Molec. Aspects Med. Vol. 18, pp. 187-247, 1997 0 1997 Elsevier Science Ltd. All rights resetved Printed in Great Britain 0098-2997197 $32.00+0.00
PII: SOO98-2997(96)00015-5
MOLECULAR
MECHANISMS AND FUTURE ANTIESTROGENS
USES
OF
V. Craig Jordan and William J. Gradishar The Robert H. Lurie Cancer Center and Medical Oncology, Northwestern Medical School, Chicago, IL 60611, USA
University
Contents
PREFACE
168
DEDICATION
169
CHAPTER 1
The Development of Tamoxifen
171
CHAPTER 2
Current Clinical Use of Tamoxifen
177
CHAPTER 3
Concerns About Tamoxifen
181
CHAPTER 4
New Directions in Research
187
CHAPTER 5
Molecular Mechanisms of Antiestrogen Actions
195
CHAPTER 6
Triphenylethylenes
201
CHAPTER 7
Pure Antiestrogens
217
CHAPTER 8
Targeted Antiestrogens
221
CHAPTER 9
Comparison of New Antiestrogen
REFEREiNCES
(Tamoxifen Analogs)
and Conclusions
223 225
Preface Twenty five years ago there was no tamoxifen for the treatment of breast cancer, only ICI 46,747 an antiestrogen with potent antifertility properties in laboratory rats. Today tamoxifen is the most prescribed cancer medicine and the endocrine treatment of choice for all stages of breast cancer. The low incidence of side-effects and the fact that tamoxifen increases the survival of breast cancer patients has revolutionized the approach to the treatment of breast cancer in general practice. However, the success of tamoxifen as a cancer therapy and the finding that tamoxifen can maintain bone density and lower circulating cholesterol has provided the incentive to develop new drugs for the treatment of osteoporosis and coronary heart disease. These drugs may have the added advantage of preventing breast cancer as a beneficial side-effect. This issue describes the development of antiestrogens as clinically useful agents from the time when Dr Leonard Lerner published his pioneering paper on the first non-steroidal antiestrogen MER-25 (Lerner et al., 1958) to the present. Lerner’s discovery was greeted with intense interest by the pharmaceutical industry because one of the fascinating effects of the drug was its action as a postcoital contraceptive. Regrettably MER-25 did not achieve its promise as a clinically useful antiestrogen but a successor compound MRL-41, also described by Dr Lerner (Holtkamp et ul., 1960), was successfully developed, not as a contraceptive but as an inducer of ovulation in subfertile women. MRL-41 or clomiphene became established as the therapeutic cornerstone in reproductive endocrinology, for the induction of ovulation. The reason for the intense interest in the value of antiestrogens as regulators of reproduction was because the oral contraceptive, conceived by Pincus and Chang at the Worcester Foundation for Experimental Biology (now the Worcester Foundation for Biomedical Research) in Shrewsbury, Massachusetts, had been so successful clinically. A ‘morning after pill’ would have been a valuable innovation but this was not to be. Nevertheless, the new antiestrogens, nafoxidine (Upjohn), CI 628 (Park-Davis), clomiphene (Merrel), and ICI 46,474 (ICI now Zeneca) were all investigated extensively in the laboratory throughout the 1960s and became standard laboratory reagents to investigate estrogen action. During the 196Os, there was only modest interest in cancer research in general and almost no interest in breast cancer therapeutics in particular. This was all to change in the 1970s with a ‘declaration of war on cancer’. However, several clinical trials with antiestrogens were completed in the 1960s but toxicity and an overall lack of interest by the pharmaceutical industry retarded progress. Only by a series of fortunate circumstances did ICI 46,474 become tamoxifen and this issue is dedicated to those who helped to play the key roles in this success story. 0 1997 Elsevier Science Ltd
168
Dedication Dr Leonard Lerner provided the scientific breakthrough that proved it was possible to design a compound to block estrogen action. Dr Elwood Jensen described the target site specific effects of estrogen in a laboratory model and developed the ‘estrogen receptor assay’ to predict which breast cancer patients would respond to antiestrogen therapy. Dr Jack Gorski first reported the isolation of the estrogen receptor as a soluble protein and in parallel with Jensen’s group developed intracellular models of estrogen action. Dr Arthur Walpole, head of the Reproduction Research program at Zeneca (then ICI), was the driving force behind the testing of ICI 46,474 (tamoxifen) as a therapy for advanced breast cancer. Finally Dr Bill McGui:re, whose clinical application of the steroid receptor concept, provided the rationale and target for therapeutic intervention at all stages of breast cancer. On a personal note, Dr Edward Clark, the former acting head of the Department of Pharmacology at the University of Leeds, UK deserves credit for preparing one of us (VCJ) for his subsequent career. Dr Clark was convinced that Jensen and Gorski’s reports of the isolation of a soluble receptor protein for estrogen was going to be pivotal for the future development of antiestrogens. Dr Clark is a synthetic bioorganic chemist who had a particular interest in Lerner’s work and convinced me in 1969 to read for a PhD degree in his laboratory in Leeds. My topic was to be simple. All I had to do was isolate the estrogen receptor and crystallize it with an estrogen and an antiestrogen. I was then to discover the change in shape of the complex by Xray diffraction in the Astbury Department of Biophysics. I encountered some difficulties and changed my project to the ‘structure-activity relationships of some triphenylethylenes and triphenylethanes.’ As it turned out this was a good strategic decision as no-one has yet succeeded in doing my first project! I am personally grateful to Dr Clark, my PhD supervisor, for the rigorous training and education I received. The late Dr Arthur Walpole was the examiner for my PhD in 1972 and provided me with all the opportunities to turn ICI 46,474 into tamoxifen. I met my friends and mentors Elwood Jensen (Chicago) and the late Bill McGuire (San Antonio) whilst I was a visiting scientist at the Worcester Foundation, conducting the first systematic laboratory study of tamoxifen as a antitumor agent. Their assistance and council over the past 20 years is truly appreciated. When I was recruited to develop a breast cancer research program at the University of Wisconsin in 1980: one of the reasons I accepted the challenge was the opportunity to collaborate with Jack Gorski, who is a Professor in the Biochemistry Department. Over the 1.5 years of my association with the University of Wisconsin I enjoyed tremendous help from Jack and his staff. Jack willingly served on the committees for all eleven of my PhD students at Wisconsin from Anna Riegel (n&e Tate) in 1983, who came to Madison as a Fulbright/Hays Scholar from Leeds University, to John Pink in 1995. I 169
170
V. C. Jordan and W. J. Gradishar
am extremely grateful for all the help Jack has given so much of his intellect.
to my students
and for sharing
Finally I want to thank my friend Len Lerner who sees me as a ‘younger brother.’ I am grateful for his inspiration and his desire to ‘pass the baton’ to me to realize a dream of developing antiestrogens as therapeutic agents with applications throughout medicine. Although circumstances have never permitted us to work together we were both delighted to be linked by the award of the 8th Bruce F. Cain Memorial Award by the American Association for Cancer Research in 1989. The award recognized our preclinical work that spanned 40 years, and lead to the application of antiestrogens as an effective cancer therapy.
Northwestern
Robert University
V. Craig Jordan H. Lurie Cancer Center Medical School Chicago
Top left to right: Drs Len J. Lerner, Elwood V. Jensen, Jack Gorski Bottom left to right: Drs Arthur L. Walpole, William L. McGuire, Edward R. Clark
Chapter 7
The Development of Tamoxifen
Historical Perspective In 1896 George Beatson (Beatson, 1896) demonstrated that removal of the ovaries from premenopausal women could cause the regression of breast cancer. By the turn of the century it was established (Boyd, 1900) that about l/3 of all premenopausal women with advanced breast cancer could benefit from oophorectomy and from that time, a principal strategy for the treatment and prevention of breast cancer has been either to block or to restrict the action of estradiol in its target tissue, the breast. However, the successful clinical development of antiestrogenic drugs did not initially focus on the therapy for breast cancer but drew upon expertise in several unrelated disciplines. Most of the early interest in antiestrogens was focused on reproductive endocrmology but it was clear from the beginning of clinical studies that the effects of the (drugs on cholesterol biosynthesis would play a pivotal role in assessing safety considerations for long-term therapy. Ultimately the discovery of the estrogen receptor (ER) (J ensen and Jacobson, 1962; Gorski et al., 1968) in the 1960s and the application of this basic knowledge to understand hormone-dependent breast cancer growth (McGuire et al., 1975) focused interest on the development of antiestrogens as targeted agents to block estrogen action in the tumor directly. In 1958, Lerner and coworkers reported the biological properties of the first nonsteroidal antiestrogen MER-25 (Fig. 1). The compound was found to be an antiestrogen in all species tested and was found to have no other hormonal or antihormonal properties. However, the discovery was of importance because MER-25 was found to be a post coital contraceptive in laboratory animals. Obviously one application could have been as a ‘morning after pill’ but after clinical evaluation in numerous situations the results were disappointing. MER-25 underwent initial evaluation for the induction of ovulation (Tyler et al., 1960) and the treatment of chronic cystic mastitis, breast and endometrial carcinoma (Kistner and Smith, 1960; Smith et al., 1963), but the low potency and severe CNS side-effects prohibited further clinical development (Lerner, 1981). It is relevant to point out that the antiestrogen MER-25 is a structural derivative of the cholesterol lowering drug triparanol (MER-29). In the late 1950s there was initial enthusiasm about the potential benefits of triparanol as a hypocholestremic drug (Hollander et al., 1960). However, the finding that triparanol caused an accumulation of desmosterol (Avigan et al., 1960; Gaylor, 1963; Frantz et al., 1966; Steinberg et al., 171
V. C. Jordan and W. J. Gradishar
172
1961); (an intermediate in cholesterol biosynthesis) and the linking of this biochemical effect to cataract formation (Laughlin and Carey, 1962; Kirby et al., 1962; von Sallman et al., 1963), caused withdrawal of the drug in 1962. Nevertheless triparanol was first evaluated as a potential therapy for breast cancer (Kraft, 1962) but again the results were disappointing. A successor compound to MER-25, MRL-41 or clomiphene, was a more potent antiestrogen but drug development for long-term use was to be retarded because of toxicological concerns. Clomiphene is an effective antifertility agent in laboratory animals (Holtkamp et al., 1960) but paradoxically induces ovulation in subfertile women (Greenblatt et al., 1961, 1962; Kistner, 1965). Again the prospect of developing a ‘morning after pill’ for women was not realised. Although clomiphene showed some activity in the treatment of advanced breast cancer (Herbst et al., 1964; Hecker et al., 1974), the drug was only developed for short-term use for the induction of ovulation (Huppert, 1979) because desmosterol was noted in patient sera during prolonged treatment (W. S. Merrill Company, 1967).
MER 25
r /\/“\_ P
MER 29 (TRIPARANOL)
Fig. 1.
A comparison
of the structures of the first non-steroidal antiestrogen the hypocholestremic drug triparanol (MER-29).
MER-25
and
Molecular Mechanisms and Future Uses of Antiestrogens
173
Clomiphene is marketed as an impure mixture of geometric isomers (Fig. 2) that have opposing biological activities: one isomer is an estrogen and one isomer is an antiestrogen (Palopoli et aE., 1967). Unfortunately the isomers were initially (1967) given the incorrect designation but this was corrected by 1976 (Ernst et al., 1976). Although breast cancer clinical trials were still being reported with the impure mixture of isomers in 1974 (Hecker et al., 1974), the antiestrogenic isomer eventually entered into clinical trial for breast cancer treatment but the studies were dropped by the NIH due to the interest in tamoxifen (Holtkamp, 1987). derivative In contrast, the compound nafoxidine (U- 11, lOOA) is a dihydronaphthalene that cannot isomerize (Fig. 2). The drug is a potent antiestrogen with antifertility 1965). Nafoxidine properties in laboratory animals (Duncan et al., 1963; Greenwald, exhibits antitumor properties in laboratory models (Bloom et al., 1967) but following extensive testing as a treatment for breast and renal cancer, the drug was not developed further because of unacceptable side-effects experienced by all patients (Legha et al., 1976). Although the discovery of ICI 46,474 (tamoxifen), the antiestrogenic, pure trans isomer of a substituted triphenylethylene, was made by Drs Harper, Richardson and Walpole (Harper and Walpole, 1966; Bedford and Richardson, 1966) as part of the Fertility Control Program at ICI Pharmaceuticals (now Zeneca), Cheshire, UK, the
I-
(CLOMIPHENE (mbdure of cl8 and trans isomers)
TAMOXIFEN (trans isomer only)
Fig. 2. The triphenylethylene derivatives, clomiphene and tamoxifen, that were developed into clinically useful agents in the 1960s and 1970s. The clinically unsuccessful compound nafoxidine is shown for comparison.
174
V. C. Jordan and W. J. Gradishar
study of cancer therapies was Dr Walpole’s long-term interest (Jordan, 1988). The story of ICI’s involvement with the hormonal treatment of breast cancer goes back to the early 1940s following the laboratory discovery of chlorotriphenylethylene as an orally active estrogen (Robson et al., 1938). ICI supplied the triphenylethylenes initially used by Haddow, Watkinson and Paterson in their landmark study of the antitumor effects of synthetic estrogens in advanced breast and prostate cancer (Haddow et al., 1944). These studies paved the way for the standard use of high dose estrogen therapy to treat both breast and prostate cancer for the next two decades. In 1949, Walpole and Paterson (Walpole and Paterson, 1949) studied the antitumor actions of non-steroidal estrogen therapy on breast cancer in the hope of finding the reason why some patients responded and others did not. These early studies by Walpole were unsuccessful but the subcellular mechanism of hormonal-dependent growth was eventually discovered by Dr Elwood Jensen with his pioneering work on the ER (Jensen and Jacobson, 1962). Although Harper and Walpole (1967) s h owed that tamoxifen was an antifertility agent and an antiestrogen in the laboratory rat, like clomiphene it induced ovulation in subfertile women (Williamson and Ellis, 1973). Fortunately preliminary clinical studies demonstrated efficacy for tamoxifen in the treatment of advanced breast cancer (Cole et al., 1971, 1972; Ward, 1973) and the stage was now set for the successful development of tamoxifen as the first clinically acceptable antiestrogen for the treatment of breast cancer.
Clinical Development Tamoxifen (Nolvadexm) had several advantages at the outset: (1) higher antitumor potency compared with other compounds being evaluated at the time (Table 1); (2) a low reported incidence of side-effects for the drug itself (Ward, 1973); (3) a lower incidence of side-effects compared with other endocrine therapies available to the clinician (i.e. high-dose estrogens or androgens (Cole et al., 1971)); and (4) the laboratory finding (Harper and Walpole, 1967), confirmed in the clinic (Cole et al., 1972) that desmosterol levels were not affected by tamoxifen. Long-term therapy (i.e. years) could be considered without the fear of cataract formation. This has proved to be true after 20 years of usage. The evolution of strategies to determine the optimal application of tamoxifen for the treatment of breast cancer has taken more than 20 years but the process has been facilitated by a dialogue between the laboratory and the clinical trials community. The principal areas of contact are illustrated in Fig. 3. The clinical development of tamoxifen from the first preliminary clinical reports, suggesting its efficacy as an agent for the palliation of advanced breast cancer in postmenopausal women, to the present has been extensively reviewed (Furr and Jordan, 1984; Lerner and Jordan, 1990; Jordan, 1993; Jaiyesimi et al., 1995; Osborne, in press). However, the laboratory studies investigating the biology of tamoxifen have played, and continue to play, a pivotal role in the clinical investigation of the treatment strategies, concepts and concerns with antiestrogens. Long-term adjuvant therapy (i.e. greater than 1 year) (Jordan, 1978, 1983, 1994; Jordan et al., 1979), prevention (Jordan, 1974, 1976; Jordan et al., 1991), the association of tamoxifen with endometrial cancer (Satyaswaroop et
Molecular Mechanisms Table 1. Preliminary
and Future Uses of Antiestrogens
clinical trials of antiestrogens
for the treatment
of metastatic
Total patients (% response)
Toxicities
Compound Reference
Daily dose
Triparanol, Kraft (1962) MER-2.5, Kistner and Smith (1960) Clomiphene, Herbst et al. (1964) Hecker et al. (1974) Nafoxidine, Legha et al. (1976)
250-1000 500-4500 100-300
8 (11) 4 (25) 56 (34)
180-240
198 (31)
20-40
114 (31)
(mg)
TamoxiFen, Cole et al. (1971) Ward (1973)
175 breast cancer
None” Acute psychotic episodeb None reported or mild’
Bilateral cataracts Ichthyosis Cutaneous photophobiad Transient thrombocytopenia’
“Withdrawn from the market by the William S. Merrel Co. in cooperation with the FDA in April 1962. hDoes not include patients treated by Dr Roy Hertz when therapy was stopped due to hallucinations (Lerner, 1981). ‘Visual rsymptoms (W. S. Merrill Company, 1967). ‘Eighty to one hundred percent of patients affected (Legha et al., 1976). “The particular advantage of this drug is the low incidence of side effects’ (Cole et al., 1971) ‘The side effects were usually trivial’ (Ward, 1973).
al., 1984; Gottardis et al., 1988), carcinogenesis and tamoxifen (Han and Liehr, 1992; Williams et al., 1993; Greaves et al., 1993) and the targeting of novel antiestrogens to prevent osteoporosis (Jordan et al., 1987; Lerner and Jordan, 1990) are all laboratory concepts that have had a profound impact on the clinical applications of
CLINICAL TESTS TAMOXIFEN Advanced Disease Adjuvant Single Agent
Tamoxifen
Pure Antiestrogens PREVENTION Raloxifene for Osteoporosis
High Affinity Preserve Bone Long Term Tamoxlfen
Pure Antiestrogens
LABORATORY TESTS Fig. 3. The development of tamoxifen during the 1970s with laboratory results being translated into clinically useful treatment strategies culminating in the current evaluation of tamoxifen as a preventive. An investigation of tamoxifen’s pharmacology in the laboratory establislhed the concept of antiestrogens with high affinity for the estrogen receptor, pure antiestrogens and the target site specific actions on bones that has culminated in the testing of raloxifene to treat and prevent osteoporosis.
176
V. C. Jordan and W. J. Gradishar
The enormous clinical success of tamoxifen and the discovery of its target site-specific actions has provided an enormous incentive to develop new and improved compounds with applications to treat breast cancer and diseases associated with the menopause (Tonetti and Jordan, 1996). antiestrogens.
We will first describe the effectiveness of tamoxifen in the treatment of breast cancer and focus on the potential concerns that have been raised about the use of extended durations of adjuvant tamoxifen therapy and the testing of tamoxifen as a breast cancer preventive in high risk women. The broad application of tamoxifen has encouraged far ranging studies of its toxicology and side-effects. At present there are several concerns regarding tamoxifen, but there is little clear cut clinical evidence or description of serious unpredicted side-effects, to support the hypothetical toxicities. Nevertheless with the reported concerns in mind we will describe the properties of an ideal antiestrogen to be used as a preventive agent, a long-term adjuvant therapy or as a treatment for osteoporosis. We will then evaluate each available new compound, based upon the criteria required for the ideal therapeutic agent, so that clinicians can make an objective assessment of the information that is used to support the claims for new antiestrogens currently under investigation.
Chapter2
Current Clinical Use of Tamoxifen
Tamoxifen is the endocrine treatment of choice for all stages of breast cancer. There are currently 8 million women years of clinical experience with the drug and it is the world’s most prescribed cancer medicine. In 1973 Nolvadex@, the ICI brand of tamoxifen was approved for the treatment of breast cancer by the Committee on the Safety of Medicines in the United Kingdom. Similar approval was given in the United States for the treatment of advanced disease in postmenopausal women by the Food and Drug Administration (FDA) on 30 December 1977. Tamoxifen was approved for the treatment of advanced ER-positive breast cancer in premenopausal women in 1989. With more than 20 years of clinical experience world-wide, tamoxifen is established as the endocrine therapy of choice for metastatic breast cancer in pre- and postmenopausal patients. Extensive testing as an adjuvant therapy and proven efficacy to enhance survival (NATO, 1983, 1988; Fisher et al., 1983, 1986, 1989; Breast Cancer Trials Committee, Scottish Trials Office, 1987) lead to FDA approval for the use of tamoxifen as an adjuvant therapy with chemotherapy (1985), as an adjuvant therapy alone (1986) in node-positive, postmenopausal patients and pre- and postmenopausal patients with ER-pomsitive, node-negative disease (1990). Tamoxifen was approved for the treatment of male breast cancer in 1993. The currently accepted strategy for the adjuvant treatment of breast cancer is to employ at least 5 years of therapy. This duration is the standard used in clinical trials in the United States. However, based upon theoretical considerations and its safety profile., tamoxifen treatment has been used until relapse in both a clinical trials setting (Falkson et al., 1990; Tormey et al., 1992) and in clinical practice on a case by case basis. If all the published clinical trials data kept are considered it is not possible to define the optimal duration of tamoxifen treatment (Bilimoria et al., 1996). Nevertheless a recent clinical alert from the National Cancer Institute has recommended stopping tamoxifen at 5 years in ER-positive, node-negative women because no improvement can be detected in an National Surgical Adjuvant Breast and Bowel Program (NSABP) study by continuing tamoxifen to 10 years. However, we believe that additional published data are required before rigid guidelines can be establi’shed. Furthermore, the small Scottish trial of 5 years versus indefinite 177
178
V. C. Jordan and W. J. Gradishar
tamoxifen shows no benefit for indefinite therapy but strongly encourages further evaluation in clinical trial (Stewart et al., 1996). The Cancer Research Campaign in the United Kingdom has opened a registration trial (ATTOM) of stopping tamoxifen trials at 2 years or continuing for at least 5 additional years. Similarly the clinical trials center in Oxford is preparing a study, ATLAS, with sites around the world, to define the optimal duration of adjuvant tamoxifen treatment. Despite our inability to define the optimal duration for tamoxifen treatment under all circumstances it is reassuring that the reporting of major toxicities has remained so low during a period when the duration of tamoxifen treatment has not been restricted (Jordan, 1994).
