition of Tam-resistance in a mouse model of human breast cancer correlates with decreased levels of the corepressor N-CoR (37).] If this is the case, treatment with a different SERM such as Arzox would not be advantageous because the down-regulation of corepressor(s) has already occurred. However, there are mechanistic distinctions between different SERM:ER complexes (38, 39) that may result in the binding of different corepressors, which may be the reason cross-resistance does not necessarily occur.

In addition, these differences in SERM:ER complexes may also affect coactivator binding (40). The development of T47D:Tam tumors from T47D:E2 tumors occurs very rapidly (31), after ∼3 months. A different mechanism of resistance may evolve that results in an increase in coactivator(s) that leads to Tam-stimulated growth. Because distinct SERM:ER complexes are formed, it is possible that different coactivators specifically activate transcription of specific complexes. For example, L7/SPA (the only antagonist-specific coactivator defined to date) (41) increases transcription of the Tam:ER complex but not of the pure antiestrogen ICI164,384:ER complex, which proves that this coactivator specifically up-regulates based on the ligand (42).

In previous studies, we have described an in vitro antiestrogen classification system that differentiates antiestrogens through the use of mutant ERs (43, 44, 45). These ERs are stably transfected into ER-negative MDA-MB-231 cells and contain mutations at amino acid 351: D351 (wild type), D351Y, and D351G. A TGFα assay system is used to classify antiestrogens as Tam-like, Ral-like, or pure antiestrogen. As shown in Table 1, there are differences between the growth responses of MCF-7:Tam tumors and T47D:Tam tumors to E2 and antiestrogens. When TGFα is measured in the ER-expressing MDA-MB-231 cells, the response is identical to that of the T47D:Tam tumors. This is a significant observation because these models reflect the inherent differences between breast cancer cells that dictate responses to antiestrogens. At this point, the most likely candidates for future studies are coactivators and corepressors. Studies are in progress to determine the relative levels of known coregulators in our models in vivo and in vitro.

Many tumors that first respond to Tam will ultimately develop acquired Tam resistance (7). However, approximately two-thirds of the tumors that become resistant to Tam continue to express ER (46, 47); therefore, many of these respond to second-line endocrine therapies. Consequently, cross-resistance to Tam is an important issue in the development of novel SERMs for the treatment of breast cancer.

Tam is a prodrug being converted to the active metabolite 4-OHT, which blocks estrogen binding to the ER (48). Serum levels of 4-OHT during long-term adjuvant Tam (150 ng/ml) are ∼2–5 ng/ml (49). Thus, the prolonged serum half-life of Tam (7 days; Ref. 50) and the stable levels of 4-OHT provide adequate blockade of the breast cancer ER in patients. Azox is designed to improve bioavailability over the 2% observed with Ral (21). A recent Phase I clinical trial of Arzox in heavily treated postmenopausal breast cancer patients showed no objective responses, but Phase II studies are evaluating the efficacy if the drug in patients who failed prior Tam (51). Nevertheless, a pharmacokinetic evaluation of Arzox, the parent drug, show steady-state levels between <1–20 ng/ml for daily dosing regimens between 10 and 100 mg Arzox. The levels of the desmethyl active metabolite of Arzox, LY335362, were below the level of detection (<0.05 ng/ml). Clearly, the polyphenolic nature of the benzothiophene antiestrogens results in rapid clearance and emphasize the need for strict compliance in high-estrogen environments.

The experimental doses of Arzox and LY117018 were selected to directly compare the ratios to the doses of Tam used in the clinic. Tam is used at 20 mg daily (1), whereas Ral is recommended for the treatment of osteoporosis at a dose of 60 mg daily (three times the Tam dose; Ref. 19). The increased metabolic stability and oral bioavailability of Arzox at the doses tested were illustrated when Arzox and LY117018 (a raloxifene analogue) were directly compared in the T47D:Tam model (Fig. 7). In this situation, 5 days of Arzox or LY117018 treatment did not stimulate growth but when combined with E2 capsule, which released E2 7 days week, they were both unable to completely block E2-stimulated growth. We found a difference between the two feeding schedules and profound differences between Arzox and LY117018 when 5 versus 7 days of treatment was compared (Fig. 8A). Arzox completely inhibited E2-stimulated tumor growth on a 7 day treatment schedule, whereas both Arzox and LY117018 were much less effective on a 5 day treatment schedule. Tam is known to accumulate to high levels in patients (1). This is beneficial in the treatment of breast cancer to produce continuous ER blockade. Clearly, the issue of bioavailability and compliance will become very important when evaluating the test results of new, rapidly excreted SERMs. It is clear from these data that the low bioavailability of polyhydroxylated SERMs (21) will be reduced further by sporadic dosing schedules. Compliance will be crucial for maintaining the effectiveness of Arzox in premenopausal breast cancer patients or if Arzox is evaluated as a preventative in premenopausal women.