The success of tamoxifen as a therapeutic agent has been demonstrated convincingly through the Oxford overview analysis of randomized clinical trials (Early Breast Cancer Trialists’ Collaborative Group, 1988, 1992). However, the direct investigation of potential side-effects has demonstrated additional benefits for patients treated with tamoxifen that have ultimately resulted in the widespread testing of new antiestrogens as potential preventives for osteoporosis, coronary heart disease, breast and endometrial cancer (Lerner and Jordan, 1990; Jordan, 1995a; Tonetti and Jordan, 1996).
Additional Benefits of Tamoxifen A decade ago when the strategy of long-term tamoxifen therapy was being incorporated into treatment arms of all clinical trials using tamoxifen, preliminary studies were undertaken to evaluate the possibility that an ‘antiestrogen’ might have a detrimental effect on bone density and also place patients at risk for coronary heart disease. However, it was known that tamoxifen is not a pure antiestrogen but has estrogen-like effects in animals and postmenopausal women (Furr and Jordan, 1984; Lerner and Jordan, 1990; Jordan, 1993). Tamoxifen maintains bone density in et al., 1987, 1988) and has been ovariectomized rats (Jordan et al., 1987; Turner shown to have either a beneficial effect in maintaining bone density in the lumbar spine and neck of the femur in postmenopausal women (Turken et al., 1989; Love et al., 1992, 1994; Ward et al., 1993; Kristensen et al., 1994) or have no detrimental effects (Fornander et al., 1990). These data have been summarized recently (Bilimoria et al., 1996) and suggest that tamoxifen has an estrogen-like effect on maintaining bone density in postmenopausal women. Similarly, tamoxifen lowers circulating cholesterol. However, high density lipoprotein (HDL) cholesterol levels are unaffected (Bertelli et al., 1988; Love et al., 1991, 1995) and only low density lipoprotein (LDL) cholesterol is reduced. Overall these data suggest that tamoxifen could lower the incidence of coronary heart disease if long-term treatment schedules are used. Preliminary studies from the Scottish trial demonstrate that 5 years of tamoxifen will reduce the risk of fatal myocardial infarction (McDonald and Stewart, 1991) and there is a reduced incidence of hospitalizations from any cardiac condition in the Stockholm trial (Rutqvist and Matteson, 1993). A recent update of the Scottish Trial (McDonald ef al., 1995) has shown a clear decrease in coronary heart disease if women have ever taken tamoxifen. However, most protection is seen for current users.
Molecular
Mechanisms
and Future Uses of Antiestrogens
179
Antiestrogens and Lipids: Molecular Mechanisms There is intense interest in the ability of antiestrogens to decrease coronary heart disease and two lines of investigation have demonstrated possible mechanisms. First, the decrease in circulating cholesterol has recently been shown to be caused by a block in the conversion of delta 8 cholestenol to lathosterol (Gylling et al., 1995). (Fig. 4). Both tamoxifen and toremifene cause an increase in circulating delta 8 cholestenol but there is very little effect on the conversion of desmosterol to cholesterol. This is the primary mechanism for triparanol blocking cholesterol biosynthesis that was associated with cataract formation in treated patients (Avigan et al., 1960). Some of the beneficial effects of tamoxifen may be related to its antioxidant ability. Tamoxifen protects rat cardiac membranes from damage caused by lipid peroxidation (Wiseman et al., 1993a). Oxidative damage to LDL is also thought to play a role in the development of atherosclerosis and Wiseman et al. (1993b) have shown that tamoxifen and its metabolites can protect human LDL against copper-ion dependent lipid peroxidation.
Contralateral Breast Cancer If the beneficial effects of tamoxifen on bones and cholesterol are expressions of the estrogen-like effects of tamoxifen, the effects of tamoxifen on reducing contralateral breast cancer must be considered to be an expression of the antiestrogenic/antitumor properties of the drug. The incidence of second primary breast cancer in patients treated with tamoxifen is reduced by approximately 40% compared to
Lanosterol
Dihydrolanosterol + AS-dimethylsterol 1 A*-monomethylsterol + A*-cholestenol X+Lathosterol
TAMOXIFEN & TOREMIFENE
4
Desmosterol
Cholesterol Triparanol
Fig. 4.
The inhibition
of cholesterol biosynthesis by tamoxifen, Adapted from Gylling et al. (1995).
toremifene
and triparanol.
180
V. C. Jordan and W. J. Gradishar
not receiving tamoxifen (Fisher et al., 1989; Early Breast Cancer Trialists’ Collaborative Group, 1992; Fornander et al., 1989). A significant reduction in contralateral breast cancer (0.5 matched odds ratio) for patients receiving 2 years of adjuvant tamoxifen has recently been reported by Cook et al. (1995) based on an epidemiology study. patients
In summary tamoxifen has been shown to produce significant benefits to patients as an anticancer agent by increasing patient survival and reducing the risk of developing contralateral breast cancer (Early Breast Cancer Trialists’ Collaborative Group, 1992). Furthermore, the estrogen-like effects of tamoxifen appear to provide benefits to postmenopausal patients who would normally be denied a hormone replacement therapy to prevent osteoporosis or coronary heart disease (Bilimoria et al., 1996). Nevertheless there are legitimate concerns about the potential toxicities associated with long-term tamoxifen treatment that have required consideration by the clinical community.
Chapter 3
Concerns About Tamoxifen
During the past half decade several important aspects of the pharmacology of tamoxifen have emerged that have had an impact on the clinical use of the drug. There are three principal areas of concern. First, an increased incidence in endometrial cancer has been reported (Fornander et al., 1989; Fisher et al., 1994) in tamoxifsen treated patients and one study found higher grade disease and patients with a poorer prognosis associated with tamoxifen treatment compared to patients with de nova disease (Magriples et al., 1993). Based on laboratory evidence (Satyaswaroop et al., 1984; Gottardis et al., 1988), an increased detection of endometrial cancer could be ascribed to the estrogen-like effects of tamoxifen. Second, laboratory studies have shown that tamoxifen produces liver tumors in rats (Williams et al., 1993; Greaves et al., 1993) and concerns have been raised that tamoxifen could produce second primary tumors in the liver, stomach, colon and rectum of women (Rutqvist et al., 1995). Finally, laboratory evidence (Osborne et al., 1987; Gottardis and Jordan, 1988; Gottardis et al., 1989a; Wolf et al., 1993) shows that tamoxifen-stimulated breast tumors can develop so the antitumor action of long-term adjuvant tamoxifen therapy could eventually fail. Tamoxifen might ultimately encourage tumor growth in patients. However, each of these concerns must be placed in perspective based on the current clinical data base of 8,000,OOO woman-years of experience accumulated from the worldwide use of tamoxifen in patients for nearly a quarter of a century.
Endomietrial Cancer Much controversy has surrounded the associations between tamoxifen use and the detection of endometrial cancer. However, it is now possible to provide a reasonable picture of the actual incidence of endometrial cancer and provide a balanced view of the concerns. Recent reviews (Assikis and Jordan, 1995; Assikis et al., 1996; Jordan and Assikis, 1995) of the literature have only identified about 400 cases of endometrial cancer associated with tamoxifen use world-wide and our original reviews (Assikis and Jordan, 1995; Jordan and Assikis, 1995) has now been updated to the end of 1995 in Table 2. The disease is found predominantly in postmenopausal women and there is not a strong association between the duration of tamoxifen use and the risks of developing endometrial carcinoma. Indeed it is interesting to re-evaluate earlier studies that claim an association between long-term tamoxifen and 181
182
V. C. Jordan and W. J. Gradishar Table 2. The incidence and characteristics of endometrial tumors associated with the use of tamoxifen. The cases accumulated are those reported up to the end of 1995
Endometrial carcinomas Premenopausal patients Postmenopausal patients Stage 1 disease Good Grade (1+2) More than 2 years tamoxifen Less than 2 years tamoxifen
349 2 200 184/234 (79%) 185/225 (82%) 108 91
The information is reported in full elsewhere (Assikis et al., 1996). It should be noted that disease and patient characteristics are not provided by the reporting authors uniformly.
endometrial cancer. Re-analysis of the Stockholm study (Fornander et al., 1989) that originally concluded that randomization to 2 years of adjuvant tamoxifen did not cause an increase in endometrial cancer, but randomization to 5 years of adjuvant tamoxifen caused a six-fold increase in the risk of endometrial cancer actually demonstrates that 12 of 16 received only 2 years of drug (Jordan and Morrow, 1994). Clearly pre-existing disease was being detected. Based on the known long genesis of cancer in humans it would be inappropriate to suggest that early detection of endometrial cancer was caused by short courses of tamoxifen. It is also important to point out that DNA adducts are absent from uterine samples of patients taking tamoxifen (Carmichael et al., 1996). Detection bias, through the investigation of symptoms, is almost certainly responsible for the disease found in many patients receiving tamoxifen. It is important to appreciate that epidemiology studies do not show a statistically relevant increase in the incidence of endometrial cancer after a short (2 year) course of tamoxifen (Cook et al., 1995; van Leeuwen et al., 1994). The finding, by Magriples et al. (1993), that tamoxifen use is associated with poor prognosis disease has not been confirmed by any other study (Fisher et al., 1994; Rutqvist et al., 1995; Barakat et al., 1994). Overall, the stage and grade of endometrial cancer associated with tamoxifen use is proportionally the same as Surveillance, Epidemiology, and End Results (SEER) data (Assikis and Jordan, 1995). Therefore, it is fair to say that the overall consensus is that the benefits of tamoxifen in the treatment of breast cancer far outweigh the risks associated with a two-fold elevation in early stage low grade endometrial carcinoma (Jaiyesimi et al., 1995; Osborne, in press; Bilimoria et al., 1996; Early Breast Cancer Trialists’ Collaborative Group, 1992; Jordan, 1995b). However, as a precaution, patients should be screened to determine whether they have pre-existing endometrial carcinoma before starting a course of adjuvant tamoxifen therapy. Additionally patients who present with spotting and bleeding during treatment must be investigated with a thorough gynecological examination. There is, however, no justification for an extensive screening program to detect endometrial cancer in asymptomatic women taking tamoxifen (Barakat, 1997).
Molecular
Mechanisms
and Future Uses of Antiestrogens
183
Hepatocellular Carcinoma and Other Malignancies Several investigators report that tamoxifen is both an initiator and a promoter of rat liver carcinogenesis (Williams et al., 1993; Greaves et al., 1993; Hard et al., 1993; Dragan et al., 1994, 1995, 1996). Tamoxifen, at high doses, causes DNA adducts in rat liver (Han and Liehr, 1992; Hard et al., 1993; White et al., 1992). However, only low adduct formation is noted in mouse liver DNA, (White et al., 1992) a species that does not produce tumors in response to high daily doses of tamoxifen (Furr and Jordan, 1984). It is also reassuring to note that there is no increase in DNA adduct formation in the livers of patients receiving tamoxifen (Martin et al., 1995). As a result, it has been argued that the rat studies are not relevant to human usage (Jordan and Morrow, 1994; Jordan, 1995b, 199%). An exarnination of the data from the rat carcinogenesis studies demonstrate that the animals receive tamoxifen (S-SO mg/kg daily) from puberty for more than 50% of their life (Jordan and Morrow, 1994). In contrast, the therapeutic dose of tamoxifen, as an anticancer agent in rats, is 250 pg/kg (Jordan, 1983) which is comparable to the therapeutic dose in a 70 kg patient of 285 ,ug/kg or 20 mg of tamoxifen administered twice daily. The duration of adjuvant therapy for postmenopausal patients is usually 5 years. This would be equivalent to 8% of a woman’s life. Thus, the animal experiment at the lowest dose to produce tumors, 5 mg/kg, is equivalent to a teenage girl (i.e. 14 years of age) receiving 20 times the daily dose of tamoxifen until she is 40 years old. This is 40 tablets a day. The reason that such large doses have to be administered to the rat to produce drug levels comparable to the human is that the drug is cleared from the rat ten times faster than in humans (Jordan and Morrow, 1994). Thus, artificially high levels of drug must be given, far outside the therapeutic range, that ultimately cause damage in the rat liver. In recent years, concerns about carcinogenesis with tamoxifen have lead to a report of increases in co10 rectal cancer and stomach cancer (Rutqvist et al., 1995). These results have not been supported by either individual reports from clinical trials (Fisher et al., 1994) or from the current (1996) Oxford overview analysis. The finding of liver carcinogenesis in the rat would be cause for concern with any new drug that is about to go into clinical trial. However, tamoxifen had been used extensively for 20 years before the investigation of rat liver carcinogenesis. There has not been a significant increase in hepatocellular carcinoma since the two initial cases reported. in 1989 (Fornander et al., 1989). Similarly epidemiology studies (Muhleman et al., 1994) have not shown a rise in hepatocellular carcinoma in breast cancer patients since tamoxifen was approved for use in the United States in 1978. In contrast., oral contraceptives cause a ten-fold increase in the risk for the development of hepatocellular carcinoma (Prentice, 1991), but this risk is considered to be acceptable to regulatory authorities because of the rarity of the disease.
Molecular Mechanism of Carcinogenesis During the past 5 years there has been intense interest in discovering event for tamoxifen-induced rat liver carcinogenesis and determining
the initiating the relevance
184
V. C. Jordan and W. J. Gradishar
for humans. Han and Liehr (1992) first noted an accumulation of DNA adducts in the liver of Sprague-Dawley rats on repeated injections of 20 mg/kg (cf. human dosage of 0.3 mg/kg). This has been adequately confirmed by numerous investigators and the focus of investigation has been the identification of the actual DNA adduct. Several candidates have been proposed: an epoxide (Styles et al., 1994; Lim et al., 1994; Phillips et al., 1994a), 4-hydroxytamoxifen (Randerath et al., 1994; Moorthy et al., 1996), Metabolite E (Pongracz et al., 1995) or cr-hydroqamoxifen (Potter et al., 1994; Phillips et al., 1994b, 1994~). Recently, Osborne et al. (1996) prepared a-acetoxytamoxifen which is able to react with DNA to a greater extent (1 in 50 bases) than a-hydroxytamoxifen (1 in lo5 DNA bases). The products of the reaction were identical to those isolated from DNA of rat hepatocytes or the livers of rats treated with tamoxifen. The adduct of tamoxifen and DNA has been identified at the nucleoside deoxyguanosine in which the a position of tamoxifen is linked covalently to the exocyclic amino of deoxyguanosine (Fig. 5). These important observations have provided a framework to study the metabolic activation of tamoxifen in human systems and to identify any DNA adducts in human tissues. The metabolic activation of tamoxifen and its metabolite cc-hydroxytamoxifen has been compared using primary cultures of rat, mouse and human hepatocytes (Phillips et al., 1996a). Although DNA adducts are readily identified in rat and mouse
Fig. 5. The formula of cc-hydroxytamoxifen (I), a-acetoxytamoxifen (II) and the adduct formed between a-hydroxytamoxifen and deoxyguanosine (III) Adapted from Osborne er al. (1996).
Molecular Mechanisms and Future Uses of Antiestrogens
185
hepatocytes (90 and 15 adducts/lOa nucleotides, respectively), DNA adducts were not detected in tamoxifen-treated human hepatocytes. Additionally human hepatocytes also appeared to produce SO-fold lower levels of a-hydroxytamoxifen from tamoxifen compared to rat hepatocytes. Further studies showed that if cells were treated with had 300-fold lower levels of adducts a-hydroxytamoxifen human hepatocytes compared to rat hepatocytes. Studies in humans have confirmed that the human is not as susceptible as the rat to DNA adduct formation with tamoxifen. The pattern of DNA adducts found in the rat liver is not found in humans treated with tamoxifen (Martin et al., 1995), DNA adducts are not found in lymphocytes (Phillips et al., 1996b) and there is a lack of genotoxicity of tamoxifen in human endometrium (Carmichael et al., 1996). In the latter studies, DNA adducts could be produced in endometrial samples with a-hydroxytamoxifen but not with tamoxifen. The authors proved that tissue was capable of metabolizing tamoxifen to cr-hydroxytamoxifen but apparently it is incapable of producing adducts. Endometria from patients taking tamoxifen for up to 9 years were analyzed for DNA adducts. No evidence for any DNA adducts induced by tamoxifen was found in any of the patients examined. The authors concluded that the genotoxic events observed with tamoxifen in the rat may not apply to the human endometrium (Carmichael et al., 1996). This conclusion supports the previous suggestl.on that tamoxifen, or indeed any new antiestrogen which has partial agonist actions, will cause the activation and detection of pre-existing disease (Jordan and Morrow, 1994).
Drug Resistance Drug resistance to tamoxifen therapy can take many forms (Morrow and Jordan, 1993; Tonetti and Jordan, 1995). Obviously if tumors are ER-negative there is only a small probability of a response to antiestrogen therapy. In the case of metastatic breast cancer about 10% of ER- and progesterone receptor (PR)-negative patients respond to any form of endocrine modulation (Jordan et al., 1988). Similarly the overview analysis (Early Breast Cancer Trialists’ Collaborative Group, 1992) of clinical trials suggest that postmenopausal, node-positive patients with receptor poor disease will only benefit from adjuvant tamoxifen with a small survival advantage compared with highly receptor-positive disease. Adjuvant tamoxifen produces the best survival benefits in patients with receptorpositive disease, however, the duration of therapy may have a profound effect on the overall effectiveness of treatment. One goal of current clinical studies is to determine the optimal duration of tamoxifen. Two (NATO, 1983) and five (Breast Cancer Trials Committee, Scottish Trials Office, 1987) years of tamoxifen treatment will produce a survival advantage in unselected postmenopausal patients with predominately nodepositive disease. However, the emergence of drug resistance to tamoxifen will limit the effectiveness of indefinite tamoxifen treatment (Bilimoria et al., 1996). It is interesting to note that the NSABP B-14 trial evaluating 5 versus 10 years of tamoxifen in node-negative patients, was recently stopped, not because significant drug resistance was observed with 10 years of therapy, but because 10 years of therapy
166
produced economic
V. C. Jordan and W. J. Gradishar
no significant benefit over 5 years of therapy. The issue, therefore, and a consideration of a possible increase in iatrogenic diseases.
becomes
Nonetheless, there are ongoing clinical trials in place in the United Kingdom (ATTOM and ATLAS) that are addressing the duration of tamoxifen in large patient populations. Each study is recruiting 20,000 patients to establish guidelines for the duration of tamoxifen therapy in node-positive and node-negative populations. In the laboratory, tamoxifen will inhibit estrogen-stimulated growth of MCF-7 breast tumors implanted into athymic mice (Osborne et al., 1985). Nevertheless, continuous therapy with tamoxifen results in the emergence of tamoxifen-stimulated breast tumors that will grow in response to either estrogen or tamoxifen (Osborne et al., 1987; Gottardis and Jordan, 1988; Gottardis et al., 1989a; Wolf et al., 1993). Since there are clinical reports of tamoxifen-stimulated tumors that have a withdrawal response to tamoxifen (Canney et al., 1987; Howell et al., 1992), new second-line agents (or first-line agents) are necessary to control tumors that grow after extended tamoxifen treatment.
Chapter 4
New Directions in Research
The Ideal Agents for Therapeutic Evaluation There are three distinct goals for drug discovery to exploit the current therapeutic applications of antiestrogens. The opportunities are illustrated in Fig. 6. Tamoxifen is being used experimentally as a preventive for breast cancer in high risk women and
BIOLOGY INITIATION
ANTIESTROGEN TREATMENT STRATEGY
1
-
? PRGMOTlON WllH ESTROGEN
I I I I I I
+
@ DETECTION
--)
@O@
O@O O@
-
SURGERY
@O
PURE ANllESTRGGENS
w Fig. 6. Treatment opportunities with antiestrogens. New antiestrogens could be discovered that do not develop antiestrogen-stimulated tumor growth and are not cross-resistant with tamoxifen. Pure antiestrogens could be used for advanced disease but targeted antiestrogens could be used as preventives for breast cancer and as specific hormone replacement therapies for post-menopausal women. 187
188
V. C. Jordan and W. J. Gradishar
also as a treatment for all stages of breast cancer. One strategy is to develop agents that are not cross-resistant with tamoxifen and have a safer toxicity profile. Another strategy is to develop a pure (i.e. non-estrogenic) antiestrogen that does not have stimulatory effects in the uterus and does not cause premature drug resistance. These agents could find use as second-line therapies after the failure of tamoxifen or as optimal estrogen antagonists for the first-line treatment of metastatic breast cancer. Finally, an ideal agent for adjuvant therapy, and as a preventive, could be designed to exploit the beneficial effects of tamoxifen on bones and lipids but solve the problems suggested by an increased detection of endometrial cancer and the theoretical concerns about DNA adduct formation in rat liver. However, quality of life issues will become of primary importance for the successful application of antiestrogens in women without a diagnosis of breast cancer. Clearly, if a new agent is unpleasant to take, compliance will be a problem. As a result a critical area for research consideration must be the control of hot flashes and menopausal symptoms; a feeling of well-being is essential in any new agent. A compound that mimics estrogen in the brain would be a major advantage for patient compliance and in the long run, an estrogen-like effect may also prevent Alzheimers syndrome. (Tang et al., 1996). The properties of an ideal agent that could be employed more widely, for example as a hormone replacement therapy, or a prevention maintenance therapy in postmenopausal women, are illustrated in Fig. 7. If these properties can be achieved,
Pfopertiesofldeal TargetedAntiestrogens - rnk*
Value of ideal Targeted Andestogens Eliminate6 hd Hashea
Fig. 7. Properties of an ideal targeted antiestrogen to be developed as a new hormone replacement therapy, or prevention maintenance therapy, for postmenopausal women.
Molecular
Mechanisms
and Future Uses of Antiestrogens
189
the agents could be applied to a host of novel situations throughout medicine as treatments and preventatives for osteoporosis and coronary heart disease. To this end we will describe the progress that has been made during the past decade to achieve the three separate goals: the development of a safer antiestrogen, a pure antiestrogen and a targeted antiestrogen.
Discovery of New Antiestrogens The intense investigation of the pharmacology of tamoxifen during the 1970s and 1980s established a data base for the development of new compounds to treat breast cancer and for broader therapeutic applications. During the 1960s and 1970s all the availablle antiestrogens were noted to have a low affinity for the ER (Jordan, 1984). This observation lead to the widely held belief that antiestrogens had to be compounds with low affinity for the ER so that the receptor complex would break up before the functions required for estrogen action could be completed (Jordan, 1984). The compounds were thought of as weak estrogens that inhibited full estrogen action by saturating all available ER. The discovery that the metabolites of tamoxifen, 4-hydroxytamoxifen and 3,4_dihydroxytamoxifen, have a high affinity for the ER and inhibit full estrogen action, changed the understanding of antiestrogen action (Jordan
METABOLITE
MIMICRY N’
P-’
4-HYDROXYTAMOXIFEN
DROLOXIFENE
Fig. 8. The mono- and dihydrovlated been copied to develop the antiestrogen
antiestrogenic metabolites of tamoxifen that have TAT-59 (a pro drug) and droloxifene (3-hydroxytamoxifen).
190 et al.,
V. C. Jordan and W. J. Gradishar
Receptor binding and biological in the same molecule.
1977).
functions
activity
are now viewed
as two separate
The principle of an antiestrogen with high affinity for the ER has been exploited with the antiestrogens, TAT-59 and 3-hydroxytamoxifen (droloxifene), that mimic the metabolites of tamoxifen (Fig. 8). TAT-59 is a phosphorylated derivative of 4-hydroxytamoxifen that requires dephosphorylation to the active antiestrogen (Toko et al., 1992). In contrast, droloxifene is derived from the tamoxifen metabolite 3,4_dihydroxytamoxifen. This metabolite of tamoxifen is unusual because it has antiestrogenic activity in both rat and the mouse uterine weight tests (Jordan et al., 1977, 1978), whereas tamoxifen and 4-hydroxytamoxifen are both classified as estrogen in short-term mouse assays (Jordan et al., 1977). 3,4_Dihydroxytamoxifen is very unstable and is readily deactivated by the enzyme catechol ortho methyl transferase so this tamoxifen metabolite would be unpromising as a therapeutic agent (Jordan et al., 1984). However, removal of the 4-hydroxy to produce droloxifene retains antiestrogenic properties (Roos et al., 1983) and, most importantly, antitumor properties. The Eli Lilly Company has made triphenylethylene structure but
a systematic study of compounds that avoid the retain potent antiestrogenic properties. The
NAFOXIDINE
TRIOXIFENE Fig. 9.
A comparison
of the structures
of nafoxidine
and trioxifene.
Molecular Mechanisms and Future Uses of Antiestrogens
191
antiestrogen trioxifene was discovered (Jones et al., 1979) during the late 1970s and a comparison of the structure of trioxifene with nafoxidine illustrates that the main differe:nce is a methanone bridge that alters the triphenylethylene-like structure (Fig. 9). Trioxifene is active as an antitumor agent in the laboratory (Rose et al., 1981) and in the .treatment of advanced breast cancer (Manni et al., 1981; Witte et al., 1986; Lee et al., ‘1986), but it does not have the severe side-effects noted earlier with nafoxidine (Legha et al., 1976). Although the drug showed some promise it has not been developed as a clinically useful agent because side-effects were believed to be a potential problem when compared to tamoxifen.
In contrast to tamoxifen the novel antiestrogens, LY117018 and LY156758 (Fig. lo), have a high affinity for the ER and are less estrogenic in the rat and mouse uterus than tamoxifen (Black et al., 1983; Jones et al., 1984; Jordan and Gosden, 1983a, 1983b) as well as potent inhibitors of estrogen-stimulated effects in vitro (Lieberman et al., 1.983; Scholl et al., 1983; Poulin et al., 1989). The compounds exhibit antitumor properties in animal models but are not superior to tamoxifen (Clemens et al., 1983; Wakeli,ng and Valcaccia, 1983; Gottardis and Jordan, 1987). However, and perhaps most importantly, the weakly estrogenic, antiestrogen keoxifene (LY1.56758) was found to maintain bone density in the ovariectomized rat (Jordan et al., 1987). The
4-HYDROXYTAMOXIFEN (1977)
2) /
0
LY117018 (1980)
r/0
0
LY156,758
LY 139,481
KEOXIFENE (1982)
RALOXIFENE (1994)
Fig. 10. The development of the novel, weakly estrogenic, antiestrogen LY 117018 from the tamoxiffzn metabolite 4-hydroxytamoxifen Subsequent structure-activity relationships improved the estrogenic/antiestrogenic ratio while retaining high affinity for the estrogen receptor.
V. C. Jordan and W. J. Gradishar
192
drug has subsequently been renamed treatment of osteoporosis following 1994; Sato et al., 1995) of the original
raloxifene and has entered clinical trials for the confirmation (Evans et al., 1994; Black et al., animal studies (Jordan et al., 1987).
The possibility that a pure antiestrogen could be developed with high binding affinity for the ER combines the observation that MER-25, the first antiestrogen, has virtually no estrogenic properties in any animal species (Lerner et al., 19.58), with the knowledge that binding affinity and biological activity are separate functions of the same molecule (Jordan et al., 1977). The antiestrogens ICI 164,384 and ICI 182,780 are derivatives of estradiol with an optimal binding affinity for the ER, but these structural analogues are unique because they do not have any estrogenic properties
Alkylating Antiestrogens
Affinity Chromatography
Long Alkylating Side Chains
Pure Antiestrogens
Laboratory Studies
HO
I
I
OH
ICI164,364
0 OH
Clinical Studies
HO
ICI 182,780 Fig. 11. The discovery of pure antiestrogens occurred via an investigation of 6,7 substituted alkylating derivatives of estradiol and attempts to purify the estrogen receptor using affinity chromatography linked to estradiol through the 7-a position. ICI 164,384 has been used extensively in laboratory studies to describe the unique pharmacology of this class of drugs. ICI 182,780 is being evaluated clinically to treat advanced breast cancer.
Molecular
Mechanisms
and Future Uses of Antiestrogens
193
and they have a novel subcellular mechanism of action (Wakeling, 1994) (see below). The serendipitous discovery of pure antiestrogens occurred through two essentially unsuccessful research endeavors that converged thus providing the optimal intellectual environment for new drug discovery (Fig. 11). Derivatives of estradiol or estrone substituted in the 6 and 7 position were being evaluated as potential alkylating antiestrogens in the late 1970s through an ICI-Leeds University joint research scheme (Jordan et al., 1981). Independently, scientists in France were attempting to purify the ER using estradiol linked at the 7 position through a tenmembered carbon side chain to Sephadex columns (Bucourt et al., 1978). Dr Alan Wakeling brought both of these independent ideas together to discover the structurefunction relationships of a new class of compounds that have no estrogenic properties in any test system (Wakeling and Bowler, 1987; Bowler et al., 1989; Wakeling et al., 1991). The pure antiestrogen ICI 164,384 has been used extensively in laboratory studies (Wakeling, 1994), but the more potent ICI 182,780 (Wakeling et al., 1991) is
TAMOXIFEN o-N< I
TOREkFENE
IDOXIFENE Fig. 12.
Substituted
tamoxifen
analogs.
194
V. C. Jordan and W. J. Gradishar
being evaluated as a second-line therapy for the treatment of breast failure of long-term adjuvant tamoxifen (Howell et al., 1995).
cancer
after the
Finally, there has been a search for the past 1.5 years for tamoxifen derivatives that might have less clinical toxicity (Fig. 12). T oremifene, or chlorotamoxifen (Kangas et al., 1986; Kangas, 1990a), is less potent than tamoxifen as an antiestrogen (DiSalle et al., 1990) and an antitumor agent (Robinson et al., 1988) and this has translated into higher daily doses of toremifene being used in clinical trials to treat advanced breast cancer (Hayes et al., 199.5). The most interesting feature of the toxicology is that the compound does not produce liver tumors in rats (Hard et al., 1993; Hirsimaki et al., 1993). Clearly this property of toremifene could become important if tamoxifen is proven to be carcinogenic in the human liver. In contrast to toremifene, idoxifene (Fig. 12) was designed to be metabolically resistant so that there would be less likelihood of carcinogenic potential. Substitution of a halogen atom at the 4 position of tamoxifen to prevent metabolic activation to 4-hydroxytamoxifen is known to reduce antiestrogenic potency (Allen et al., 1980). It is therefore an advantage, but not a requirement, for the antiestrogen tamoxifen to be metabolically activated (Allen et al,, 1980; Lieberman et al., 1983). To exploit this concept, idoxifene was designed with an iodine atom at the 4 position of tamoxifen to prevent toxicity through 4-hydroxylation and a pyrrolidino side chain to avoid theoretical toxicities associated with demethylation (McCague et al., 1989, 1990). The compound is entering clinical trial but it is unclear whether idoxifene will be protected from carcinogenic potential. In summary numerous new compounds have been discovered that are now actively being evaluated in clinical trial and should be approved for different treatment applications within the next 5 years. However, before we consider the current status of the evaluations of each new agent we will briefly describe the subcellular mechanism of actions of the two principal classes of agents: compounds based on the tamoxifen molecule and the pure antiestrogens. This is important because it provides insight into the current problems of understanding target tissue responses and the complex issue of drug resistance to antihormones.
Chapter
5
Molecular Mechanisms of Antiestrogen Actions
Subcellular Mechanism of Action of Antiestrogens in Breast Cancer The recognition that the ER is a nuclear transcription 1991) which bind estrogens or antiestrogens and can enormous interest replicate or not, has focused antiestrogen action. A generally accepted model for Fig. 12;. Estradiol (or indeed any estrogen) binds produces a change in shape to expose fully the DNA
factor program on the estrogen to the binding
(Green and Chambon, breast cancer cells to basic mechanism of action is illustrated in nuclear ER that then domain on the protein
STOP GROWTH lTamoxifen Analogues
Fig. 13. Molecular model of estrogen actions in the breast cancer cell. Estradiol (E2) binds to the nuclear estrogen receptor (ER) which changes shape to bind to estrogen response elements (EREs) that control the transcription of estrogen responsive genes. All antiestrogens can bind to the ER but cause an inappropriate change in shape so that binding to the EREs is imperfect. The pure antiestrogens have an additional mechanism of action by promoting the cytoplasmic destruction of the newly synthesized ER.
196
V. C. Jordan and W. J. Gradishar
complex. The activated receptor complex then dimerizes and binds to an estrogen response element (ERE) located in the promoter region of estrogen responsive genes. Once bound to the ERE, the ER acts as an anchor for other transcription factors that when assembled and associated with RNA polymerase, produces a transcription complex. The estrogen responsive gene is then transcribed and subsequently translated to proteins that are either involved in growth responses or differentiation responses, for example progesterone receptor synthesis. The regulation of the ER is complex and not clearly understood, although some general principles can be stated that appear to be important for the development of hormone independent growth. Only estrogens will cause synthesis of the ER while other steroid hormones (e.g. progesterone) are inactive. Nevertheless, there appear to be two models of receptor regulation (Pink and Jordan, 1996) (Fig. 14). In Model 1, MCF-7 breast cancer cells have their ER downregulated by estradiol. Conversely, in times of estrogen deprivation ER levels increase (Welshons and Jordan, 1987; Katzenellenbogen et al., 1987) and estrogen independent clones emerge (Jiang et al., 1992; Pink et al., 1995, 1996a, in press). However, antiestrogens are still effective at regulating growth in short-term laboratory experiments. Ultimately the drug resistance and tumor growth that is observed during indefinite tamoxifen therapy occurs despite the fact the tumor remains ER-positive. It is clear that the drugreceptor complex has discovered new ways of activating growth through the ER. In Model II, exemplified by the T47D cell line, estrogen is necessary to induce the synthesis of the ER and maintain the sensitivity of the cell to the influences of estrogen and antiestrogens. During prolonged periods of estrogen deprivation, the ER
Model I (Inhibitory) E2
,_,---mRNA +--___.\ /’ 1’
IiR
-
Ek
*
\
\
REGULATORY MECHANISM
ER ER
Model II (Stimulatory) E2
-
REGULATORY * MECHANISM
ER ER ERER
ER
Fig. 14. The model systems for the regulation of estrogen receptor function in target tissues. Either the estrogen receptor is down regulated by estrogen (Model I) or upregulated by estrogen (Model II). There would clearly be a physiological need, in rapidly responding tissues, to sequester estrogen rapidly. High concentration of receptor would then be down regulated once estrogen appears. In contrast, sustained, protein synthetic responses of a target tissue, that occur periodically, would require a receptor mechanism that is switched off in the absence of estrogen (Model II) (Pink and Jordan, 1996).
Molecular Mechanisms and Future Uses of Antiestrogens
197
can be lost (Murphy et al., 1992; Pink et al., 1996b) with an associated refractoriness to antiestrogens. This method of drug resistance may be important during long-term total estrogen withdrawal in patients (i.e. sequential combinations of aromatase inhibitors and pure antiestrogens). It is possible that the two mechanisms for regulating the ER may be different stages in the evolution of complete hormone independent growth. Unfortunately we currently have little information about the true state of affairs in breast tumors, but it would be reasonable to assume that because breast tumors are heterogeneous they will contain equilibrium mixtures of cells each seeking to survive through selection in an antiestrogenic environment. The current molecular model of estrogen action provides several potential points of weakness that can be exploited by antiestrogens. It is now apparent that antiestrogens can be divided into two major categories based on their mechanism of action. Type I antiestrogens are the analogs of tamoxifen or structural derivatives of the triphenylethylene type of drug. Type II are the pure antiestrogens. All compounds are competitive inhibitors of the binding of estradiol to the ER but there the similarity ends. Type I antiestrogens appear to form a receptor complex that is incompletely converted to the fully activated form (Tate et al., 1984; Martin et al., 1988; Pham et al., 199 1; Tzukerman et al., 1994; McDonnell et al., 1995; Allan et al., 1992). As a result of the imperfect changes in the tertiary structure of the protein, the complex is only partially active in initiating the programmed series of events necessary to orchestrate gene activation (Metzger et al., 1988; Jordan, 1984). Studies in vitro demonstrate that very low concentration of triphenylethylene-type antiestrogens can cause a single round of replication in breast cancer cells but high concentration of these antiestrogens are completely inhibitory (Berthois et al., 1986). It is possible that the modest partial estrogen-like action at low concentrations causes the tamoxifen flare that is sometimes observed when therapy is started in patients with boney metastases (Reddel and Sutherland, 1984). Once steady state levels of the drug have been achieved (approximated 4-8 weeks with 20 mg/day), symptoms will have dis’appeared and the patient will experience a response to therapy (Furr and Jordan, 1984). It is important therefore, to be able to identify tumor flare and not prematurely terminate a beneficial therapy. Nevertheless, a recent report (Vogel et al., 1995) has demonstrated that clinicians often prematurely terminate antiestrogen treatment based on changes in bone scintigraphy misinterpreted as progressive disease. Since there are clear toxicological advantages in disease control with antiestrogens, a premature change to chemotherapy may be inappropriate. Several type II antiestrogens are available for study in the laboratory (Wakeling, 1994; Van de Velde et al., 1994; Dukes et al., 1994; von Angerer et al., 1990) but only ICI 182,780 is being developed clinically (Wakeling et al., 1991). Initially it was believed that pure antiestrogens prevent the dimerization of receptor complexes thereby preventing binding to EREs (Fawell et al., 1990). Clearly if receptor complexes do not bind to any EREs then no genes can be activated and the compound would be a ‘pure’ antiestrogen. However, numerous reports (Pink and Jordan, 1996; Pham et al., 1991; Salbbah et al., 1991) now demonstrate that pure antiestrogen-ER complexes can
198
V. C. Jordan and W. J. Gradishar
bind to EREs but the transcriptional unit is inactive. What is unique about the type II antiestrogens is the observation that they provoke the destruction of the ER in breast cancer cells in culture (Dauvois et al., 1992), mouse uterus (Gibson et al., 1991) and in breast tumors in situ (DeFriend et al., 1994). The ER is synthesized in the cytoplasm and transported to the nucleus where it functions as a transcription factor. A pure antiestrogen binds to the newly synthesized receptor in the cytoplasm and prevents transport to the nucleus (Davois et al., 1993). The paralyzed receptor complex is then rapidly destroyed (Davois et al., 1993). The complete destruction of available ER will prevent any estrogen regulated events from occurring. Normal cells will become quiescent whereas hormone dependent tumors will rapidly regress because senescent tumor cells cannot be replaced by replication. The elucidation of the subcellular model for the molecular mechanism of action of estrogen has provided an insight into the inhibitory actions of antiestrogens in breast cancer. Most of the work has been completed on tumor cells in culture and extrapolated to the clinic. However, the pharmacology of antiestrogens is complex and the target site specific effects observed in viva on bones and lipids are not adequately explained by studies in breast cancer cells. Alternate explanations for the estrogenic effects of tamoxifen are currently being sought at the molecular level.
Target Site Specific Effects of Antiestrogens Although studies of the molecular biology of the ER have gone some way in explaining the inhibitory effects of antiestrogens there is currently no universal explanation for the diverse pharmacological effects observed with tamoxifen in women. Tamoxifen appears to exhibit inhibitory effects on breast cancer growth but produce estrogen-like effects in postmenopausal women that maintains bone density (Turken et al., 1989; Love et al., 1992, 1994; Ward et al., 1993; Kristensen et al., 1994)) reduces circulating gonadotropins, luteinizing hormone (LH) and folliclestimulating hormone (FSH) (J or d an et al., 1987) and produces estrogen-like effects on vaginal cytology (Boccardo et al., 1981). The ER has been cloned and sequenced (Green et al., 1986; Greene et al., 1986), and, until the discovery of ER,8, there was no evidence that there were different ER receptors located at different sites around a woman’s body. However, at least two alternative mechanisms, illustrated in Fig. 15, could be used by the antiestrogen-ER complex to stimulate the selective activation of estrogen responsive genes. We will consider each hypothesis in turn: the role of associated proteins in the cell nucleus and the concept of an ‘antiestrogen response element’ (AERE). ER-associated proteins (Halachmi et al., 1993) may be differentially located at target sites throughout a woman’s body and these could be selectively used by the cells to govern estrogen regulated growth or differentiation. The molecular events needed to provoke cells to pass through the cell cycle have to be quantitatively and qualitatively precise. Any disruption of the correct sequence or defect in the amounts of required catalysts would lead to inappropriate scheduling of replication which clearly would be catastrophic for a growing tumor that depends for its survival on the equilibrium between replicating and dying cells. It is possible that the changes in the tertiary
Molecular
Mechanisms
199
and Future Uses of Antiestrogens
No replication
)’
*Differentiation, prolonged response
_)
\;-_
7
differentiation only
replication and differentiation
Fig. 15. A model of estradiol (E,) action at physiological sites around a woman’s body. (a) The nuclear estrogen receptor (ER) may bind to associated proteins (ASP) to modulate the rapid response of replication or the prolonged effects of differentiation. There are currently two potential models to describe the estrogen-like effects of tamoxifen at physiological sites. (b) The antiestrogen (AE) binds to the ER but the ASP have only a weak affinity for the receptor complex and cannot support transcription of genes necessary to support replication. types of receptor The relentless signaling may, however, support other differentation responses because the EREs are less specific. (c) The AE binds to the ER, but the AER complex interacts with an antiestrogen response element (AERE) that triggers differentiation only. The AER blocks EREs but the AERE plays a dominant role.
200
V. C. Jordan and W. J. Gradishar
shape of the antiestrogen-ER complex may be inappropriate to bind to associated proteins to accomplish the rapid responses needed for replication. The low affinity, and perhaps the inability of the cells to create higher concentrations of associated proteins, might cause a paralysis in the ability of the breast cancer cell to survive. In contrast, the physiological effects of tamoxifen on bones, cholesterol and gonadotropins depend on long-term signaling that lasts days, months or even years for completion. Normal cells may be able to respond to the signal from the altered ER complex, by either synthesizing more associated proteins over days and months to establish the appropriate signal transduction pathway, or by producing the correct expansion of estrogen responsive pathways. Overall the partial estrogen-like response would build over time in selected normal tissues to produce an estrogenic effect. In contrast, cell replication in a tumor could only succeed through selection of resistant clones that can exploit the weak estrogen-like effects of antiestrogens by expansion of the pool of associated proteins necessary for replication. The second potential mechanism to explain the estrogen like effects of antiestrogens depends on the antiestrogen-ER complex using an alternate response element from the ERE in the promoter region of estrogen responsive genes. The concept depends upon the ability of the antiestrogen-ER complex to alter the shape of the protein so that binding to the ERE is inappropriate for gene activation but the complex can seek out an alternate site in the promoter region to initiate gene transcription (Yang et al., 1996). This could be an AERE. It is possible that the explanation for the target site specific action of type I antiestrogens could be any one or a combination of the hypotheses illustrated in Fig. 15. However, it is important to elucidate the precise mechanism because the idea of being able to target antiestrogens to switch on or switch off gene function in specific sites has fundamental importance for the prevention of osteoporosis, coronary heart disease, breast and endometrial cancer. With this background of tamoxifen’s toxicology, novel drug discovery mechanisms of actions of antiestrogens we will now consider the progress been made in the development of new clinically useful antiestrogens.
and that
the has
Numerous compounds are being evaluated in clinical trial but for convenience have divided them into three major categories based on their proposed applications.
we
Triphenylethylenes
(tamoxifen
analogs)
Pure antiestrogens Targeted
antiestrogens
We will consider each new compound by discussing support the clinical testing currently in progress.
the
laboratory
evidence
to
Chapter
6
Triphenylethylenes (Tamoxifen Analogs)
Toremifene Toremifene (Fig. 12) or chlorotamoxifen began development in Finland by Farmos Pharmalceuticals (Orion) in 1978. The drug is FDA approved for the treatment of metastatic breast cancer and is marketed in the US under the trade name of Farnesdonm by Scheering Plough, New Jersey. The recommended treatment regimen for metastatic breast cancer in postmenopausal women is at least 60 mg daily.