In conclusion, the primary goal of the study was to evaluate the SERMs Arzox and LY117018 in E2- and Tam-stimulated breast cancer models to determine whether they are cross-resistant with Tam. These findings demonstrate that the differences in tumor characteristics between breast cancers can dictate the effectiveness of antiestrogen therapies and reflect the putative differences in the mechanisms of actions of SERMs. Because Arzox is not cross-resistant in the T47D:Tam tumor model but is cross-resistant in the MCF-7:Tam tumor model, this SERM may be effective in some patients who have failed Tam therapy. However, the disparate actions of Arzox in the two models suggests that sequential treatment with Arzox after Tam may not provide an optimal treatment option. Nevertheless, these studies suggest that clinical studies should be pursued not only in advanced breast cancer but also in adjuvant therapy. The observation that Arzox appears to be less susceptible to the development of drug resistance compared with Tam indicates that longer adjuvant treatment schedules may be possible.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

        
1

Supported by the following: NIH SPORE in breast cancer 1P50 CA89018-01 (to V. C. J.); Department of Defense Breast Cancer Training Grants DAMD 17-94-J-4466 and DAMD 17-96-1-6169 (to J. M. S.); Breast Cancer Program Development Grant P20 CA65764 (to V. C. J.); Coordenação de Aperfeiçoamento de Pessoal de Nível Superior Scholarship (to R. C. D.); a grant from Eli Lilly, Indianapolis, IN (to V. C. J.); and the generous support of the Lynn Sage Breast Cancer Foundation of Northwestern Memorial Hospital and the Avon Products Foundation.

                        
4

The abbreviations used are: Tam, tamoxifen; ER, estrogen receptor; E2, 17β-estradiol; Arzox, arzoxifene; Ral, raloxifene; 4-OHT, 4-hydroxytamoxifen; SERM, selective ER modulator; TGF, transforming growth factor.

Fig. 1.

Chemical structures of Tam, Ral, Arzox (LY353381), and LY117018.

Fig. 1.

Chemical structures of Tam, Ral, Arzox (LY353381), and LY117018.

Close modal
Fig. 2.

Comparison of Tam-stimulated MCF-7:Tam tumor growth in response to Tam, or Arzox, or LY117018 treatment (mean ± SE). Fifty ovariectomized athymic mice were bilaterally transplanted with MCF-7:Tam tumors (10/group) and were treated for 10 weeks with 0.3 cm E2 capsule, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg LY117018, or control.

Fig. 2.

Comparison of Tam-stimulated MCF-7:Tam tumor growth in response to Tam, or Arzox, or LY117018 treatment (mean ± SE). Fifty ovariectomized athymic mice were bilaterally transplanted with MCF-7:Tam tumors (10/group) and were treated for 10 weeks with 0.3 cm E2 capsule, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg LY117018, or control.

Close modal
Fig. 3.

Growth of T47D:E2 tumors in ovariectomized athymic mice (mean ± SE). Fifty mice (10/group) were bilaterally transplanted with T47D:E2 tumors and were treated for 21 weeks with 1 cm E2 capsule, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg LY117018, or control.

Fig. 3.

Growth of T47D:E2 tumors in ovariectomized athymic mice (mean ± SE). Fifty mice (10/group) were bilaterally transplanted with T47D:E2 tumors and were treated for 21 weeks with 1 cm E2 capsule, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg LY117018, or control.

Close modal
Fig. 4.

Inability to develop Arzox-stimulated tumors in T47D:E2 tumors in ovariectomized athymic mice (mean ± SE). Forty mice were bilaterally transplanted with T47D:E2 tumors and were treated for 8 weeks with 1 cm E2 capsule. Mice were then separated into groups of 10 and treated for an additional 4 weeks with 1 cm E2 capsule, 0.5 mg Tam, 1.5 mg Arzox, or control.