Laboratory Studies Toremifene has a biphasic effect on the growth of ER-positive, MCF-7 breast tumor cells in vitro (Kangas et al., 1986; Kangas, 1990a). At low concentrations (10h7 to 10W6 M) toremifene is an estrogen antagonist but at high concentration ( > lop6 M) toremifene is oncolytic and the effects cannot be reversed by estrogen (Warri et al., 1993; Grenman et al., 1991; Robinson et al., 1990). Nevertheless, toremifene can be washed out of the cells and estradiol can re-stimulate growth. Toremifene and other antiestrogens increase the production of TGF-/? (Colletta et al., 1990; Knabbe et al., 1991) and toremifene-induced cell death has been documented as apoptotic (Warri et al., 1993). Toremifene is an antiestrogen in the immature rat uterine weight test and exhibits the properties of a partial estrogen antagonist (Kangas, 1990b; DiSalle et al., 1990) (i.e. toremifene causes modest increases in uterine weight when administered alone). Toremifene is effective in controlling the growth of DMBA-induced rat mammary carcinomas (DiSalle et al., 1990; Huovinen et al., 1993, 1994, 1995), but appears to be approxirnately l/3 less potent than tamoxifen (Robinson et al., 1988). This is consistent with the larger dose of toremifene (60 mg daily) that is used clinically compared with tamoxifen (20 mg daily). The MCF-7 (ER-positive) and MDA-MB-231 (ER-negative) breast cancer cell lines will grow in athymic mice. However, toremifene will only block the estrogenstimulatmed growth of MCF-7 cells but does not inhibit the growth of MDA-MB-231 cells (Robinson and Jordan, 1989). The drug does not control the growth of mixed tumors containing both MDA-MB-231 and MCF-7 cells (Robinson and Jordan, 1989). Nevertheless, toremifene has been reported to have a cytolytic effect on controlling the growth, in vivo, of an ER-negative, glucocorticoid sensitive mouse uterine
V. C. Jordan and W. J. Gradishar
202
can produce acquired resistance sarcoma (Kangas et al., 1986). Toremifene long-term therapy. Athymic animals implanted with MCF-7 cells will eventually tumors in response to toremifene treatment (Osborne et al., 1994).
after grow
Toxicology Toremifene does not produce a mutagenic effect in either the Ames test or the sister chromatid exchange assay (Kangas et al., 1986; Styles et al., 1994). Large daily doses (5-20 mg) o f t oremifene do not produce DNA adducts in the rat liver and long-term therapy does not result in hepatocarcinogenesis (Hard et al., 1993; Hirsimaki et al., 1993). However, recent studies demonstrate that large doses of toremifene (750 mg toremifene/kg food) can promote rat liver and kidney carcinogenesis (Dragan et al., 1995).
Clinical Pharmacology and Endocrinology Toremifene is extensively metabolized in animals and patients(Antilla et al., 1990; Kangas, 1990b). The principal metabolites are shown in Fig. 16. Toremifene can be
CI
Toremifene
N-desmethyltoremifene
-a HO
&
4 hydroxytoremifene
c1
T6k IV
TOR VI
Fig. 16. The metabolites of toremifene described in animals and man. The routes appear to be the same as those previously described for tamoxifen (Jordan, 1984), demethylation and deamination of the dimethylaminoethoxy side-chain and 4-hydrovlation to a metabolite with high binding affinity for the ER.
Molecular
Mechanisms
and Future Uses of Antiestrogens
203
measured using high performance liquid chromatography (HPLC) (Wiebe et al., 1990; Webster et al., 1991; Berthou and Dreano, 1993; Bishop et al., 1992) and the time to steady state (Wiebe et al., 1990; Bishop et al., 1992; Kohler et al., 1991) and terminal elimination half-life (Antilla et al., 1990) have been determined as 2 weeks and 5 days, respectively. Toremifene exhibits weak estrogen-like properties in the postmenopausal patient. LH and FSH are slightly depressed during therapy and sex hormone binding globulin (SHBG) is increased (Hamm et al., 1991; Kivinen and Maenpaa, 1990; Szamel et al., 1994). Although toremifene exhibits antiestrogenic effects on the vaginal mucosa of estrogen-primed women (Homesley et al., 1993; Maenpara et al., 1990), there is no effect in blocking short-term estrogen action in the uterus ((Maenpaa et al., 1990). Toremifene and tamoxifen produce the same estrogenlike effects on the histology of the postmenopausal endometrium (Tomas et al., 1995).
Clinical Evaluation The initial phase I studies, started in the early 198Os, demonstrated that toremifene was well1 tolerated with minimal toxicity, and activity in breast cancer was observed (Wiebe et al., 1990; Hamm et al., 1991; Tominga et al., 1990). Several phase II clinical trials of toremifene have been reported (Table 3) in postmenopausal patients with advanced disease, who did not receive prior hormonal or cytotoxic chemotherapy (Valavaa.ra et al., 1988; Gunderson, 1990; Modig et al., 1990; Valavaara and Pyrhonen, 1989; Hietanen et al., 1990; Valavaara, 1990). In 46 previously untreated patients with ER-positive, metastatic breast cancer, Valavaara et al. (1988) reported a 63% objective response rate (CR-37%; PR-26%) following treatment with toremifene (60 mg/day, PO). Responses were observed in soft tissue and visceral sites of disease. No significant differences in response rates could be detected when related to different ER concentrations. Toxicity was mild with hot flashes occurring in 22% of patients. Gunderson (1990) reported a 48% response rate, including six complete responders, using a:n identical treatment schedule of toremifene in a group of 23 patients with advanced disease in whom 20 had received no prior therapy. The median duration of complete and partial responders was 14 and 1.5 months, respectively. Hot flashes were reported in approximately half of the patients. Similar findings have been reported by Modig et al. (1990). In an effort to determine if lower daily doses of toremifene were equally effective as higher doses, toremifene treatment was evaluated by initially administering an oral loading dose: 120 mg on day 1, 60 mg on days 2 and 3, and thereafter 20 mg daily (Valavaara and Pyrhonen, 1989). Of the 14 patients treated according to this schema, three achieved a partial response (21%) and seven achieved stabilization of disease. No complete responses were observed. In a multicenter study involving 38 patients with advanced disease, oral toremifene was administered at a dose of 240 mg/day (Hietanen et al., 1990). An objective response rate of 68% was observed (CR-26%; PR-42%). This study suggested that higher doses of toremifene are more effective in inducing objective responses compared to low-dose toremifene (20 mg/day). Toremifene in patients
has been directly compared with advanced disease (Table
to tamoxifen as first-line hormonal therapy 4) (Hayes et al., 1995; Nomura et al., 1993;
204
V. C. Jordan and W. J. Gradishar
Konstantinova and Gershanovich, 1990; Stenbygaard et al., 1993). Three small randomized trials, lacking the statistical power to make a valid comparison between tamoxifen and toremifene, have been published. In a randomized trial comparing toremifene (40 mg/day, p.o.) to tamoxifen (20 mg/day, p.o.), Nomura et al. (1993) observed similar response rates and median time to disease progression in both treatment arms. Konstantinova and Gershanovich (1990) reported on 47 patients randomized to one of two doses to toremifene (60 mglday, p.o. or 240 mglday, p.o.) or tamoxifen (40 mglday, p.0.). Patients treated with 60 mglday of toremifene had a response rate of 50% compared to 35% in the higher dose toremifene arm and 36% in the tamoxifen arm. In contrast, Stenbygaard et al. (1993) reported inferior response rates for patients treated with toremifene (240 mg/day, p.o.) compared to tamoxifen (40 mg/day, p.0.). In a recently reported international trial (Hayes et al., 1995), 648 previously untreated, hormone receptor-positive or -unknown, metastatic breast cancer patients were randomized between tamoxifen (20 mglday, p.o.) or two different doses of toremifene (60 or 200 mg/day, p.0.). Tamoxifen produced a response rate of 19% and a median survival of 32 months. Toremifene produced a response rate of 21% with the 60 mg dose and 23% with the 200 mg dose. Median survival of toremifene-treated
Table 3. Phase II trials of toremifene in postmenopausal women with advanced breast cancer with no prior endocrine therapy of chemotherapy Study
Patient population
Dose/schedule
Response
Toxicity
Valavaara et al. (1988)
46 postmenopausal ER+, metastatic breast cancer
60mgpoqd
Mild, hot flashes-22%
Valavaara and Pyrhonen (1989) Gunderson (1990) Hietanen et al. (1990) Modig et al. (1990)
14 postmenopausal, ER+ or unknown
20mgpoqd
RR CR SD 54% 17% 26% Soft tissue, visceral sites most likely to respond RR CR SD 21% 0 58%
23 postmenopausal, ER+ or unknown 38 postmenopausal, ER+ or unknown 12 postmenopausal, hormone receptor + or unknown, inoperable or metastatic breast cancer 113, ER+, postmenopausal, metastatic breast cancer. First-line rx for advanced disease.
60mgpoqd
Valavaara (1990)
240 mgpoqd 60mgpoqd
60mgpoqd
RR 48% RR 68% RR 50%
CR 26% CR 26% CR 25%
SD 26% SD SD 42%
RR CR SD 45% 15% 35% Duration of response correlated with ER concentration
Mild, hot flashes-28% Mild Mild None
NA
Abbreviations: rx, treatment; RR, response rate; po, oral; CR, complete response; qd, daily; SD, stable disease; NA, not available.
Molecular
Mechanisms
and Future Uses of Antiestrogens
205
patients was 38 months (60 mg/day) and 30 months (ZOOmg/day). The median time to disease progression was not statistically different between treatment arms (Hayes et al., 1995). Response rates, times to disease progression and overall survival for patients on each arm were superior for ER-positive patients compared to ER-negative patients. However, no statistical difference in any of these endpoints was detected between treatment arms. Furthermore, quality-of-life assessments were not different between treatment arms. Toxicity was mild in all patients, but toremifene-treated patients experienced less nausea (26% versus 37%). The data from this large trial (Hayes et al., 1995) support the use of toremifene as an alternative first-line therapy to tamoxifen in hormone receptor-positive, postmenopausal patients with advanced disease. Toremifene appears to have cross-resistance to tamoxifen, since overall response rates to toremifene following tamoxifen therapy in several phase II studies are low
Table
4. Clinical
trials
Study
comparing
toremifene to tamoxifen advanced breast cancer
Patient population
Dose/ schedule
Konstantinova and Gershanovich (1990)
Nomura
Toxicity
RR
CR
NA
TOR 60 mg
50%
0
17
PO qd TOR240mg
35%
6%
14
PO qd TAM 40 mg
36%
1%
PO qd 57
TOR 40 mg
RR 26%
CR 14%
57
PO qd TAM 20mg
28%
5%
PO qd
et al. (1993)
patients
Response
16
et al. (1993)
Stenbygaard
in postmenopausal
with
NA
31
TOR240mg
RR 29%
CR 3%
31
PO qd TAM 40 mg
42%
16%
RR 19%
CR 5.8
SD 31.7
Mild; nausea 37%
TOR 60 mg
21%
5.6
38
26%
PO qd TOR 200 mg
23%
5.6
30.1
26%
PO qd
Hayes et al. (1995) 648, peri- or post-menopausal hormone receptor+ or unknown metastatic breast cancer
TAM 20 mg
NA
PO qd
PO qd Abbreviations: TAM, tamoxifen; RR, response rate; TOR, oral; SD, stable disease; qd, daily; NA, not available.
toremifene;
CR, complete
response;
po,
V. C. Jordan and W. J. Gradishar
206
(Table 5) (Ebbs et al., 1990; Hindy et al., 1990; Jiinsson et al., 1991; Asaishi et al., et al., 1994). The largest experience was a 1993; Vogel et al., 1993; Pyrhonen multicenter trial reported by Vogel et al. (1993) in which 102 perimenopausal or postmenopausal women with metastatic breast cancer, refractory to tamoxifen, received 200 mg/day of toremifene. Patients in this trial were heavily pretreated with 65% having failed chemotherapy and 72% having failed two or more hormonal therapies. Forty-nine percent of patients had visceral-dominant disease. The objective
Table 5. Clinical trials of toremifene
in patients
with advanced breast cancer refractory
to tamoxifen
Study
Patient population
Dose/schedule
Response
Toxicity
Ebbs et al. (1990)
9 TAM-refractory metastatic breast cancer 17 postmenopausal metastatic breast cancer; prior hormonal rx or RT if d/c 2 months prior 35 TAM-refractory, ER+ or unknown, post-menopausal inoperable or metastatic breast cancer 5 1 TAM-refractory, ER+ or unknown metastatic breast cancer 50 TAM-refractory, ER+ metastatic breast cancer. 48% patients with disease progression during adjuvant TAM 52% patients with advanced disease 102 TAM-refractory, metastatic breast cancer. I: 28-1” refractory II: 43-relapse after TAM response III: 31-relapse while receiving TAM 831102 assessable 81% ER+ and/or PRt 50% visceral disease
200 mg po qd
RR 33%
CR 0%
Mild
60, 120, or 300 mg po
RR 25% 33%
CR 0% 0%
Hindy et al. (1990)
Jonsson et al. (1991)
Asaishi et al. (1993)
Pyrhonen et al. (1994)
Vogel et al. (1993)
qd
240 mg po qd
120 mg po qd
120 mg po bid
(60 mg) (300 mg)
Mild
RR SD 0% 26% TTP in SD 8 months
RR CR 14% 0 TTP in SD 6 months RR CR 4% 2%
Minimal; nausea, hot flashes
SD 19%
NA
SD 22%
Minimal: sweating 12%: nausea 4%
SD 23%
Minimal: nausea, hot flashes, dry eyes
TTP in SD 2 months 200 mg po qd
RR 5%
CR 2%
TTP in SD 8 months
Abbreviations: TAM, tamoxifen; po, oral; CR, complete response; NA, not available; qd, daily; SD, stable disease; rx, treatment; RR, response rate; TTP, time to progression.
Molecular
Mechanisms
and Future Uses of Antiestrogens
response rate was 5% with only two patients achieving a CR. An additional patients maintained stable disease status for a median of 8 months.
207
23% of
Toremifene was observed to have an antitumor effect in mice with ER-negative uterine sarcomas and, as a result, a cytolytic effect independent of the estrogen receptor was postulated (Kangas et al., 1986). In a small trial of nine patients with ER-unknown breast cancer progressing following tamoxifen therapy, 33% of patients (3/9) responded to toremifene (200 mg/day, p.o.) (Ebbs et al., 1990). In an effort to confirm a mechanism of action independent of the ER, the Cancer and Leukemia Group B (CALGB) conducted a phase II trial to test the efficacy of high-dose toremifene (400 mgiday) in a population of patients with hormone receptor-negative, metastatic disease with limited prior chemotherapy exposure (Perry et al., 1995). Twenty patients were enrolled, but no objective responses were observed. These findings reaffirm that toremifene is primarily active in patients with hormone receptor-positive, metastatic breast cancer.
Droloxifene Droloxifene or 3-hydroxytamoxifen (Fig. 8) began development in Germany Pharmaceuticals in the late 1970s and subsequently by Fujisawa in Japan. has been tested for the treatment of metastatic breast cancer but is being by Pfizer for the treatment of osteoporosis in postmenopausal women.
by Klinge The drug developed
Laboratory Studies N-desmethyldroloxifene Droloxifene (Roos et al., 1983) and its major metabolite (Loser et al., 1985a) have a ten-fold higher binding affinity for MCF-7 (ER-positive) breast cancer cells than tamoxifen. Droloxifene inhibits estrogen-stimulated cell et al., 1991; Kawamura et al., 1993) by replication (Loser et al., 1985b; Eppenberger appears to be arresting cells in GdG, (Hasman et al., 1994). In addition, droloxifene a more potent inducer of TGF-/I than either tamoxifen or toremifene (Knabbe et al., 1991). Similarly droloxifene inhibits IGF-l-stimulated growth of MCF-7 cells (Hasman et al., 1994). In contrast, droloxifene does not inhibit the growth of the ERthat negative cell line, MDA-MB-231 (Kawamura et al., 1993). This demonstrates droloxifene is active through the ER. Droloxifene exhibits antiestrogenic activity in the immature rat uterine weight test but also causes a partial increase in uterine wet weight when administered alone action of (Wosikiowski et al., 1993; Loser et al., 1985b). The partial agonist droloxifene is only slightly less potent than tamoxifen (Eppenberger et al., 1991; Wosikowski et al., 1993). However, droloxifene does maintain bone density in the ovariectomized rat (Ke et al., 1995). Droloxifene exhibits antitumor activity in several rat and mouse models. Droxloxifene inhibits the growth of the transplantable rat mammary tumor R3230AC, and both DMBA-(Loser et al., 1985b; Kawamura et al., 1991) and NMU-(Winterfeld et al., 1992) induced rat mammary tumors, but only ERpositive breast tumors transplanted into athymic mice (Kawamura et al., 1993; Wosikowski et al., 1993).
208
V. C. Jordan and W. J. Gradishar
Toxicology Droloxifene does not produce DNA adducts or hepatocellular carcinomas in male or female rats fed daily doses of 36 mg/kg (Hasman et al., 1994). One male and one female rat (2% total) developed a hepatocellular carcinoma following treatment for 24 months with 90 mglkglday. By comparison, tamoxifen produced hepatocellular carcinomas in 100% animals following 36 mg/kg/day for 24 months (Hasman et al., 1994). Droloxifene is inactive in the ability to transform Syrian hamster embryo cells in vitro whereas tamoxifen and 4-hydroxytamoxifen produce a significant level of transformation (Metzler and Schiffmann, 1991).
Clinical Pharmacology and Endocrinology Droloxifene is rapidly absorbed and excreted and does not appear to accumulate like tamoxifen and toremifene. Droloxifene can be monitored in serum using HPLC (Grill and Pollow, 1991; Lien et al., 1995). Under chronic dosing conditions steady state levels of parent drug were 83.5 _+32.5 ng/ml (40 mg daily) and 146 + 115.4 ng/ml (100 mg daily) with a half-life of 28 and 27 hours, respectively. Steady state levels were achieved rapidly within 5 hours (Grill and Pollow, 1991). There are several metabolites of droloxifene (Fig. 17) and all are present in serum as both free and glucuronide conjugates. This profile contrasts with tamoxifen that does not have serum glucuronide metabolites (Grill and Pollow, 1991). Droloxifene causes a dose-related decrease in LH and FSH in postmenopausal patients (Lien et al., 1995). There is only a modest decrease at the 20 and 40 mg dose daily, whereas there is a definite decrease at the 100 mg daily dose. Similarly there is only a marginal rise in SHBG at the 20 and 40 mg daily dose but a moderate increase occurs with 100 mg droloxifene daily. These conclusions have recently been confirmed in patients treated with 40 mg droloxifene daily (Geisler et at., 1995).
Clinical Evaluation Table 6 summarizes the results of clinical trials evaluating droloxifene in patients with metastatic breast cancer (Ahlemann et al., 1988; Abe et al., 1990, 1991; Bellmunt and Sole, 1991; Haarstad et al., 1992; Raushning and Pritchard, 1994; Buzdar et al., 1994). The majority of patients on these trials had been previously treated with chemotherapy and/or hormone therapy (Ahlemann et al., 1988; Abe et al., 1990, 1991; Bellmunt and Sole, 1991; Haarstad et al., 1992; Raushning and Pritchard, 1994; Buzdar et al., 1994). The daily dose of droloxifene ranged from 20 to 300 mg. Response rates ranged from 0 to 70% with most responses occurring in peri- or postmenopausal patients. Trials evaluating different doses of droloxifene have not convincingly demonstrated a dose-response effect (Bellmunt and Sole, 1991; Raushning and Pritchard, 1994; Buzdar et al., 1994). The largest clinical trial involving patients with metastatic breast cancer treated with droloxifene was recently updated by Raushning and Pritchard (1994). This phase II study compared droloxifene in doses of 20, 40 and 100 mg daily in postmenopausal women with metastatic, inoperable recurrent, or primary locoregional breast cancer
Molecular
Mechanisms
and Future Uses of Antiestrogens
who hatd not received prior hormonal eligible: and 268 evaluable for response. 47% in the 40 mg group and 44% occurred within 2 months of starting been extremely well tolerated with flashes, fatigue and nausea.