Fig. 4.

Inability to develop Arzox-stimulated tumors in T47D:E2 tumors in ovariectomized athymic mice (mean ± SE). Forty mice were bilaterally transplanted with T47D:E2 tumors and were treated for 8 weeks with 1 cm E2 capsule. Mice were then separated into groups of 10 and treated for an additional 4 weeks with 1 cm E2 capsule, 0.5 mg Tam, 1.5 mg Arzox, or control.

Close modal
Fig. 5.

Long-term attempt to develop Arzox-stimulated tumors in T47D:E2 tumors in ovariectomized athymic mice (mean ± SE). Forty mice bearing T47D:E2 tumors were treated with 1 cm E2 capsule until the tumors reached an average size of 0.3 cm2. At this point, mice were separated into groups of 10 and treated for an additional 16 weeks with 1 cm E2 capsule, 0.5 mg Tam, 1.5 mg Arzox, or control.

Fig. 5.

Long-term attempt to develop Arzox-stimulated tumors in T47D:E2 tumors in ovariectomized athymic mice (mean ± SE). Forty mice bearing T47D:E2 tumors were treated with 1 cm E2 capsule until the tumors reached an average size of 0.3 cm2. At this point, mice were separated into groups of 10 and treated for an additional 16 weeks with 1 cm E2 capsule, 0.5 mg Tam, 1.5 mg Arzox, or control.

Close modal
Fig. 6.

Arzox and LY117018 do not stimulate growth in T47D:Tam tumor compared with E2 and Tam in ovariectomized athymic mice (mean ± SE). T47D:Tam tumors were bitransplanted into 60 athymic mice and were treated as follows: 1 cm E2 capsule, 0.5 mg Tam, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg LY117018, or control.

Fig. 6.

Arzox and LY117018 do not stimulate growth in T47D:Tam tumor compared with E2 and Tam in ovariectomized athymic mice (mean ± SE). T47D:Tam tumors were bitransplanted into 60 athymic mice and were treated as follows: 1 cm E2 capsule, 0.5 mg Tam, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg LY117018, or control.

Close modal
Fig. 7.

Arzox and LY117018 are not cross-resistant to Tam in T47D:Tam tumors grown in ovariectomized athymic mice (mean ± SE). Seventy ovariectomized athymic mice (10/group) were bilaterally transplanted with T47D:Tam tumors and treated for 15 weeks with 0.3 cm E2 capsule, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg Arzox+0.3 cm E2 capsule, 1.5 mg LY117018, 1.5 mg LY117018+0.3 cm E2 capsule, or control.

Fig. 7.

Arzox and LY117018 are not cross-resistant to Tam in T47D:Tam tumors grown in ovariectomized athymic mice (mean ± SE). Seventy ovariectomized athymic mice (10/group) were bilaterally transplanted with T47D:Tam tumors and treated for 15 weeks with 0.3 cm E2 capsule, 1.5 mg Tam, 1.5 mg Arzox, 1.5 mg Arzox+0.3 cm E2 capsule, 1.5 mg LY117018, 1.5 mg LY117018+0.3 cm E2 capsule, or control.

Close modal
Fig. 8.

Comparison of 5 and 7 day dosing of Arzox or LY117018 in T47D:E2 tumors grown in ovariectomized athymic mice (mean ± SE). A, seventy ovariectomized athymic mice (10/group) were bilaterally transplanted with T47D:E2 tumors and treated for 12 weeks with 0.3 cm E2 capsule, 1.5 mg Tam (5 day)+0.3 cm E2 capsule, 1.5 mg Arzox (5 day)+0.3 cm E2 capsule, 1.5 mg Arzox (7 day)+0.3 cm E2 capsule, 1.5 mg LY117018 (5 day)+0.3 cm E2 capsule, 1.5 mg LY117018 (7 day)+0.3 cm E2 capsule, or control. B, mean tumor growth of mice treated with antiestrogens at week 12.

Fig. 8.

Comparison of 5 and 7 day dosing of Arzox or LY117018 in T47D:E2 tumors grown in ovariectomized athymic mice (mean ± SE). A, seventy ovariectomized athymic mice (10/group) were bilaterally transplanted with T47D:E2 tumors and treated for 12 weeks with 0.3 cm E2 capsule, 1.5 mg Tam (5 day)+0.3 cm E2 capsule, 1.5 mg Arzox (5 day)+0.3 cm E2 capsule, 1.5 mg Arzox (7 day)+0.3 cm E2 capsule, 1.5 mg LY117018 (5 day)+0.3 cm E2 capsule, 1.5 mg LY117018 (7 day)+0.3 cm E2 capsule, or control. B, mean tumor growth of mice treated with antiestrogens at week 12.

Close modal
Table 1

A classification of agents that either stimulate or block Tam-resistant tumor growth (MCF-7:Tam or T47D:Tam)

The tumor growth characteristics are compared with the SERM classification system based on the stimulation or blockade of the TGFα gene in the ER-negative MDA-MB-231 breast cancer cell line stably transfected with the wild-type ER cDNA (36). The numbers in parentheses are the references.
 MCF-7:Tam T47D:Tam TGFα assay 
E2 Stimulate (26) Stimulate (31) Stimulate (52) 
Tam-like Stimulate (35) Stimulate (35) Stimulate (45, 53) 
Ral-like Stimulate (Fig. 2) Block (Fig. 7) Block (43) 
Pure antiestrogen Block (35, 54) Blocka Block (44) 
The tumor growth characteristics are compared with the SERM classification system based on the stimulation or blockade of the TGFα gene in the ER-negative MDA-MB-231 breast cancer cell line stably transfected with the wild-type ER cDNA (36). The numbers in parentheses are the references.
 MCF-7:Tam T47D:Tam TGFα assay 
E2 Stimulate (26) Stimulate (31) Stimulate (52) 
Tam-like Stimulate (35) Stimulate (35) Stimulate (45, 53) 
Ral-like Stimulate (Fig. 2) Block (Fig. 7) Block (43) 
Pure antiestrogen Block (35, 54) Blocka Block (44) 
a

Unpublished observations.