209
therapy. Of 369 patients randomized, 292 were Response rates were 30% in the 20 mg group, in the 100 mg daily group. Most responses therapy. In all trials reported, droloxifene has the most common toxicities cited being hot
ldoxifene The drug was originally designed Sutton (Surrey, UK) and offered
at the Cancer Research to the pharmaceutical
-“<
Campaign Laboratory in industry through British
-E-
0
0 minor metabolite
HO
HO
DROLOXIFENE
N-DESMETHYLDROLOXIFENE
HO
4-METHOXYDROLOXIEENE Fig. 17 The metabolites of droloxifene described in patient serum. The drug and metabolites are rapidly conjugated to glucuronides that are the major component detected in the blood. ![nterestingly the 4-methoxy metabolite of droloxifene has been identified despite the earlier observation that 3,4_dihydroxytamoxifen is metabolically stable in vitro in the presence of an inhibitor of catechol-o-methyl transferase (Jordan et al., 1984). The enzyme would prefer to direct a methyl to the 3-hydroxyl not the 4-hydroxyl.
10 peri or postmenopausal, ER+ or unknown, metastatic breast cancer. Four received prior hormone or chemotherapy.
Ahlemann
16 pre- or postmenopausal, hormone receptor +, -, or unknown. Metastatic breast cancer. All pretreated.
94 pre- or post-menopausal, hormone receptor +, -, or unknown. Primary or recurrent advanced breast cancer. Majority pretreated.
Abe et al. (1991)
Abe et al. (1990)
et al. (1988)
Patient population
Study
qd
PO qd
20, 40, or 80 mg
120 mgpo
100 mg po q 2 or 3 days
treated patients
CR 0% 4% 0%
RR 20 mg: 14% 40 mg: 5% 100 mg: 17% Most responses in peri- or post-menopausal patients.
SD 32% 46% 48%
CR 0%
RR 29%
Side effects mild in all groups, but fewest occurred in 20 mg group.
Mild in two patients
Mild Hot Flashes
CR 30% 40% 20%
RR 70% 60% 80%
30% RR in perior post-menopausal patients. 40% RR in ER+ or unknown disease.
Toxicity
SD 43%
with advanced breast cancer Response
Overall q 2 days: q 3 days:
in previously
Dose/schedule
Table 6. Clinical trials of droloxifene
Haarstad et al. (1992)
26 postmenopausal women; 1 male. Metastatic disease. All pretreated with endocrine rx and 35% with chemotherapy
124 postmenopausal, hormone receptor + or unknown (65%); metastatic disease; 68% pretreated
Bellmunt (1991)
and Sole
Patient population _ -
Study
PO qd
100 mg
20, 40, or 100 mg PO od
Dose/schedule
20 mg: 40 mg: 100 mg:
Table 6. contd.
CR SD 0% 19% SD duration 22 weeks
RR 15%
SD 36-40%
CR 0% 3% 10%
RR 17% 30% 31%
Response
Nausea 22% Hot flashes 15% Vomiting 15% GI discomfort 18% Anorexia 15% Two patients with severe fatigue.
Toxicity
$-.
5 b 2 E 5 0
response;
20, 40 or 100, 200 or 300 mg PO qd.
20, 40 or 100 mg PO qd.
Dose/schedule
*Updated results from Bellmunt and Sole (1991). Abbreviations: po, oral; qd, daily; RR, response rate; CR, complete
Buzdar et al. (1994)
23 patients with metastatic, refractory breast cancer. ER+ or previously responsive to endocrine therapy; median number prior hormone rx-3; chemotherapy rx-2.
369 postmenopausal, hormone receptor + or unknown, metastatic or inoperable recurrent or primary locoregional breast cancer
Raushning Pritchard
and (1994)*
Patient population
Study
Mild. Hot flashes, headache, fatigue, nausea and leg cramps most common.
Adverse effects mild and equal between groups.
SD 42% 35% 27%
RR 30% 47% 44% CR 6% 8% 12%
Toxicity
Response
PR, partial; SD, stable disease.
No CR or PR 13% minor response
20 mg: 40 mg: 100 mg:
Table 6. contd.
$ ZJ
&
5 0
Molecular Mechanisms and Future Uses of Antiestrogens Biotechnology. Idoxifene development by SmithKline
(Fig. 12) is currently Beecham in the UK
undergoing
213 evaluation
and
Laboratory Studies Idoxifene has a binding affinity for the ER that is about twice that of tamoxifen and this translates to a modest increase, compared to tamoxifen, in the ability to inhibit the growth of ER-positive, MCF-7 breast cancer cells in culture (Chander et al., 1991). As would be expected with a compound that cannot be metabolically activated to the 4-hydroxy derivative (McCague et al., 1990), idoxifene is a less potent (approximately ten-fold) antiestrogen in immature rat uterine weight tests. However, idoxifene has less uterotropic activity when administered alone (Chander et al., 1991). Idoxifene demonstrates antitumor properties in the NMU-induced rat mammary carcinoma model in the dose range l-2 mg/kg (Chander et al., 1991). Although this range is somewhat higher than reported for tamoxifen, this would be expected from a drug that cannot be metabolically activated. The authors did show superior activity to tamoxifen at the 1 mg/kg dose (Chander et al., 1991).
Toxicology The drug was designed to avoid the toxicology problem of liver carcinogenic@ produced by tamoxifen (McCague et al., 1989, 1990). A recent report demonstrates that tamoxifen, toremifene and idoxifene, given as acute doses, will cause aneuploidy in rat liver (Sargent et al., 1996). However, idoxifene has not been evaluated as a rat liver carcinogen. It is known that N-demethylation is the major metabolic route for tamoxifen so a pyrrolidino group was used to resolve liver toxicity through a reduction in formaldehyde production. Pyrrolidino tamoxifen (at half the dose) gives levels of DNA adducts as high as tamoxifen in rat liver (White et al., 1992). Mice show interstitral hyperplasia in the ovaries and the expected decrease in uterine weights after 4 weeks of treatment with 25-50 mg/kg of daily idoxifene. A single dose study in mice at 100 mg/kg shows no mortality or behavioral changes (Chander et al., 1991). Tamoxifen produces similar actions in the mouse but the metabolic profile in the mouse differs from the rat (Robinson et al., 1991) and mice do not produce liver tumors in response to tamoxifen (Furr and Jordan, 1984). Idoxifene is designed to be an agent that is ‘metabolism restricted’ and as such requires careful evaluation for unusual or toxic routes of metabolism not previously noted with tamoxifen.
Clinical1Pharmacology and Endocrinology Preliminary studies in the laboratory with idoxifene demonstrate no metabolism of idoxifene over 48 hours of administration (Carnochan et al., 1994). The studies are being used to establish a data base for the evaluation of [lz41] idoxifene by PET imaging during clinical studies. Idoxifene can be measured by HPLC (Coombes et al., 1995), but although 4’-hydroxyidoxifene (Fig. 18) is the most likely metabolite it has not been reported in clinical studies. Idoxifene has an initial half-life of 15 hours and a terminal half-life of 23.3 days (e.g. three times greater than tamoxifen). Idoxifene causes a modest decrease in LH and FSH but no increase in SHBG.
214
V. C. Jordan and W. J. Gradishar
Clinical Evaluation Only one clinical trial in humans has been reported with idoxifene. Coombes et al. (1995) reported the results of a phase I clinical trial in which 20 patients with advanced breast cancer (ER-positive or unknown) were treated with one of four dose levels of idoxifene. The majority of patients previously received tamoxifen, secondline hormone therapy, and chemotherapy. Partial responses were observed in 14% of patients and additional 29% of patients had stable disease for 1.4-14 months (Table 7) Toxicity was mild and not dose related.
TAT-59 TAT-59 is a prodrug and is being developed in Japan for the treatment of advanced breast
by the Taiho cancer.
Pharmaceuticals
Co. Ltd.
3
NN
0
& 1:
,:I
1:
IDOXIFENE
I
3
NN
0
c
I
Jsi+ 1 ;
OH
3
4’-HYDROXYIDOXIFENE Fig. 18.
The possible metabolite
of the metabolically
resistant
antiestrogen
idoxifene.
Molecular
Mechanisms
and Future Uses of Antiestrogens
215
Table 7. Clinical trial involving idoxifene Study Coombes (1995)
et al.
Patient population
Dose/schedule
Response
Toxicity
20 patients
10, 20, 40, 60 mg po qd. (14/20 patients received idoxifene longer than 2 weeks.)
RR PR SD 14% 14% 29% SD duration 1.5-1.4 months
Mild, not dose related Nausea 15% Anorexia 15% Fatigue 20%
with metastatic breast cancer. lo/20 patients prior TAM rx. 16/20 patients prior second-line hormonal rx. ER+ or unknown. 13120 patients received prior chemotherapy
Abbreviations: RR, response disease; qd, daily.
rate;
TAM,
tamoxifen;
PR, partial
response;
po, oral; SD, stable
o\b,O
HO/ \oa
TAT-59
dephosphorylation
HO
DP-TAT-59 (active metabolite) Fig.
19.
The metabolic
activation
of the antiestrogen
TAT-59
to the 4-hydroxy
derivative.
V. C. Jordan and W. J. Gradishar
216
Laboratory Studies TAT-59 is active in inhibiting the growth of ER-positive, DMBA-induced rat carcinomas (Toko et al., 1990). The drug is converted to its mammary dephosphorylated metabolite (Toko et al., 1990) that has high binding affinity for the ER (Toko et al., 1992) (Fig. 19). The drug is active in inhibiting the growth of estrogen-stimulated, ER-positive breast cancer cells transplanted into athymic mice (Koh et al., 1992; Iino et al., 1994).
Clinical Evaluations Clinical studies been published.
using
TAT-59
for the treatment
of advanced
breast
cancer
have not
Chapter 7
Pure Antiestrogens
Pure antiestrogens were first described in the mid-1980s (Wakeling and Bowler, 1987). The compound ICI 182,780 (ZM.182780) is being developed by Zeneca Pharmaceuticals, UK. for the treatment of advanced breast cancer following the failure of long-term adjuvant tamoxifen therapy. Pure antiestrogens could also find application in gynecology and other non-malignant conditions.
Laboratory Studies ICI 182,780 is a competitive inhibitor of estrogen action by blocking estrogen binding to the ER (Wakeling et al., 1991) and causing a destruction of the ER (Dauvois et al., 1992; Gibson et al., 1991). The antiestrogen is a potent inhibitor of the growth of MCF-7 cells (10-9-10-7M) (Wakeling et al., 1991; Thompson et al., 1989) and causes a. more complete inhibition of growth compared to tamoxifen (Thompson et cell line, MDA-MB-231, is unaffected by pure al., 1989). The ER-negative antiestrogens. ICI 182,780 is a potent and complete antiestrogen when given orally or subcutaneously to immature rats. Furthermore ICI 182,780 can inhibit the partial estrogen-like effects of tamoxifen on the rat uterus (Wakeling et al., 1991). This may be important because tamoxifen can eventually encourage the growth of MCF-7 breast c,ancer cells implanted into athymic mice (Osborne et al., 1987; Gottardis et al., 1989a). Similarly tamoxifen-stimulated endometrial carcinoma has been reported to grow in athymic animals (Satyaswaroop et al., 1984; Gottardis et al., 1988). Pure antiestrogens will inhibit the growth of tamoxifen-stimulated breast and endometrial tumors in the laboratory (Gottardis et al., 1989b; Gottardis et al., 1990). The antiestrogen ICI 182,780 will control the growth of tamoxifen-stimulated tumors for prolonged periods, however growth eventually occurs. Preliminary studies (Osborne et al., 1995:) suggest that ICI 182,780 resistant tumors may be developed but further work is required to describe the model in detail. It is known in the laboratory that ER+ breast cancer cells will develop subclones that are ERif they are maintained in an estrogen-deprived state (Pink and Jordan, 1996). Whether this is true clinically remains to be determined. Rats with established DMBA-induced tumors show a more rapid decrease in tumor size and uterine weight with a combination of the luteinizing hormone-releasing hormone (LHRH) analog, goserelin, and ICI 164,384 compared
217
218
V. C. Jordan and W. J. Gradishar
with goserelin alone (Nicholson et al., 1990). The complete estrogen blockade that might eventually premenopausal patients with advanced disease.
combination be valuable
may provide a more for the treatment of
Toxicology There have been no reports of genotoxicity or carcinogenesis with ICI 182,780. Unlike estradiol or tamoxifen, the administration of ICI 164,384 to neonatal female rats did not accelerate the onset of puberty or lead to abnormalities in the development of the reproductive tract (Wakeling and Bowler, 1988). Although ZM 189,154, a pure nonsteroidal antiestrogen not intended for clinical application does not affect bone density in the rat (Dukes et al., 1994), ICI 182,780 reduces cancellous bone volumes in female rats (Gallagher et al., 1993).
Clinical Pharmacology and Endocrinology There are no reports about the metabolism of ICI 182,780 but injection of 18 mg/day produces blood levels of 25 ng/ml after 1 week of treatment. ICI 182,780 is determined by radioimmunoassay (DeFriend et al., 1994). Patients treated with ICI 182,780 for a few days have a significant decrease in the Ki67, PgR and ER in their breast tumors (DeFriend et al., 1994). ICI 182,780 has no effect on LH, FSH, or SHBG (DeFriend et al., 1994). The pharmacokinetics of ICI 182,780 were evaluated following 1 month and 6 month treatments with 250 mg/month. The AUC were 140.5 and 206.8 ng/day/ml on the first and sixth month of dosing, respectively, These data were consistent with drug accumulation after multiple doses and suggested that lower doses of the drug may be effective in maintaining therapeutic serum drug levels. A recent therapeutic study using depot ICI 182,780 showed rises in gonadotrophins but interestingly, no associated rise in circulating cholesterol. Howell et al. (1996) noted that all patients had previously been treated with tamoxifen, thus accounting but the fact that tamoxifen caused a decrease in for rises in gonadotropins, cholesterol, that was maintained during ICI 182,780 treatment is very intriguing. One is the possibility that ICI 182,780 is explanation that should be investigated, metabolized locally in the liver to an estrogen prior to biliary excretion. Serial endometrial ultrasound was performed in five responding patients. The endometrial thickness in all five patients was greater than that found in the normal postmenopausal uterus. However, this increased thickness was attributed to prior tamoxifen treatment. ICI 182,780 inhibits further endometrial proliferation but did not cause regression of pre-existing hypertrophied endometrium (Howell et al., 1996). in both ovariectomized (Dukes et al., 1992) and intact adult female (Dukes et demonstrate a complete blockade of the uterotrophic effects of estradiol by ICI 182,780. The pure antiestrogens could find use in the treatment of endometrial disorders and endometrial carcinoma. A clinical comparison with progestins might demonstrate fewer side-effects with a pure antiestrogen. Studies
al., 1993) monkeys
Molecular
Mechanisms
and Future Uses of Antiestrogens
Table 8. Clinical trials evaluating the pure antiestrogen
219
ICI 182,780
Study
Patient population
Dose/schedule
Response
Toxicity
DeFriend et al. (1994)
56 postmenopausal patients with histologically, confirmed primary breast cancer
In ER+ tumors, ER, PR and Ki 67 were significantly reduced after rx.
Minor/moderate headache 6
Howell et al. (1996)
19 postmenopausal, TAM-resistant, advanced breast cancer
Controls 10 patients 6mg,IMx7 days preop 21 patients 18mg,IMx7 days preop 16 patients 250 mg IM/ month
PR 37%
None
SD 32%
Soft tissue, bone, and visceral sites responded. Abbreviations:
TAM, tamoxifen;
IM, intramuscular.
Clinical Evaluations from clinical trials evaluating the activity of pure antiestrogens are limited (Table 8). DeFriend et al. (1994) assessed the tolerance, pharmacokinetics and shortterm biological activity of ICI 182,780 in 56 women with primary breast cancer. Patients were randomized to a control group (n = 19) in which no preoperative therapy was administered, or to one of two preoperative treatment groups (treatment treatment group 2: 18 mg IM x 7 days, group I : 6 mg IM x 7 days, n = 21 patients; n = 16 patients). ICI 182,780 was not associated with any significant toxicity.
Data
In a group of 19 tamoxifen-resistant, advanced breast cancer patients, Howell et al. (1996) reported a 37% partial response rate using ICI 182,780 (250 mg IM/month). Soft-tissue, bone and visceral sites of disease responded. In addition, 32% of patients maintained stable disease status. No significant toxicity was observed. The lack of cross-resistance with tamoxifen in 69% of patients suggest that ICI 182,780 may be useful as a first-line therapy in advanced disease or as a second-line therapy in advanced disease where tamoxifen has been previously used. The activity of pure antiestrogens in advanced disease suggest that they may also have efficacy in the adjuvant setting, although no data are available.
Chapter 8
Targeted Antiestrogens
Raloxifene (originally called keoxifene) is being developed by Eli Lilly Laboratories as a treatment for osteoporosis. The drug being referred to as a selective estrogen recept0.r modulator (SERM) which builds on the concept that targeted antiestrogens can be ifound to have estrogenic effects on the cardiovascular system and bone but an antiestrogenic action on the breast and uterus (Lerner and Jordan, 1990; Tonetti and Jordan, 1996). Raloxifene is also being evaluated as an antitumor agent in ER-positive advanced breast cancer patients.
Laboratory Studies Raloxifene has a binding affinity for the ER equivalent to that of estradiol (Black et al., 1983). The compound is a potent inhibitor of the growth of breast cancer cells in culture (Poulin et al., 1989). Raloxifene is an antiestrogen in the immature rat uterine weight ‘test but has little agonist action on the uterus well established that raloxifene can maintain bone (Jordan et al., 1987; Evans et al., 1994; Black et al., the drug also reduces circulating cholesterol (Black et Raloxifene has antitumor activity in induced (Gottardis and Jordan, Raloxifene-stimulated breast tumors inhibits the tamoxifen-stimulated EnCalOl, in athymic mice (Gottardis
when administered alone. It is density in the ovariectomized 1994; Sato et al., 1995) rat, but al., 1994).
the DMBA- (Clemens et al., 1983) and NMU1987) rat mammary carcinoma models. have not been described, but raloxifene partially growth of the human endometrial carcinoma, et al., 1990).
Toxicollogy No reports
of DNA adducts
or hepatocarcinogenesis
have appeared
with raloxifene.
Clinical Pharmacology and Endocrinology An analytical method to determine raloxifene and its metabolites has not been published. The drug causes a decrease in LDL cholesterol, but HDL cholesterol remains unchanged during treatment with 200 and 600 mg daily (Draper et al., 1993). 221
222
V. C. Jordan and W. J. Gradishar
Clinical Evaluation In the 1980s a series of phase I studies of raloxifene were carried out in healthy male subjects. A once-daily, oral dosing schedule was well tolerated, and acceptable blood levels were achieved. No clinically adverse events were detected, and there was evidence for an antiestrogenic effect (Draper et al., 1995). Physicians at the M.D. Anderson Hospital reported the results of a phase II trial of raloxifene in female patients with metastatic breast cancer who were refractory to tamoxifen therapy (Buzdar et al., 1988). Raloxifene, 200 mglday, was administered orally in accordance with the highest dose given in phase I studies. Fourteen patients received raloxifene daily for up to 8 months. The drug was well tolerated with no significant clinical or laboratory abnormalities detected, but no objective responses were observed (Buzdar et al., 1988). It is clear that raloxifene has no deleterious side-effects on breast cancer patients. As a result of the intense interest in raloxifene as a potential treatment for osteoporosis, a small international clinical trial is underway to evaluate the activity of raloxifene in hormone receptor-positive, postmenopausal patients with metastatic breast cancer who have not received prior hormonal therapy or chemotherapy for metastatic disease. The ability of raloxifene to maintain bone density in the rat (Jordan et al., 1987; Evans et al., 1994; Black et al., 1994; Sato et al., 1995) has encouraged the clinical testing of raloxifene as a treatment for osteoporosis. Large international clinical trials are currently underway to evaluate the effect of raloxifene on the progression of osteoporosis.
Chapter9
Comparison of New Antiestrogen and Conclusions
The successful- development of tamoxifen has created numerous new opportunities for the development of drugs that could be applied throughout medicine. In this review we have described the properties of several new agents being tested clinically but it is important to stress that the current development of these new agents is focused on several different therapeutic goals. Although this fact makes a direct comparison of compounds difficult, it is useful to evaluate the available data in the areas that have caused the most concern with tamoxifen. The principal areas of evaluation in the laboratory and clinic are shown in Table 9. Obviously additional information is available within the individual pharmaceutical companies to justify current clinical trials with regulatory authorities. However, it is possible to cite only published data in the medical literature. Nevertheless, compounds that are intended for prolonged usage must be evaluated clinically to the same high standards already achieved with tamoxifen to avoid misinterpretation of laboratory data that describes clinical. safety of new agent prematurely. Only carefully organised prospective clinical studies can provide the appropriate safety data. Additionally, for those agents that are intended to be used for the treatment of advanced breast cancer, it is essential to demonstrate the new drug is not cross
Table 9. A comparative
summary
DNA adducts Rat liver tumors Endometrial cancer (lab) Endometrial cancer (clinic) Cross-resistance with tamoxifen Advanced breast cancer Adjuvant therapy Cholesterol (+) (labor clinic) Preserves bones (+) (labor clinic)
of published reports about the toxicology antiestrogens Toremifene
Droloxifene
No No
No No
and application
of new
ICI 182,780 ICI 164,384
Raloxifene Keoxifene
Inhibit
Inhibit
Yes Yes
Yes
No Yes
Yes Yes
Yes
No
Yes Yes
a characteristic
was found
No, a report where a characteristic was not found; yes, a report where and a da.sh indicates no pubished studies. 223
224
V. C. Jordan and W. J. Gradishar
resistant with tamoxifen. All patients who present with stage I and II breast cancer that is ER-positive will be treated with adjuvant tamoxifen. There is clearly no point in treating disease that is resistant to tamoxifen with a cross resistant agent. This can be tested in the laboratory and must be addressed.