1
Osborne C. K. Tamoxifen in the treatment of breast cancer.
N. Engl. J. Med.
,
339
:
1609
-1618,  
1998
.
2
Fisher B., Costantino J. P., Wickerham D. L., Redmond C. K., Kavanah M., Cronin W. M., Vogel V., Robidoux A., Dimitrov N., Atkins J., Daly M., Wieand S., Tan-Chiu E., Ford L., Wolmark N. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study.
J. Natl. Cancer Inst. (Bethesda)
,
90
:
1371
-1388,  
1998
.
3
Early Breast Cancer Treatment Collaborative Group. Tamoxifen for early breast cancer: an overview of the randomised trials.
Lancet
,
351
:
1451
-1467,  
1998
.
4
Canney P. A., Griffiths T., Latief T. N., Priestman T. J. Clinical significance of tamoxifen withdrawal response.
Lancet
,
1
:
36
1987
.
5
Howell A., Dodwell D. J., Anderson H., Redford J. Response after withdrawal of tamoxifen and progestogens in advanced breast cancer.
Ann. Oncol.
,
3
:
611
-617,  
1992
.
6
Legault-Poisson S., Jolivet J., Poisson R., Beretta-Piccoli M., Band P. R. Tamoxifen-induced tumor stimulation and withdrawal response.
Cancer Treat. Rep.
,
63
:
1839
-1841,  
1979
.
7
Robertson J. F. Oestrogen receptor: a stable phenotype in breast cancer.
Br. J. Cancer
,
73
:
5
-12,  
1996
.
8
Howell A., DeFriend D. J., Robertson J. F., Blamey R. W., Anderson L., Anderson E., Sutcliffe F. A., Walton P. Pharmacokinetics, pharmacological and anti-tumour effects of the specific anti-oestrogen ICI 182780 in women with advanced breast cancer.
Br. J. Cancer
,
74
:
300
-308,  
1996
.
9
Buzdar A. U., Jonat W., Howell A., Plourde P. V. ARIMIDEX: a potent and selective aromatase inhibitor for the treatment of advanced breast cancer.
J. Steroid Biochem. Mol. Biol.
,
61
:
145
-149,  
1997
.
10
Dauvois S., Danielian P. S., White R., Parker M. G. Antiestrogen ICI 164,384 reduces cellular estrogen receptor content by increasing its turnover.
Proc. Natl. Acad. Sci. USA
,
89
:
4037
-4041,  
1992
.
11
Black L. J., Goode R. L. Uterine bioassay of tamoxifen, trioxifene and a new estrogen antagonist (LY117018) in rats and mice.
Life Sci.
,
26
:
1453
-1458,  
1980
.
12
Jordan V. C., Gosden B. Differential antiestrogen action in the immature rat uterus: a comparison of hydroxylated antiestrogens with high affinity for the estrogen receptor.
J. Steroid Biochem.
,
19
:
1249
-1258,  
1983
.
13
Jordan V. C., Gosden B. Inhibition of the uterotropic activity of estrogens and antiestrogens by the short acting antiestrogen LY117018.
Endocrinology
,
113
:
463
-468,  
1983
.
14
Black L. J., Jones C. D., Falcone J. F. Antagonism of estrogen action with a new benzothiophene derived antiestrogen.
Life Sci.
,
32
:
1031
-1036,  
1983
.
15
Clemens J. A., Bennett D. R., Black L. J., Jones C. D. Effects of a new antiestrogen, keoxifene (LY156758), on growth of carcinogen-induced mammary tumors and on LH and prolactin levels.
Life Sci.
,
32
:
2869
-2875,  
1983
.
16
Gottardis M. M., Jordan V. C. Antitumor actions of keoxifene and tamoxifen in the N-nitrosomethylurea-induced rat mammary carcinoma model.
Cancer Res.
,
47
:
4020
-4024,  
1987
.
17
Anzano M. A., Peer C. W., Smith J. M., Mullen L. T., Shrader M. W., Logsdon D. L., Driver C. L., Brown C. C., Roberts A. B., Sporn M. B. Chemoprevention of mammary carcinogenesis in the rat: combined use of raloxifene and 9-cis-retinoic acid.
J. Natl. Cancer Inst. (Bethesda)
,
88
:
123
-125,  
1996
.
18
Delmas P. D., Bjarnason N. H., Mitlak B. H., Ravoux A. C., Shah A. S., Huster W. J., Draper M., Christiansen C. Effects of raloxifene on bone mineral density, serum cholesterol concentrations, and uterine endometrium in postmenopausal women [see comments].
N. Engl. J. Med.
,
337
:
1641
-1647,  
1997
.
19
Ettinger B., Black D. M., Mitlak B. H., Knickerbocker R. K., Nickelsen T., Genant H. K., Christiansen C., Delmas P. D., Zanchetta J. R., Stakkestad J., Gluer C. C., Krueger K., Cohen F. J., Eckert S., Ensrud K. E., Avioli L. V., Lips P., Cummings S. R. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators [see comments].
JAMA
,
282
:
637
-645,  
1999
.
20
Cummings S. R., Eckert S., Krueger K. A., Grady D., Powles T. J., Cauley J. A., Norton L., Nickelsen T., Bjarnason N. H., Morrow M., Lippman M. E., Black D., Glusman J. E., Costa A., Jordan V. C. The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation [see comments].
JAMA
,
281
:
2189
-2197,  
1999
.
21
Snyder K. R., Sparano N., Malinowski J. M. Raloxifene hydrochloride.
Am. J. Health Syst. Pharm.
,
57
:
1669
-1678; 16761668,  
2000
.
22
Sato M., Turner C. H., Wang T., Adrian M. D., Rowley E., Bryant H. U. LY353381.HCl: a novel raloxifene analog with improved SERM potency and efficacy in vivo.
J. Pharmacol. Exp. Ther.
,
287
:
1
-7,  
1998
.
23
Soule H. D., Vazguez J., Long A., Albert S., Brennan M. A human cell line from a pleural effusion derived from a breast carcinoma.
J. Natl. Cancer Inst. (Bethesda)
,
51
:
1409
-1416,  
1973
.
24
Levenson A. S., Jordan V. C. MCF-7: the first hormone-responsive breast cancer cell line.
Cancer Res.
,
57
:
3071
-3078,  
1997
.
25
Osborne C. K., Hobbs K., Clark G. M. Effect of estrogens and antiestrogens on growth of human breast cancer cells in athymic nude mice.
Cancer Res.
,
45
:
584
-590,  
1985
.
26
Gottardis M. M., Jordan V. C. Development of tamoxifen-stimulated growth of MCF-7 tumors in athymic mice after long-term antiestrogen administration.
Cancer Res.
,
48
:
5183
-5187,  
1988
.
27
Osborne C. K., Coronado E. B., Robinson J. P. Human breast cancer in the athymic nude mouse: cytostatic effects of long-term antiestrogen therapy.
Eur. J. Cancer Clin. Oncol.
,
23
:
1189
-1196,  
1987
.
28
Wolf D. M., Jordan V. C. Characterization of tamoxifen stimulated MCF-7 tumor variants grown in athymic mice.
Breast Cancer Res. Treat.
,
31
:
117
-127,  
1994
.
29
Casey G., Lo-Hsueh M., Lopez M. E., Vogelstein B., Stanbridge E. J. Growth suppression of human breast cancer cells by the introduction of a wild-type p53 gene.
Oncogene
,
6
:
1791
-1797,  
1991
.
30
Hollstein M., Rice K., Greenblatt M. S., Soussi T., Fuchs R., Sorlie T., Hovig E., Smith-Sorensen B., Montesano R., Harris C. C. Database of p53 gene somatic mutations in human tumors and cell lines.
Nucleic Acids Res.
,
22
:
3551
-3555,  
1994
.
31
MacGregor Schafer J., Lee E-S., O’Regan R. M., Yao K., Jordan V. C. Rapid development of tamoxifen stimulated mutant p53 breast tumors (T47D) in athymic mice.
Clin. Cancer Res.
,
6
:
4373
-4380,  
2000
.
32
O’Connor P. M., Jackman J., Bae I., Myers T. G., Fan S., Mutoh M., Scudiero D. A., Monks A., Sausville E. A., Weinstein J. N., Friend S., Fornace A. J., Jr., Kohn K. W. Characterization of the p53 tumor suppressor pathway in cell lines of the National Cancer Institute anticancer drug screen and correlations with the growth-inhibitory potency of 123 anticancer agents.
Cancer Res.
,
57
:
4285
-4300,  
1997
.
33
Robinson S. P., Jordan V. C. Antiestrogenic action of toremifene on hormone-dependent, -independent, and -heterogeneous breast tumor growth in the athymic mouse.
Cancer Res.
,
49
:
1758
-1762,  
1989
.
34
O’Regan R. M., Cisneros A., England G. M., MacGregor J. I., Muenzner H. D., Assikis V. J., Bilimoria M. M., Piette M., Dragan Y. P., Pitot H. C., Chatterton R., Jordan V. C. Effects of the antiestrogens tamoxifen, toremifene, and ICI 182,780 on endometrial cancer growth [see comments].
J. Natl. Cancer Inst. (Bethesda)
,
90
:
1552
-1558,  
1998
.
35
Lee E. S., MacGregor Schafer J., Yao K., England G., O’Regan R., Reyes A. D., Jordan V. C. Cross-resistance of triphenylethylene type antiestrogens but not ICI 182,780 in tamoxifen-stimulated breast tumors grown in athymic mice.
Clin. Cancer Res.
,
6
:
4893
-4899,  
2000
.
36
Levenson A. S., Jordan V. C. Selective oestrogen receptor modulation: molecular pharmacology for the millennium.
Eur. J. Cancer
,
35
:
1628
-1639,  
1999
.
37
Lavinsky R. M., Jepsen K., Heinzel T., Torchia J., Mullen T. M., Schiff R., Del-Rio A. L., Ricote M., Ngo S., Gemsch J., Hilsenbeck S. G., Osborne C. K., Glass C. K., Rosenfeld M. G., Rose D. W. Diverse signaling pathways modulate nuclear receptor recruitment of N-CoR and SMRT complexes.