Overall, much progress has been made during the past decade. However, as illustrated in Table 9 there are still significant gaps in our knowledge about the efficacy and long-term safety of the new agents. A focused program of development in each of the targeted areas of therapeutics will result in the introduction of numerous new ‘antiestrogens’ (SERMS) into clinical practice by the end of the 1990s. Overall, the fascinating and multifaceted pharmacology of the nonsteroidal antiestrogens is converging to make new drugs available in diverse areas of medicine. This means that the knowledge of the value and side-effects of an anticancer drug tamoxifen, originally the exclusive domain of the oncologist, must be distributed to a wider constituency. As the original suggestions (Lerner and Jordan, 1990) that targeted antiestrogens should be sought to treat diseases of the menopause, and that as a beneficial side-effect the prevention of breast cancer becomes a reality, then issues of duration of treatment, endometrial cancer and agent-resistant breast cancer must become centre stage. It is known that long durations of treatment will be essential to maintain bone density and protect against coronary heart disease. But what of the growth of pre-existing endometrial cancer and the eventual development of agent resistant breast cancer? It is clear that, based on the experiences with tamoxifen and endometrial cancer and the epidemiology of breast cancer, these concerns will laboratory models exist to test the extrapolate into rare events. Nevertheless, hypothesis and provide guidance for prospective clinical trials and patient monitoring. Patient piece of mind and safety must be the goal of ethical pharmaceutical companies as we enter a new era of women’s health.
Acknowledgements We are especially grateful for Susan Tripoli typing and correcting our manuscript, Henry Muenzner for completing all the diagrams and to Ruth O’Regan, MD for her invaluable help with the references. We are deeply grateful to the Lynn Sage Foundation for their continuing support for our Breast Cancer Program. The program is also supported in part by a National Cancer Institute Breast Cancer Program Development Grant R21CA-65764.
References Abe, O., Enomote, K., Fujiwara, K., Izuo, M., Iino, Y., Tominaga, T., Hayashik Takatani, O., Kugai, N. and Yoshida, M. (1990). Phase I study of FK 435. Japanese ~oumal of C’ancer Clinics 36, 903-913. Abe,
O., Enomote, K., Fujiwara, K., Izumo, M., Iino, Y., Tominaga, T., Hayashik Takatani, O., Kugai, N. and Yoshida, M. (1991). Japanese early phase II study of droloxifene in the treatment of advanced breast cancer. American Journal of Clinical Oncology 14, 540-541.
Ahlemann, L. M., Staab, H. J., Loser, R., Seibel, K. and Huber, H. J. (1988). Inhibition of growth of human cancer by intermittent exposure to the antiestrogen droloxifene. Tumor Diagnosis and Therapy 9, 41-46. Allan,
G. F., Leng, X., Tsai, S. T., Weigel, N. L., Edwards, D. P., Tsai, M. J. and O’Malley, B. W. (1992). Hormone and antihormone induce distinct conformational changes which are central to steroid receptor activation. Journal of Biology and Chemistry 267, 19.513-19520.
Allen, 1~. E., Clark, E. R. and Jordan, V. C. (1980). Evidence for the metabolic activation of nonsteroidal antioestrogens: a study of structure activity relationships. British j%umal of Pharmacology 71, 87-91. Antilla, M., Valvaara, R., Kivinen, S. and Maenpaa, J. (1990). toremifene. Journal of Steroid Biochemistry. 36, 249-252.
Pharmacokinetics
of
Asaishi, K., Tominaga, T., Abe, O., Izuo, M. and Nomula, Y. (1993). Efficacy and safety of high dose NK 622 (toremifene citrate) in tamoxifen failed patients with breast cancer [in Japanese]. Can to Kagaku Ryoho 20, 91-99. Assikis, V. J. and Jordan, V. C. (1995). Gynecological effects of tamoxifen and the association with endometrial cancer. International Journal of Gynecology and Obstetrics 49, 241-257. Assikis, V. J., Neven, P., Jordan, V. C. and Vergote I. (1996). A realistic clinical perspective of tamoxifen and endometrial carcinogenesis. European Journal of Cancer 32A, 1464-1476. M. J. and Mosettig, E. (1960). Avigan, J., Steinberg, D., Vroman, H. E., Thompson, Studies of cholesterol biosynthesis: the identification of desmosterol in serum and tissues of animals and man treated with MER-29. Journal of Biology and Chemistry 235, 3123-3126. Barakat, R. R. (1997). Benign and hyperplastic Oncology 11 (Suppl), 35-37.
changes
associated
with tamoxifen
use.
Barakat, R. R., Wong, G., Curtin, J. D., Vlamis, V. and Hoskins, W. J. (1994). Tamoxifen use in breast cancer patients who subsequently develop corpus cancer is not
V. C. Jordan and W. J. Gradishar
226
associated with a higher Oncology 55, 164-168.
incidence
of adverse
Beatson, G. T. (1896). On the treatment of inoperable suggestions for a new method of treatment 104-107.
histological
features.
Gynecology
cases of carcinoma of the mamma: with illustrative cases. Lancet 2,
Bedford, G. R. and Richardson, D. N. (1966). Preparation and identification trans isomers of a substituted triarylethylene. Nature 212, 733-734. Bellmunt, J. and Sole, L. (1991). droloxifene in advanced breast 536-539.
and
of cis and
European early phase II dose finding study of cancer. American Journal of Clinical Oncology 14,
Bertelli, G., Pronzato, P., Amoroso, D., Cusmano, Bertolini, S. and Rosso, R. (1988). Adjuvant influence on plasma lipids and antithrombin Treatment 12, 307-3 10.
M. P., Conte, P. F., Montagna, G., tamoxifen in primary breast cancer: III. Breast Cancer Research and
Berthois, Y., Katzenellenbogen, J. A. and Katzenellenbogen, B. S. (1986). Phenol red in tissue culture media is a weak estrogen: implications concerning the study of estrogen responsive cells in culture. Proceedings of the National Academy of Science USA 83, 2496-2500. Berthou, F. and Dreano, Y. (1993). High performance liquid chromatographic analysis of tamoxifen, toremifene and their major human metabolites. r. Chromatogr B. Biomed. Appl. 616, 117-127. Bilimoria, M. M., Assikis, V. J. and Jordan, V. C. (1996). Should adjuvant tamoxifen treatment be stopped at 5 years ? Cancer Journal from Scientific American 2, 140-150. Bishop, J., Muray, R., Webster, L., Pitt, P., Stokes, K., Fennessy, A., Olveri, I. and Leber, G. (1992). Phase I clinical and pharmacokinetics study of high dose toremifene in postmenopausal patients with advanced breast cancer. Cancer Chemotherapy and Pharmacology 30, 174- 178. Black, L. J., Jones, C. D. and Falcone, J. F. (1983). Antagonism of estrogen action with a new benzothiophene-derived antiestrogen. Life Sciences 32, 1031-1036. Black, L. J., Sato, M., Rowley, E. R., Magee, D. E., Bekele, A., Williams, D. C., Cullinan, G. J., Bendele, R., Kauffman, R. F. and Bensch, W. R. (1994). Raloxifene (LY139481 HCl) prevents bone loss and reduces serum cholesterol without causing uterine hypertrophy in ovariectomized rats. Journal of Clinical Investigations 93, 63-69. Bloom, H. J. G., Roe, F. J. C. and Mitcheley, B. C. V. (1967). Sex hormones and renal neoplasia: inhibition of tumor of hamster kidney by an estrogen antagonist, an agent of possible therapeutic value in man. Cancer 20, 2118-2120. Boccardo, F., Bruzzi, P., Rubagotti, A., Nicolo, G. U. and Rosso, R. (1981). Estrogen-like action of tamoxifen on vaginal epithelium in breast cancer patients. Oncology 38, 281-2825.
Molecular
Mechanisms
and Future Uses of Antiestrogens
Bowler, J., Lilley, T. J., Pittman, J. D. and Wakeling, antiestrogens. Steroids 54, 71-99. Boyd, S. (1990). 1161-1167.
On oophorectomy
in cancer
227
A. E. (1989). Novel steroidal
of the breast.
British Medical
pure
Journal
2,
Breast Cancer Trials Committee, Scottish Trials Office (1987). Adjuvant tamoxifen in the management of operable breast cancer. The Scottish Trials. Lancet ii, 171-175. Bucourt, R., Vignau, M., Torelli, V., Richard-Roytt, A., Geynet, C., Secco-Millet, C., Redcailh, G. and Baulieu, E. E. (1978). New biospecific adsorbents for the purification of estradiol receptor. Journal of Biology and Chemistry 253, 8221-8228. Buzdar, A. U., Kau, S., Hortobagyi, G. N., Theriault, R. L., Booser, D., Holmes, F. A., Walters, R. and Krakoff, I. H. (1994). Phase I trial of droloxifene in patients with metastatic breast cancer. Cancer Chemotherapy and Pharmacology 33, 313-316. Buzdar, A. U., Marcus, C., Holmes, F., Hug, V. and Hortobagyi, G. N. (1988). Phase II evaluation of LY1567.58 in metastatic breast cancer. Oncology 45, 344-345. Canney, P. A., Griffiths, T., Latief, T. N. and Priestman, T. J. (1987). (Letter). significance of tamoxifen withdrawal response. Lancet i, 36.
Clinical
Carmichael, P. L., Ugwumadu, A. H. N., Neven, P., Herwe, A. J., Poon, G. K. and Phillips, D. H. (1996). Lack of genotoxicity of tamoxifen in human endometrium. Cancer Research 56, 1475-1479. Carnochan, P., Trivedi, M., Young, H., Eccles, S., Potter, G., Haynes, B. and Ott, R. (1994). Biodistribution and kinetics of pyrrolidino-4-iodo-tamoxifen: prospects for pharmacokinetic studies using PET. Journal of Nuclear Biology and Medicine 38, 96-98. Chander, S. K., McCague, R., Lugmani, Y., Newton, C., Dowsett, M., Jarman, M. and Coombes, R. C. (1991). Pyrrolidino-4-iodotamoxifen and 4-iodotamoxifen, new analogues of the antiestrogen tamoxifen for the treatment of breast cancer. Cancer Research 51, 5851-5858. Clemen’s, J. A., Bennett, D. R., Black, L. J. and Jones, C. D. (1983). Effects of a new antiestrogen keoxifene (LY156758) on growth of carcinogen induced mammary tumors and on LH and prolactin levels. L;fe Sciences 32, 2869-2875. Cole, M. P., Jones, C. T. A. and Todd, I. D. H. (1971). A new antioestrogenic agent in late breast cancer: an early clinical appraisal of ICI 46,474. British Journal of Cancer 25,270-275. Cole, M. P., Jones, C. T. A. and Todd, I. D. H. (1972). The treatment of advanced ca:rcinoma of the breast with the antineoplastic agent tamoxifen (ICI 46,474)-a selries of 96 patients. Advances in Antimicrobial Antineoplasia and Chemotherapy 2, 529-53 1. Colletta., A. A., Wakefield, L. M., Howell, F. V., Van Roozendaal, K. E., Danielpour, D., Ebbs, S. R., Sporn, M. B. and Baum, M. (1990). Antiestrogens induce the secretion
228
V. C. Jordan and W. J. Gradishar of active transforming growth Journal qf Cancer 62,405-409.
factor
beta
from
human
fetal
fibroblasts.
British
Cook, L. S., Weiss, N. S., Schwartz, S. M., White, E., M&night, B., Moore, D. E. and Daling, J. R. (1995). Population based study of tamoxifen therapy and subsequent ovarian, endometrial and breast cancers. Journal of the National Cancer Institute 87, 1359-1364. Coombes, R. C., Haynes, B. P., Dowsett, M., Quigley, M., English, J., Judson, I. R., Griggs, L. J., Potter, G. A., McCague, R. and Jarman, M. (1995). Idoxifene: report of a phase I study in patients with metastatic breast cancer. Cancer Research 55, 1070-1074. Dauvois, S., Danielian, P. S., White, R. and Parker, M. G. (1992). Antiestrogen ICI 164,384 reduces cellular estrogen receptor by increasing its turnover. Proceedings of the National Academy of Sciences USA 89, 4037-4041. Dauvois, S., White, R. and Parker, M. G. (1993). The antiestrogen ICI 182 780 disrupts estrogen receptor nucleocytoplasmic shuttling. Journal of Cell Science 106, 1377-1388. DeFriend, D. J., Howell, A., Nicholson, R. I., Anderson, E. T., Dowsett, M. L., Mansel, R. E., Blarney, R. W., Blandred, N. J., Robertson, J. F. and Saunders, C. (1994). Investigation of a pure new antiestrogen (ICI 182,780) in women with primary breast cancer. Cancer Research 54,408-414. DiSalle, E., Zaccheo, T. and Ornati, of the new triphenylethylene Biochemistry 36, 203-206.
G. (1990). Antioestrogenic and antitumour derivative toremifene in the rat. Journal
properties of Steroid
Dragan, Y. P., Langan-Fahey, S. M., Street, K., Vaughan, J., Jordan, V. C. and Pitot, H. C. (1994). Studies of tamoxifen as a promoter of hepatocarcinogenesis in female Fisher F344 rats. Breast Cancer Research and Treatment 31, 1 l-25. Dragan, Y. P., Nawaysir, E., Fahey, S., Vaughn, J., McCague, Jordan, V. C. and Pitot, H. C. (1996). Effects of tamoxifen and two fixed ring analogs on multistage rat hepatocarcinogenesis. Carcinogenesis 17, 585-594. Dragan, Y. P., Vaughan, J., Jordan, V. C. and Pitot, H. C. (1995). Comparison of tamoxifen and toremifene on liver and kidney tumor promotion in female rats. Carcinogenesis 16,2733-2741. Draper, M. W., Flowers, D. E. and Hester, W. J. (1993). Effects of raloxifene (LY 139481 HCL) on biochemical markers of bone and lipid metabolism in healthy postmenopausal women. Proceeding of the Fourth International Symposium on Osteoporosis. Handelstrykkeriet Aalborg ApS., Aalborg, Denmark. Draper, M. W., Flowers, D. E., Neild, J. A., Huster, W. J. and Zerbe, Antiestrogenic properties of raloxifene. Pharmacology 50, 209-217.
R. L. (1995).
Dukes, M., Chester, R., Yarwood, L. and Wakeling, A. E. (1994). Effects of a nonsteroidal pure antiestrogen ZM 189,154, on estrogen target organs of the rat including bones.Joumal of Endocrinology 141, 335-341.
Molecular
Mechanisms
and Future Uses of Antiestrogens
229
Dukes, M., Miller, D., Wakeling, A. E. and Waterton, J. C. (1992). Antiuterotrophic effect of a pure antioestrogen ICI 182,780: magnetic resonance imaging of the uterus in ovariectomized monkeys. Journal of Endocrinology 135, 239-247. Dukes, M., Waterton, J. C. and Wakeling, A. E. (1993). Antiuterotrophic effect of the pure antiestrogen ICI 182,780 in adult female monkeys (Macaca nemestrina): quantitative magnetic resonance imaging. Journal of Endocrinology 138,203-210. Duncan, G. W., Lyster, S. C. and Clark, J. J. (1963). Antifertililty dihydronaphthalene derivatives. Proceeding of the Society an,d Medicine 112,439-442.
activity of two diphenylof Experimental Biology
Early Breast Cancer Trialists’ Collaborative Group (1988). Effects of adjuvant tamoxifen and of cytotoxic therapy on mortality in early breast cancer. An overview of 61 randomized trials among 28,896 women. New England Journal of Medicine 319, 1681-1692. Early Breast Cancer Trialists’ Collaborative Group (1992). Systemic treatment of early breast cancer by hormonal, cytotoxic or immune therapy. 133 randomized trials involving 31,000 recurrences and 24,000 deaths among 75,000 women. Lancet 339, l-15. Ebbs, S. R., Roberts, J. and Baum, M. (1990). Response to toremifene (Fc-1157a) therapy in tamoxifen failed patients with breast cancer. Preliminary communication. Journal of Steroid Biochemisty 36, 239. Eppenberger, U., Wosikowski, properties of Droloxifene, 14, ss-s14. Ernst,
K. and Kung, W. (1991). Pharmacologic and biologic a new antiestrogen. American Journal of Clinical Oncology
S., Hite, G., Cantrell, J. S., Richardson, A. Jr. and Benson, H. D. (1976). Stereochemistry of geometric isomers of clomiphene: A correction of the literature and re-examination of structure activity relationships. Journal of Pharmacological Science 65, 148- 150.
Evans, G. L., Bryant, H. U., Magee, D., Sato, M. and Turner, R. T. (1994). The effects of raloxifene on tibia histomorphometry in ovariectomized rats. Endocrinology 134, 2283-2288. Falkson, H. C., Gray, R., Wolberg, W. A., Gilchrist, K. W., Harris, J. E., Tormey, D. C. and Falkson, G. (1990). Adjuvant trial of 12 cycles of CMFPT followed by observation or continuous tamoxifen versus four cycles of CMFPT in postmenopausal women with breast cancer. An Eastern Cooperative Oncology Group Phase III Study. Journal of Clinical Oncology 8, 599-607. Fawell, S. E., White, R., Hoare, S., Sydenham, M., Page, M. and Parker, M. G. (1990). Inhibition of estrogen receptor DNA binding by the pure antiestrogen ICI 164 384 appears to be mediated by impaired receptor dimerization. Proceedings of the National Academy of Science 87, 6883-6887. Fisher, El., Costantino, J. P., Redmond, C. K., Fisher, E. R., Wickerman, D. L. and Clonin, W. M. (1994). Endometrial cancer in tamoxifen treated breast cancer patients:
V. C. Jordan and W. J. Gradishar
230
findings from the National 14. Journal of the National
Surgical Adjuvant Breast and Bowel Project Cancer Institute 86, 527-537.
(NSABP)
B-
Fisher,
B., Costantino, J., Redmond, C., Poisson, R., Bowman, D., Couture, J., Dimitrov, N. V., Wolmark, N., Wickerman, D. L. and Fisher, E. R. (1989). A randomized clinical trial evaluating tamoxifen in the treatment of patients with node-negative breast cancer who have estrogen receptor positive tumors. New England Journal of Medicine 320, 429-434.
Fisher, B., Redmond, C., Brown, A., Fisher, E. R., Wolmark, N., Bowman, D., Plotkin, D., Wolter, J., Bornstein, R. and Legault-Poisson, S. (1986). Adjuvant chemotherapy with and without tamoxifen in the treatment of primary breast cancer. 5 year results from the National Surgical Adjuvant Breast and Bowel Project Trial. Journal of Clinical Oncology 4, 459-471. Fisher,
B., Redmond, C., Brown, A., Wickerman, D. L., G., Lippman, M., Savlov, E. and Wittliff, J. (1983). progesterone receptor levels on the response to primary breast cancer. Journal of Clinical Oncology
Wolmark, N., Allegra, J., Escher, Influence of tumor estrogen and tamoxifen and chemotherapy in 1,227-241.
Fornander, T., Rutqvist, L. E., Cedermark, B., Glas, U., Mattsson, A., Silfverswaard, C., Skoog, L., Somell, A., Theve, T. and Wilking, N. (1989). Adjuvant tamoxifen in early breast cancer: occurrence of new primary cancers. Lancet i, 117-120. Fornander, T., Rutqvist, L. E., Sjoberg, H. E., Blomqvist, L., Mattsson, A. and Glas, U. (1990). Long term adjuvant tamoxifen in early breast cancer: effect on bone mineral density in postmenopausal women. Journal of Clinical Oncology 8, 1019-1024. Fran&
I. D., Mobberley, M. L. and Schroepfer, G. T. (1966). Effects of MER-29 on the intermediate metabolism of cholesterol. Progress in Cardiozjascular Disorders 2, 51 l-518.
Furr,
B. J. A. and Jordan, V. C. (1984). The pharmacology Pharmacology and Therapeutics 25, 127-205.
and clinical
uses of tamoxifen.
Gallagher, A., Chambers, T. J. and Tobias, J. H. (1993). The estrogen antagonist 182,780 reduces cancellous bone volumes in female rats. Endocrinology 2787-2791. Gaylor, J. L. (1963). Biosynthesis of skin sterols II. Reactions of AZ4 unsaturated of rat skin. Archives of Biochemisty and Biophysics 101, 108-l 15.
ICI 133,
sterols
Geisler, J., Ekse, D. and Hosch, S. (1995). Influence of droloxifene (3-hydroxytamoxifen) 40 mg daily on plasma gonadotrophins, sex hormone binding globulin and estrogen levels in postmenopausal breast cancer patients. Journal of Steroid Biochemistry and Molecular Biology 55, 193-195. Gibson, M. K., Nemmers, L. A., Beckman, W. C., Davis, V. L., Curtis, S. W. and Korach, K. S. (1991). The mechanism of ICI 164,384 antiestrogenicity involves rapid loss of estrogen receptor in uterine tissue. Endocrinology 129, 2000-2010. Gorski, J., Toft, D., Shyamala, studies on the interaction Research 24, 45-80.
G., Smith, D. and Notides, A. (1968). Homone receptors: of estrogen with the uterus. Recent Progress in Hormone
Molecular Mechanisms and Future Uses of Antiestrogens Gottardis,
M. M. and Jordan, V. C. (1987). The tamoxifen in the N-nitrosomethylurea-induced Carzcer Research 47. 4020-4024.
231
antitumor action of keoxifene and rat mammary carcinoma model.
Gottardis, M. M. and Jordan, V. C. (1988). Development of tamoxifen stimulated growth of MCF-7 tumors in athymic mice after long-term antiestrogen administration. Cancer Research 48, 5183-5187. Gottardis, M. M., Ricchio, M. D., Satyaswaroop, P. G. and Jordan, V. C. (1990). Effect of steroidal and nonsteroidal antiestrogens on the growth of a tamoxifen-stimulated human endometrial carcinoma (EnCalOl) in athymic mice. Cancer Research 50, 3189-3192. Gottardis, M. M., Robinson, S. P., Satyaswaroop, P. G. and Jordan, V. C. (1988). Contrasting actions of tamoxifen on endometrial and breast tumor growth in the athymic mouse. Cancer Research 48, 812-815. Gottardis, M. M., Wagner, R. J., Borden, E. C. and Jordan, V. C. (1989a). Differential ability of antiestrogens to stimulate breast cancer cell (MCF-7) growth in zlivo and in vitro. Cancer Research 49, 4765-4769. Gottardis, M. M., Jiang, S. Y., Jeng, M. H. and Jordan, V. C. (1989b). Inhibition of tamoxifen stimulated growth of an MCF-7 tumor variant in athymic mice by novel steroidal antiestrogens. Cancer Research 49, 4090-4093. Greaves, P., Goonetilleke, R., Nunn, G., Topham, J. and Orton, T. (1993). Two year carcinogenicity study of tamoxifen in Alderley Park Wistar-derived rats. Cancer Research 53, 3919-3924. Green,
S. and Chambon, P. (1991). The oestrogen receptor: from perception to mechanism. In: Nuclear Homzone Receptors (Parker, M. G., eds), pp. 1.5-38. Academic Press, New York.
Green,
S., Walter, P., Kumar, V., Krust, A., Bornert, J. M., Argos, P. and Chambon, P. (1986). Human oestrogen receptor cDNA: Sequence, expression and homology with v-erb A. Nature 320, 139-143.
Greenblatt, R. B., Barfield, W. E., Jungck, E. C. and Ray, A. W. (1961). Induction of ovulation with MRL-41. Journal of the American Medical Association 178, 101-104. Greenblalt, R. B., Roy, S., Mahesh, V. B., Barfield, W. E. and Jungck, E. L. (1962). Induction of ovulation with clomiphene citrate. American Journal of Obstetrics and Gynecology 84,900-912. Greene, ‘G. L., Gilna, P., Waterfield, M., Baker, A., Hort, Y. and Shine, J. (1986). Sequence and expression of human estrogen receptor complementary DNA. Science 231, 1150-l 154. Greenwal’d, G. S. (1965). Effects of a dihydronaphthalene pregnancy in the rat. Fertility and Sterility 16, 185-194.
derivative
(U-l 1, 1OOA) on
Grenman, R. L., Laine, K. M., Klemi, P. J., Grenman, S., Hayashida, D. J. and Joensuu, H. (1991). Effect of the antiestrogen toremifene on growth of the human mammary
232
V. C. Jordan and W. J. Gradishar
carcinoma cell line MCF-7. Journal of Cancer Research 223-226.
and Clinical Oncology 117,
Grill, H. J. and Pollow, K. (1991). Pharmacokinetics of droloxifene and its metabolites breast cancer patients. American Journal of Clinical Oncology 14(2), s21-~29.
in
Gunderson, S. (1990). Toremifene, a new antiestrogenic compound in the treatment of advanced breast cancer. Phase II study. European Journal of Cancer 24A, 785-790. Gylling, H., Pyrhonen, S., Mantyla, E., Maenpaa, H., Kangas, L. and Miettinen, T. A. (1995). Tamoxifen and toremifene lower serum cholesterol by inhibition of A8-cholestenol conversion to lathosterol in women with breast cancer. Journal of Clinical Oncology 13,2900-2905. Haarstad, H., Gundersen, S., Wist, E., Raabe, N., Mella, 0. and Kvinnsland, S. (1992). Droloxifene-a new anti-estrogen phase II study in advanced breast cancer. Acta Oncologica 31, 425-428. Haddow, A., Watkinson, J. M. and Paterson, E. (1994). Influence of synthetic upon advanced malignant disease. British Medical Journal 2, 393-398.
oestrogens
Halachmi, S., Marden, E., Martin, G., MacKay, H., Abbondanza, C. and Brown, M. (1993). Estrogen receptor associated proteins possible mediators of hormone-induced transcription. Science 264, 1455-1458. Hamm, J. T., Tormey, D. C., Kohler, P. C., Haller, D., Green, Phase I study of toremifene in patients with advanced Oncology 9, 2036-2041. Han,
X. and Liehr, J. G. (1992). Induction of covalent tamoxifen. Cancer Research 52, 1360-1363.
M. and Shemano, I. (1991). cancer. Journal of Clinical
DNA
adducts
in rodents
by
Hard, G. C., Iatropoulos, M. J., Jordan, K., Radi, K., Kaltenberg, 0. P., Imondi, A. R. and Williams, G. M. (1993). Major differences in the hepatocarcinogenicity and DNA adduct forming ability between toremifene and tamoxifen in female Crl;CD (BR) rats. Cancer Research 53, 4534-4541. Harper, M. J. K. and Walpole, A. L. (1966). Contrasting endocrine activities of cis and trans isomers in a series of substituted triphenylethylenes. Nature 212, 87. Harper, M. J. K. and Walpole, A. L. (1967). A new derivative of triphenylethylene: effect on implantation and mode of action in rats. Journal of Reproduction and Fertility 13, 101-119. Hasman, M., Rattel, B. and Loser, Letters 84, 101-l 16.
R. (1994).
Preclinical
data for droloxifene.
Cancer
Hayes, D. F., Van Zyl, J. A., Hacking, A., Goedhals, L., Bezwoda, W. R., Maillard, J. A., Jones, S. E., Vogel, C. L., Berus, R. F. and Shemano, I. (1995). Randomized comparison of tamoxifen and two separate doses of toremifene in postmenopausal patients with metastatic breast cancer. Journal of Clinical Oncology 113, 2556-2566.
Molecular Mechanisms and Future Uses of Antiestrogens
233
Hecker, E., Vegh, I., Levy, C. M., Magin, C. A., Martinez, J. C., Loureiro, J. and Garola, R. E. (1974). Clinical trial of clomiphene in advanced breast cancer. Europeam Journal of Cancer 10, 747-749. Herbst, A. L., Griffiths, C. T. and Kistner, R. W. (1964). Clomiphene citrate nated mammary carcinoma. Cancer Chemotherapy Reports 43, 39-41.
in dissemi-
Hietanen, T., Baltina, D., Johansson, R., Numminen, S., Hakala, T., Helle, L. and Valavaa.ra, R. (1990). High dose toremifene (240 mg daily) is effective as first line hormonal treatment in advanced breast cancer-an ongoing phase-II multicenter Finnish-Latvian cooperative study. Breast Cancer Research and Treatment 16, 37-40. Hindy, I., Juhos, E., Szanto, J. and Szamel, I. (1990). Effect of toremifene in breast cancer patients. Preliminary communication. Journal of Steroid Biochemistry 36, 225-226. Hirsimalti, P., Hirsimaki, Y., Nieminen, L. and Payne, B. J. (1993). Tamoxifen induces hepatocellular carcinoma in rat liver: A 1 year study with 2 antiestrogens. Archives of ‘Toxicology 67, 49-59. Hollander, W., Chobanian, A. V. and Wilkins, R. W. (1960). The effects of triparanol (M:ER-29) in subjects with and without coronary artery disease. Journal of the American Medical Association 174, !87-94. Holtkamp, D. E. (1987). Research and development of clomiphene citrate. In: Recent Advances in Human Reproduction (Asch, R. H., ed.), pp. 17-27. Fondaizione per Gli Studi sulla riproduzione Umana. Holtkamp, D. E., Greslin, S. C., Root, C. A. and Lerner, L. J. (1960). Gonadotropin inhibiting and antifecundity effects of chloramiphene. Proceedings of the Society of Experimental Biology and Medicine 105, 197-201. Homesley, H. D., Shemano, I., Gams, R. A., Harry, D. S., Hickock, P. G., Rebar, R. W., Bun, R. C., Mullin, T. J., Wentz, A. C. and O’Toole, R. V. (1993). Antiestrogenic potency of toremifene and tamoxifen in postmenopausal women. American Journal of Clinical Oncology 16, 117-122. Howell, A., DeFriend, D. J., Robertson, J. F. R., Blarney, R. W., Anderson, L., Anderson, E., Sutcliffe, F. A. and Walton, P. (1996). Pharmacokinetics, pharmacological and anti-tumour effects of the specific anti-oestrogen ICI 182,780 in women with breast cancer. British ‘Journal of Cancer 74, 300-308. Howell, .A., DeFriend, D., Robertson, J., Blarney, R. and Walton, specific antioestrogen (ICI 182,780) in tamoxifen-resistant 34!i, 29-30.
P. (1995). Response to a breast cancer. Lancet
Howell, A., Dodwell, D. J., Anderson, H. and Redford, J. (1992). Response after withdrawal of tamoxifen and progestogen in advanced breast cancer. Annals of Oncology 3, 611-617. Huovinen, R. L., Alanen, K. A. and Collan, Y. (1995). Cell proliferation in dimethylbenz (A) anthracene-induced rat mammary carcinoma treated with antiestrogen toremifene. Acta Oncologica 34, 479-485.
234
V. C. Jordan and W. J. Gradishar
Huovinen, R., Kellokumpu-Lehtinen, P.-L. I. and Collanm, Y. (1994). Evaluating the response to antiestrogen toremifene treatment in DMBA-induced rat mammary carcinoma. International Journal of Experimental Pathology 75, 257-263. Huovinen, R., Warri, A. and Collan, Y. (1993). Mitotic activity, apoptosis and TRPM-2 messenger RNA expression in DMBA-induced rat mammary carcinoma treated with antiestrogen toremifene. InternationalJournal of Cancer 55, 685-691. Huppert, L. C. (1979). Sterility 31, l-8.
Induction
of ovulation
with
clomiphene
citrate.
Fertility
and
Iino, Y., Takai, Y., Ando, T., Chwada, S., Yokoe, T., Sugamata, N., Takei, H., Houghchi, J., Iijima, K. and Morishita, Y. (1994). A new triphenylethylene derivative TAT-59: hormone receptors, insulin-like growth factor 1 and growth supression of hormone dependent MCF-7 tumors in athymic mice. Cancer Chemotherapy and Pharmacology 34, 372-376. Jaiyesimi, I. A., Buzdar, A. U., Decker, D. A. and Hortobagyi, G. N. (1995). Use of tamoxifen for breast cancer twenty eight years later. Journal of Clinical Oncology 13, 513-529. Jensen,
E. V. and Jacobson, H. I. (1962). Basic guides to the mechanism action. Recent Progress in Hormone Research 18, 387-414.
of estrogen
Jiang, S. Y., Wolf, D. M., Yingling, J. M., Chang, C. and Jordan, V. C. (1992). An estrogen receptor positive MCF-7 clone that is resistant to antiestrogens and estradiol. Molecular and Cell Endocrinology 90, 77-86. Jones,
C. D., Jevnikar, M. G., Pike, A. J., Peters, M. K., Black, L. J., Thompson, A. R., Falcone, J. F. and Clemens, J. A. (1984). Antiestrogens 2, structure activity studies in a series of 3-aroyl-2-(4-hydroxyphenyl)benzo [b] thiene-3-yl] [4-[2- (l-piperidinyl) ethoxy] phenyl] methanone hydrochloride (LY156,758) a remarkably effective estrogen antagonist with only minimal intrinsic estrogenicity. Journal of Medicine and Chemistry 27, 1057-1066.
Jones, C. D., Suarez, T., Massey, E. H., Black, L. J. and Tinsley, F. C. (1979). Synthesis and antiestrogenic activity of [3,4-dihydro-2-(4-methoxyphenyl)-1-naphthenyl] [4-(2-( 1-p-pyrrolidinyl ethoxy)-phenyl] methanone methane sulfonic acid. journal of Medicine and Chemisty 22, 962-966. Jiinsson, P. E., Malmberg, M., Bergljung, L. and Ingvar, C. (1991). Phase II study of high dose toremifene in advanced breast cancer progressing during tamoxifen treatment. Anticancer Research 11,873-876. Jordan, V. C. (1974). Antitumour activity of the antioestrogen ICI 46,474 (tamoxifen) in the dimethylbenzanthracene (DMBA)-induced rat mammary carcinoma model. Journal of Steroid Biochemisty 5, 354. Jordan, V. C. (1976). Effect of tamoxifen induced rat mammary carcinomata.
(ICI 46,474) on initiation and growth of DMBAEuropean Journal of Cancer 12,419-424.
Jordan, V.C. (1978). Use of the DMBA-induced rat mammary carcinoma system for the evaluation of tamoxifen as a potential adjuvant therapy. Rev. Endocrine Rel. Cancer October suppl, 49-55.
Molecular Mechanisms and Future Uses of Antiestrogens
235
Jordan, V. C. (1983). Laboratory studies to develop general principles for the adjuvant treatment of breast cancer with antiestrogen: problems and potential for future c’linical applications. Breast Cancer Research and Treatment 3, 573-586. Jordan, V. C. (1984). Biochemical Reviews 36.245-276.
pharmacology
of antiestrogen
action. Pharmacological
Jordan, V. C. (1988). The development of tamoxifen for breast cancer therapy: a tribute the late Arthur L. Walpole. Breast Cancer Research and Treatment 11, 197-209.
to
Jordan, V. C. (1993). Current view of the use of tamoxifen for the treatment and prevention of breast cancer. Gaddum Memorial Lecture. British Journal of Pharmacology 1.10,507-517. Jordan, V. C. (ed). (1994). Long-term OSWisconsin Press, Madison.
Tamoxifen
Treatment for Breast Cancer. University
Jordan, V. C. (1995a). Alternate antiestrogens and strategies cancer. Journal of Cell Biochemisty 22, 557-561. Jordan, V. C. (1995b). Tamoxifen and tumorigenicity: Nationa Cancer Institute 87, 623-626. Jordan,
V. C. (199%). What if tamoxifen 1973? Annals of Oncology 6, 29-33.
had been
for the prevention
a predictable
found
concern. Journal of the
to produce
Jordan, V. C. and Assikis, V. J. (1995). Tamoxifen and endometrial controversy. Clinical Cancer Research 1, 467-472.
of breast
rat liver tumors
cancer:
clearing
in
up a
Jordan, V. C. and Gosden, B. (1983a). Differential antiestrogen actions in the immature rat uterus: a comparision of hydroxylated antiestrogens with high affinity for the estrogen receptor. Journal of Steroid Biochemistry 19, 1249-1258. Jordan, V. C. and Gosden, B. (1983b). Inhibition of the uterotrophic activity of estrogens and antiestrogens by the short acting antiestrogen LY 117 018. Endocrinology 113, 463-468. Jordan, V. C. and Morrow, M. (1994). Should clinicians be concerned about the carcinogenic potential of tamoxifen? European Journal of Cancer 3OA, 1714- 1721. Jordan, V. C., Collins, M. M., Rowsby, L. and Prestwich, metabolite of tamoxifen with potent antioestrogenic ogy 75, 305-316.
G. (1977). A monohydroxylated activity. Journal of Endocrinol-
Jordan, V. C., Dix, C. J. and Allen, K. E. (1979). The effectiveness of long-term treatment in a laboratory model for adjuvant hormone therapy of breast cancer. In: Adjuvant Therapy of Cancer. II (Salmon, S. E. and Jones, S. E., eds), pp. 19-26. Grune and Stratton, New York. Jordan,
V. C., Dix, C. J., Naylor, K. E., Prestwich, G. and Rowsby, L. (1978). Nonsteroidal antiestrogens: their biological effects and potential mechanisms of action. Journal of Toxicology and Environmental Health 4, 364-390.
236
V. C. Jordan and W. J. Gradishar
Jordan, V. C., Feniuk, L., Allen, K. E., Cotton, R. C., Richardson, D., Walpole, A. L. and Bowler, J. (1981). Structural derivatives of tamoxifen and e&radio1 3-methyl ether as potential alkylating antioestrogens. European ‘Journal of Cancer 17, 193-200. Jordan, V. C., Lababidi, M. K. and Langan-Fahey, S. (1991). Suppression of mouse mammary tumorigenesis by long term tamoxifen therapy. Journal of the National Cancer Institute 83, 492-496. Jordan, V. C., Lieberman, M. E., Cormier, E., Koch, R., Bagley, J. R. and Ruenitz, P. C. (1984). Structural requirements for the pharmacological activity of non-steroidal antiestrogens in vitro. iklolecular Pharmacology 2261, 272-278. Jordan, V. C., Phelps, E. and Lindgren, J. U. (1987). Effects of antiestrogens castrated and intact female rats. Breast Cancer Research and Treatment
on bone in
10.31-35.
Jordan, V. C., Wolf, M., Mirecki, D. M., Whitford, D. A. and Welshons, W. V. (1988). Hormone receptor assays: clinical usefullness in the management of carcinoma of the breast. Critical Reviews of Clinical Laboratoy Science 26, 97-1.52. Kangas, L. (1990a). Review of the pharmacological Steroid Biochemistry 36, 191-195.
properties
of toremifene.
Kangas, L. (1990b). Biochemical and pharmacological effects of toremifene Cancer Chemotherapy and Pharmacology 27, 8-12.
Journal
of
metabolites.
Kangas, L., Nieminen, A.-L., Blanco, G., Gronroos, M., Kallio, S., Karjalainen, A., Perilla, M., Sodervall, M. and Tiovola, R. (1986). A new triphenylethylene compound Fc1157a. II antitumor effects. Cancer Chemotherapy and Pharmacology 17, 109-l 13. Katzenellenbogen, B. S., Kendra, K. L., Norman, M. J. and Berthois, Y. (1987). Proliferation, hormonal responsiveness and estrogen receptor content of MCF-7 human breast cancer cell in the short and long-term absence of estrogens. Cancer Research 47. 4355-4360. Kawamura, I., Mizota, T., Kondo, N., Shimomura, L. and Kohsaka, M. (1991). Antitumor effects of droloxifene, a new antiestrogen drug against 7,12 dimethylbenz (a) anthracene-induced mammary tumor in rats. Japan ‘Journal of Pharmacology 57,215-224. Kawamura, I., Mizota, T., Lacey, E., Tanaka, Y., Manda, T., Shimomura, K. and Kohsaka, M. (1993). Pharmacologic and biologic properties of droloxifene, a new antiestrogen. Japan Journal of Pharmacology 63,27-34. Ke, H. Z., Simmons, H. A., Pirie, C. M., Crawford, Droloxifene, a new estrogen antagonist/agonist, rats. Endocrinology 136, 2435-2441. Kirby,
D. T. and Thompson, D. D. (1995). prevents bone loss in ovariectomized
T. J,, Achorm, R. W. P., Perry, H. 0. and Winkelmann, R. K. (1962). formation after triparanol therapy. Archives Ophthalmology 68, 486-491.
Kistner, R. B. (1965). Induction of ovulation and Gynecology Surveys 20, 873-900. Kistner, R. W. and Smith, 0. estrogen antagonist-MER-25.
with clomiphene
citrate
W. (1960). Observations on the Surgical Forum 10,725-729.
(Clomid).
use
Cataract
Obstetrics
of non-steroidal
Molecular
Mechanisms
and Future Uses of Antiestrogens
237
Kivinen, S. and Maenpaa, J. (1990). Effect of toremifene on clinical hematological and hormonal parameters at different dose levels in healthy postmenopausal volunteers: phase I study. Journal of Steroid Biochemistry 36,217-220. Knabbe, C., Zugmaier, G., Schmahl, M., Dietel, M., Lippman, M. E. and Dickson, R. B. (1991). Induction of transforming growth factor beta by the antiestrogens droloxifene, tamoxifen and toremifene in MCF-7 cells. American Journal of Clinical Oncology 14, 15-20. Koh, J. I.., Kubota, T., Asanuma, F., Yamada, U., Kawamura, E., Hosoda, Y., Hashimoto, M., Yamamoto, O., Sakai, S. and Maeda, K. (1992). Antitumor effect of triphenylethylene derivative (TAT-59) against human breast carcinoma xenografts in nude mice. Journal of Surgical Oncology 51,254-258. Kohler, :P. C., Hamm, J. F., Wiebe, V. J., DeGregario, M. W., Shemano, I. and Tormey, C. (1991). Phase I study of the tolerance and pharmacokinetics of tormifene pauents with cancer. Breast Cancer Research and Treatment 16, 19-26.
D. in
clinical Konstantinova, M. M. and Gershanovich, M. L. (1990). R esults of comparative evaluation of antiestrogens toremifene and tamoxifen in locally advanced and disseminated breast cancer [in Russian] Vopr. Onkol. 36, 1182-l 186. Kraft, R. 0. (1962). Triparanol in the treatment Cancer Chemotherapy Reports 25, 113-l 15.
of disseminated
mammary
carcinoma.
Kristensen, B., Ejlertsen, B., Dalgaard, P., Larsen, L., Holmegaard, S. N., Transbol, I. and Mouridsen, H. T. (1994). Tamoxifen and bone metabolism in postmenopausal low risk breast cancer patients: a randomized study. Journal of Clinical Oncology 12, 99i!-997. Laughlin, R. C. and Carey, T. F. (1962). Cataracts in patients Journal of the American Medical Association 181, 339-340.
treated
with triparanol.