Proc. Natl. Acad. Sci. USA
,
95
:
2920
-2925,  
1998
.
38
Wijayaratne A. L., Nagel S. C., Paige L. A., Christensen D. J., Norris J. D., Fowlkes D. M., McDonnell D. P. Comparative analyses of mechanistic differences among antiestrogens.
Endocrinology
,
140
:
5828
-5840,  
1999
.
39
Liu H., Lee E-S., De Los Reyes A., Zapf J. W., Jordan V. C. Silencing and reactivation of the selective estrogen receptor modulator-estrogen receptor α complex.
Cancer Res.
,
61
:
3632
-3639,  
2001
.
40
Norris J. D., Paige L. A., Christensen D. J., Chang C. Y., Huacani M. R., Fan D., Hamilton P. T., Fowlkes D. M., McDonnell D. P. Peptide antagonists of the human estrogen receptor.
Science (Wash. DC)
,
285
:
744
-746,  
1999
.
41
Graham D. D., Bain D. L., Richer J. K., Jackson T. A., Tung L., Horwitz K. B. Thoughts on tamoxifen resistant breast cancer. Are coregulators the answer or just a red herring?.
J. Steroid Biochem. Mol. Biol.
,
74
:
225
-229,  
2000
.
42
Jackson T. A., Richer J. K., Bain D. L., Takimoto G. S., Tung L., Horwitz K. B. The partial agonist activity of antagonist-occupied steroid receptors is controlled by a novel hinge domain-binding coactivator L7/SPA and the corepressors N-CoR or SMRT.
Mol. Endocrinol.
,
11
:
693
-705,  
1997
.
43
Levenson A. S., Jordan V. C. The key to the antiestrogenic mechanism of raloxifene is amino acid 351 (aspartate) in the estrogen receptor.
Cancer Res.
,
58
:
1872
-1875,  
1998
.
44
MacGregor Schafer J., Liu H., Tonetti D. A., Jordan V. C. The interaction of raloxifene and the active metabolite of the antiestrogen (SC5705) with the human estrogen receptor (ER).
Cancer Res.
,
59
:
4308
-4313,  
1999
.
45
MacGregor Schafer J., Liu H., Bentrem D. J., Zapf J. W., Jordan V. C. Allosteric silencing of activating function 1 in the 4-hydroxytamoxifen estrogen receptor complex is induced by substituting glycine for aspartate at amino acid 351.
Cancer Res.
,
60
:
5097
-5105,  
2000
.
46
Encarnacion C. A., Ciocca D. R., McGuire W. L., Clark G. M., Fuqua S. A., Osborne C. K. Measurement of steroid hormone receptors in breast cancer patients on tamoxifen.
Breast Cancer Res. Treat.
,
26
:
237
-246,  
1993
.
47
Johnston S. R., Lu B., Dowsett M., Liang X., Kaufmann M., Scott G. K., Osborne C. K., Benz C. C. Comparison of estrogen receptor DNA binding in untreated and acquired antiestrogen-resistant human breast tumors.
Cancer Res.
,
57
:
3723
-3727,  
1997
.
48
Allen K. E., Clark E. R., Jordan V. C. Evidence for the metabolic activation of non-steroidal antioestrogens: a study of structure-activity relationships.
Br. J. Pharmacol.
,
71
:
83
-91,  
1980
.
49
Langan-Fahey S. M., Tormey D. C., Jordan V. C. Tamoxifen metabolites in patients on long-term adjuvant therapy for breast cancer.
Eur. J. Cancer
,
26
:
883
-888,  
1990
.
50
Furr B. J., Jordan V. C. The pharmacology and clinical uses of tamoxifen.
Pharmacol. Ther.
,
25
:
127
-205,  
1984
.
51
Munster P. N., Buzdar A., Dhingra K., Enas N., Ni L., Major M., Melemed A., Seidman A., Booser D., Theriault R., Norton L., Hudis C. Phase I study of a third-generation selective estrogen receptor modulator, ly353381.hcl, in metastatic breast cancer.
J. Clin. Oncol.
,
19
:
2002
-2009,  
2001
.
52
Jeng M. H., Jiang S. Y., Jordan V. C. Paradoxical regulation of estrogen-dependent growth factor gene expression in estrogen receptor (ER)-negative human breast cancer cells stably expressing ER.
Cancer Lett.
,
82
:
123
-128,  
1994
.
53
Levenson A. S., Tonetti D. A., Jordan V. C. The oestrogen-like effect of 4-hydroxytamoxifen on induction of transforming growth factor α mRNA in MDA-MB-231 breast cancer cells stably expressing the oestrogen receptor.
Br. J. Cancer
,
77
:
1812
-1819,  
1998
.
54
Osborne C. K., Coronado-Heinsohn E. B., Hilsenbeck S. G., McCue B. L., Wakeling A. E., McClelland R. A., Manning D. L., Nicholson R. I. Comparison of the effects of a pure steroidal antiestrogen with those of tamoxifen in a model of human breast cancer.
J. Natl. Cancer Inst. (Bethesda)
,
87
:
746
-750,  
1995
.