Lee, R. W., Buzdar, A. U., Blumenschein, G. R. and Hortobagyi, G. N. (1986). Trioxifene mesylate in the treatment of advanced breast cancer. Cancer 57, 40-43. Legha,
S. S., Slavik, M. and Carter, S. K. (1976). Nafoxidine-an treatment of breast cancer. Cancer 38, 1535-1541.
Lien,
E. A., Anker, G., Lonning, P. E. and Veland, D. M. (1995). Determination of droloxifene and two metabolites in serum by high pressure liquid chromatography. Therapy and Drug Monitoring 17, 259-265.
Lerner, L. J. (1981). The first non-steroidal antioestrogen-MER-25. Am’ioestrogens: Molecular Pharmacology and Ant&mow Activity and Jordan, V. C., eds), pp. 1-16. Academic Press, Sydney.
antiestrogen
for the
In: Non-steroidal (Sutherland, R. L.
Lerner, L. J. and Jordan, V. C. (1990). Development of antiestrogens and their breast cancer. Eighth Cain Memorial Lecture. Cancer Research 50,4177-4189. Lerner, L. J., Holthaus, J. F. and Thompson, C. R. (1958). A non-steroidal antagonist l-(p-2-diethylaminoethoxyphenyl)-l-phenyl-2-p-metho~henyl-ethanol. Endocrinology 63, 295-3 18.
use in
estrogen
238
V. C. Jordan and W. J. Gradishar
Lieberman, M. E., Gorski, J. and Jordan, V. C. (1983). An estrogen receptor model to describe the regulation of prolactin synthesis by antiestrogens in vitro. Journal of BioIogy and Chemistry 258, 4741-4745. Lim, C. K., Zhi-Kin, Y., Lamb, J. H., White, I. N. H., De Matteis, F. and Smith, L. L. (1994). A comparative study of tamoxifen metabolism in female rat, mouse and human liver microsomes. Carcinogenesis 15,589-593. Loser,
R., Seibel, K. and Eppenberger, U. (1985a). No loss of estrogenic and antiestrogenic activity after demethylation of droloxifene (3-OH tamoxifen). International Journal of Cancer 36, 701-703.
Loser,
R., Seibel, K., Roos, W. and Eppenberger, U. (198510). In vivo and in vitro antiestrogenic action of 3-hydroxy-tamoxifen, tamoxifen and 4-hydroxy-tamoxifen, European Journal of Cancer. Clinical Oncology 21, 900-985.
Love, R. R., Barden, H. S., Mazess, R., Epstein, S. and Chappell, R. J. (1994). Effect of tamoxifen on lumber spine bone mineral density in postmenopausal women after 5 years. Archives of Internal Medicine 154, 2585-2586. Love, R. R., Mazess, R. B., Barden, H. S., Epstein, S., Newcomb, P. A., Jordan, V. C., Carbone, P. P. and De Mets, D. L. (1992). Effects of tamoxifen on bone mineral density in postmenopausal women with breast cancer. New England Journal of Medicine 326, 852-856. Love, R. R., Wiebe, D. A., Feyzi, J. M., Newcomb, P. A. and Chappell, R. J. (1995). Effects of tamoxifen on cardiovascular risk factors in postmenopausal women after 5 years of treatment. Journal of the National Cancer Institute 86, 1534-1539. Love, R. R., Weibe, D. A., Newcomb, P. A., Cameron, L., Leventhal, H., Jordan, V. C., Feyzi, J. and DeMets, D. L. (1991). Effects of tamoxifen on cardiovascular risk factors in postmenopausal women. Annals of Internal Medicine 115,860-864. Maenpaa, J., Sonderatrom, K. O., Gronroos, M., Taina, E., Ganba, A. and Kangas, L. (1990). Effect of toremifene on estrogen primed vaginal mucosa in postmenopausal women. Journal of Steroid Biochemistry 36, !221-223. Magriples, U., Naftolin, F., Schwartz, P. E. and Carcangui, M. L. (1993). High grade endometrial carcinoma in tamoxifen treated breast cancer patients. Journal of Clinical Oncology l&485-490. Manni, A., Arafah, B. and Pearson, 0. H. (1981). Changes in endocrine status following antiestrogen administration to premenopausal and postmenopausal women. In: Nonsteroidal Antioestrogens. Molecular Pharmacology and Antitumour Activity (Sutherland, R. L. and Jordan, V. C., eds), pp. 435-452. Academic Press, Sydney. Martin, E. A., Rich, K. J., White, I. N., Woods, K. L., Powles, T. J. and Smith, L. L. (1995). 32P-Past labelled DNA adducts in liver obtained from women treated with tamoxifen. Carcinogenesis 16, 1651- 1654. Martin, P. M., Berthois, Y. and Jensen, E. V. (1988). Binding of antiestrogen exposes an occult antigenic determinant in the human estrogen receptor. Proceedings of the National Academy of Science 85,2533-2537,
Molecular
Mechanisms
and Future Uses of Antiestrogens
239
McCague, R., LeClercq, G., Legros, N., Goodman, J., Blackburn, G. M., Jarman, M. and Foster, A. P. (1989). Derivatives of tamoxifen. Dependency of estrogenicity on the 4 substituent. Journal of Medicine and Chemisty 32,2527-2533. McCague, R., Parr, I. B. and Haynes, B. P. (1990). Metabolism of the 4 iodo derivative of tamoxifen by isolated rat hepatocytes. Demonstration that the iodine atom reduces metabolic conversion and identification of four metabolites. Biochemical Pharmacology 40,2277-2283. McDonald, C. C. and Stewart, H. J. (1991). Fatal myocardial tamoxifen trail. British MedicalJournal 303, 435-437.
infarction
in the Scottish
McDonald, C. C., Alexander, F. E., Whyte, B. W., Forrest, A. P., McDonald, C. C. and Stewart, H. J. (1995). Cardiac and vascular morbidity in women receiving adjuvant tamoxifen for breast cancer in a randomized trial. British Medical Journal 311, 977-980. McDonnell, D. P., Clemm, D. L., Hermann, T., Goldman, M. E. and Pike, J. W. (1995). Analysis of estrogen receptor function reveals three distinct classes of antiestrogens. Molecular Endocrinology 9, 659-669. McGuiree, W. L., Carbone, P. P. and Vollmer, E. P. (eds) E&man Breast Cancer. Raven Press. New York. W. S. Merrill Company (1967). Clomiphene Therapeutics 8, 891-897. Metzger, D., White, J. and Chambon, in yeast. Nature 334, 31-36.
Citrate
(1975). Estrogen Receptors
(Clomid).
P. (1988). The human
Clinical Pharmacology
estrogen
receptor
in
and
functions
Metzler, M. and Schiffmann, D. (1991). Structural requirements for the in vitro transof Syrian hamster embryo cells by stilbene estrogens and formation triphenylethylene-type antiestrogens. American Journal of Clinical Oncology 14(2), s30-s35. Modig, H., Borgstrom, M., Nilsson, I. and Westman, G. (1990). Phase II clinical study of toremifene in patients with metastatic breast cancer. Journal of Steroid Biochemistry 36, 235-236. Moorthy, B., Sriram, P., Pathak, D., Bodell, W. J. and Randerath, K. (1996). Tamoxifen metabolic activation: comparison of DNA adducts formed by microsomal and chemical activation of tamoxifen and 4-hydroxytamoxifen with DNA adducts formed in viva. Cancer Research 56, 53-57. Morrow, M. and Jordan, V. C. (1993). Molecular mechanism of resistance therapy in breast cancer. Archives of Surgery 128, 1187-l 191.
to tamoxifen
Muhleman, K., Cook, L. S. and Weiss, N. S. (1994). The incidence of hepatocellular carcinoma in US white women with breast cancer after the introduction of tamoxifen in 1977. Breast Cancer Research and Treatment 30, 201-204. Murphy, C. S., Pink, J. J. and Jordan, V. C. (1992). Characterization of a receptor-negative, hormone non responsive clone derived from T47D human breast cancer cell line kept under estrogen free conditions. Cancer Research 50, 7285-7292.
240
V. C. Jordan and W. J. Gradishar
Nicholson, R. I., Walker, K. J., Bouzukar, N., Wills, R. J., Gee, J. M., Rushmere, N. K. and Davies, P. (1990). Estrogen deprivation in breast cancer. Clinical, experimental and biological aspects. Annals of tke New York Academy of Science 595, 316-327. Nolvadex Adjuvant Trial Organization [NATO] (1983). Controlled trial of tamoxifen as a single adjuvant agent in the management of early breast cancer. Interim analysis at 4 years. Lancet i, 257-261. Nolvadex Adjuvant Trial Organization [NATO] (1988). Controlled trial of tamoxifen as a single adjuvant agent in the management of early breast cancer. Interim analysis at eight years. British Journal of Cancer 57, 608-611. Nomura, Y., Tominaga, T., Abe, O., Izuo, M. and Ogawa, N. (1993). Clinical evaluation of NK 22 (toremifene citrate) in advanced or recurrent breast cancer-a comparative study by a double blind method with tamoxifen [in Japanse]. Gun to Kbgaku Ryoho 20, 247-258. Osborne, C. K. (in press). Journal of Medicine.
Tamoxifen
in the treatment
of breast
cancer.
New England
Osborne, C. K., Coronado, E. B. and Robinson, J. P. (1987). Human breast cancer in athymic nude mice: cytostatic effects of long-term antiestrogenic activity. European Journal of Cuncer 23, 1189-l 196. Osborne, C. K., Coronado-Heinsohn, E. B., Hilsenbeck, S. G., McCue, B. L., Wakeling, A. E., McClelland, R. A., Manning, D. L. and Nicholson, R. I. (1995). Comparison of the effects of a pure steroidal antiestrogen with those of tamoxifen in a model of human cancer. Journal of the National Cancer Institute 87, 746-750. Osborne, C. K., Hobbs, K. and Clark, G. M. (1985). Effects of estrogens and antiestrogens on growth of human breast cancer cells in athymic nude mice. Cancer Research 45, 584-590. Osborne, C. K., Jarman, M., McCague, R., Coronado, E. B., Hilsenbeck, S. G. and Wakeling, A. E. (1994). The importance of tamoxifen metabolism in tamoxifenstimulated breast tumor growth. Cancer Chemotherapy and Phamzacology 34, 89-95. Osborne, M. R., Hewer, A., Hardcastle, I. R., Carmichael, P. L. and Phillips, (1996). Identification of the major tamoxifen-deoxyguanosine adduct formed liver DNA of rats treated with tamoxifen. Cancer Research 56, 66-71.
D. H. in the
Palopoli, F. P., Feil, V. J., Allen, R. F., Holtkamp, D. E. and Richardson, A. Jr. (1967). Substituted aminoalkoxytriarylhaloethylenes. Journal of Medicine and Chemisty 10, 84-86. Perry, J. J., Berry, D. A., Weiss, R. B., Hayes, D. M., Duggan, D. B. and Henderson, I. C. (1995). High dose toremifene for estrogen and progesterone receptor negative metastatic breast cancer. A phase II trial of the Cancer and Leukemia Group B (CALGB). Breast Cancer Research and Treatment 36, 35-40. Pham, T. A., Elliston, J. F., Nawaz, Z., McDonnell, D. P., Tsai, M. J. and O’Malley, B. W. (1991). Antiestrogens can establish nonproductive receptor complexes and alter
241
Molecular Mechanisms and Future Uses of Antiestrogens ch[omatin structure
Science
at target enhancers.
Proceedings
of the National
Academy
of
USA 88, 3125-3129.
Phillips,, D. H., Carmichael, P. L., Hewer, A., Cole, K. J., Hardcastle, I. R., Poon, G. K. and Deogh, A. (1996a). Activation of tamoxifen and its metabolite cc-hydroxytamoxifen to DNA-binding products: comparison between human, rat and mouse hepatocytes. Carcinogenesis 17, 89-94. Phillips., D. H., Hewer, A., White, I. N. H. and Farmer, P. B. (1994a). Co-chromatography of a tamoxifen epoxide-deoxyguanylic acid adduct with a major DNA adduct formed in the livers of tamoxifen-treated rats. Carcinogenesis 15, 793-795. Phillips, D. H., Hewer, A., Grover, P. L., Poon, G. K. and Carmichael, P. L. (1996b). Tamoxifen does not form detectable DNA adducts in white blood cells of breast cancer patients. Carcinogenesis 17, 1149-l 152. Phillips, D. H., Potter, G. A., Horton, M. N., Hewer, A., Crofton-Sleigh, C., Jarman, M. and Venitt, S. (1994b). Reduced genotoxicity of [D,-ethyl]-tamoxifen inplicates cc-hydroxylation of the ethyl group as a major pathway of tamoxifen activation to a liver carcinogen. Carcinogenesis 15, 1487-1492. Phillips, D. H.,- Carmichael, P. L., Hewer, A., Cole, K. J. and Poon, G. K. (1994~). cc-‘hydroxytamoxifen, a metabolite of tamoxifen with exceptionally high DNA-binding activity in rat hepatocytes. Cancer Research. 54, 5518-5522. Pink, J. J. and Jordan, and antiestrogens
V. C. (1996). Models of estrogen receptor regulation by estrogens in breast cancer cell lines. Cancer Research 56, 2321-2330.
Pink, J. J., Bilimoria, M. M., Assikis, V. J. and Jordan, V. C. (1996b). Irreversible loss of the estrogen receptor in T47D breast cancer cells following prolonged estrogen deprivation. British Journal of Cancer (in press). Pink, J. J., Fritsch, M., Bilimoria, M. M., Assikis, V. J. and Jordan, V. C. (in press). Cloning and Characterization of a 77 kilodalton oestrogen receptor isolated from a human breast cancer cell line. British Journal of Cancer. Pink, J. J., Jiang, S. Y., Fritsch, M. and Jordan, V. C. (1995). An estrogen independent MCF-7 breast cancer cell line which contains a novel 80 kilodalton estrogen receptor related protein. Cancer Research 55, 2583-2590. Pink, J. J., Wu, S. Q., Wolf, D. M., Bilimoria, M. M. and Jordan, V. C. (1996a). A novel 80 kilodalton human estrogen receptor containing a duplication of exons 6 and 7. Nucleic Acids Research 24, 962-969. Poulin, :R., Merand, Y., Poirier, D., Levesque, C., DuFour, J. M. and Labrie, F. (1989). Antiestrogenic properties of keoxifene, trans 4-hydroxytamoxifen and ICI 164,384, a new steroidal antiestrogen in ZR-75-1 human breast cancer cells. Breast Cancer Research and Treatment 14. 655-667. Pongracz, K., Pathak, D. N., Nakamura, T., Burgingame, A. L. and Bodell, W. J. (1995). Activation of the tamoxifen derivative metabolite E to form DNA adducts: comparison with the adducts formed by microsomal activation of. tamoxifen. Cancel Research 55, 3012-3015.
242
Potter,
V. C. Jordan and W. J. Gradishar
G. A., McCague, R. and Jarman, M. (1994). A mechanistic hypothesis for DNA adduct formation by tamoxifen following oxidative metabolism. Curcinogenesis 15, 439-442.
Prentice, R. L. (1991). Epidemiologic data on exogenous hormones and hepatocellular carcinoma and selected other cancers. Preventive Medicine 20. 38-46. Pyrhonen, S., Valavaara, R., Vuorinen, J. and Hajba, A. (1994). High dose toremifene in advanced breast cancer resistant to or relapsed during tamoxifen treatment. Breast Cancer Research and Treatment 29. 223-228. Randerath, K., Moorthy, B., Mabon, N. and Sriram, P. (1994). Tamoxifen: evidence by 32P-postlabeling and use of metabolic inhibitors for two distinct pathways leading to mouse hepatic DNA adduct formation and identification of 4-hydroxytamoxifen as a proximate metabolite. Carcinogenesis 15, 2087-2094. Raushning, W. and Pritchard, K. I. (1994). Droloxifene, a new antiestrogen: its role in metastatic breast cancer. Breast Cancer Research and Treatment 31, 83-94. Reddel, R. R. and Sutherland, R. L. (1984). Tamoxifen stimulation cancer cell proliferation in vitro: a possible model for tamoxifen pean Journal of Cancer 20, 1419-1425.
of human breast tumor flare. Euro-
Robinson, S. P. and Jordan, V. C. (1989). Antiestrogen action of toremifene on hormonedependent, independent and heterogeneous breast tumor growth in the athymic mouse. Cancer Research 49, 1758- 1762. Robinson, S. P., Goldstein, D., Witt, P. L., Borden, E. C. and Jordan, V. C. (1990). Inhibition of hormone dependent and independent breast cancer cell growth in vivo and in vitro with the antiestrogen toremifene and recombinant human interferon 012. Breast Cancer Research and Treatment 15,95-101. Robinson, S. P., Langan-Fahey, S. M., Johnson, D. A. and Jordan, V. C. (1991). Metabolites, pharmacodynamics and pharmacokinetics of tamoxifen in rats and mice compared to the breast cancer patient. Drug Met. Disp. 19, 36-43. Robinson, S. P., Mauel, D. A. and Jordan, V. C. (1988). Antitumor actions in the 7, 12 dimethylbenzanthracene (DMBA)-induced rat mammary European Journal of Cancer 24, 18 17- 182 1. Robson, J. M., Schonberg, A. and Fahim, H. A. (1938). Duration synthetic estrogens. Nature 142, 292-293.
of toremifene tumor model.
of action of natural
and
Roos, W. K., Oeze, L., Loser, R. and Eppenberger, U. (1983). Antiestrogen action of 3-hydroxy-tamoxifen in the human breast cancer cell line MCF-7f Journal of the National Cancer Institute 71, 55-59. Rose, D. P., Fischer, A. H. and Jordan, V. C. (1981). Activity of the antiestrogen trioxifene against N-nitrosomethylurea-induced rat mammary carcinomas. European Journal of Cancer and Clinical Oncology 17, 893-898. Rutqvist, L. E., Johansson, N. (1995). Adjuvant primary malignancies.
H., Signomklao, T., Johansson, U., Fornander, T. and Wilking, tamoxifen therapy for early stage breast cancer and second Journal of the National Cancer Institute 87, 645-651.
Molecular
Mechanisms
and Future Uses of Antiestrogens
243
Rutqvist, L. E. and Matteson, A. (1993). Cardiac and thromboembolic morbidity among postmenopausal women with early-stage breast cancer in a randomized trial of tamoxifen. The Stockholm Breast Cancer Study Group. ~oumal of the National Cancer Institute 85, 1398- 1406. Sargent, L. M., Dragan, Y. P., Suttler, C., Bahnub, N., Martin, P., Cisneros, A., Mann, J., Thorgursson, S., Jordan, V. C. and Pitot, H. C. (1996). Induction of hepatic aneuploidy in viva by tamoxifen, toremifene and idoxifene in female Sprague-Dawley rats. Carcinogenesis 17, 10.51-1056. Sabbah, M., Grouilleux, F., Sola, B., Redeuil, G. R. and Baulieu, E. E. (1991). Structural differences between the hormone and antihormone estrogen receptor complexes bound to the hormone response elements. Proceedings of the National Academy of Science USA 88. 390-394. Satyaswaroop, P. G., Zaino, R. J. and Mortel, R. (1984). Estrogen-like effects of tamoxifen on human endometrial carcinoma transplanted into nude mice. Cancer Research 44, 4006-4010. Scholl, IM., Huff, K. K. and Lippman, M. E. (1983). Antiestrogenic MCF-7 cells. Endocrinology 113, 611-617. Smith,
10. W., Smith, G. V. and Kistner, R. W. (1963). Action phene on the human ovary. Journal of the American 122-130.
effects of LYl17018
in
of MER-25 and of clomiMedical Association 184,
Sato, M., Kim, J., Short, L. L., Szemenda, C. W. and Bryant, H. U. (1995). Longitudinal and cross-sectional analysis of raloxifene effects on tibiae from ovariectomized rats. Journal of Pharmacology and Experimental Therapy 272, 1251-1259. Steinberg, D., Avigan, J. and Feigelson, E. G. (1961). Effects of triparanol (MER-29) on cholesterol biosynthesis and on blood sterol levels in man. Journal of Clinical Investigations 40, 884-888. Stenbygaard, L. E., Herrstedt, J., Thomsen, J. F., Svendsen, K. R., Engelholm, S. A. and Dombernowsky, P. (1993). Toremifene and tamoxifen in advanced breast cancer-a double-blind cross-over trial. Breast Cancer Research and Treatment 25, 57-63. Stewart., H.J., Forrest, A.P., Everington, D., McDonald, C.C., Dewar, J.A., Hawkins, R.A., Prescott, R.J.and George, W.D. (1996). on behalf of the Scottish Cancer Trials Breast Group, Randomized comparison of 5 years of adjuvant tamoxifen with continuous therapy for operable breast cancer. British ‘Journal of Cancer 74, 297-299. Styles, J. A., Davies, A., Lim, C. K., De Matteis, F., Stanley, L. A., White, I. N., Yuan, 2. X. and Smith, L. L. (1994). Genotoxicity of tamoxifen, tamoxifen epoxide and toremifene in human lymphoblastoid cells containing human cytochrome P45O’s. Carcinogenesis 15, 5-9. Szamel, I., Hindy, I., Vincze, B., Eckardt, S., Kangas, L. and Hajba, A. (1994). Influence of toremifene on the endocrine regulation in breast cancer patients. European Journal of Cancer 3OA, 154- 158.
244
V. C. Jordan and W. J. Gradishar
Tang,
M. X., Jacobs, P., Stern, Y., Marder, K., Schofield, P., Garland, B., Andrews, H. and Mayeux, R. (1996). Effect of oestrogen during menopause on risk and age at onset of Alzheimers disease. Lancet 348, 429-433.
Tate,
A. C., Greene, G. L., DeSombre, E. R., Jensen, E. V. and Jordan, V. C. (1984). Differences between estrogen and antiestrogen-estrogen receptor complexes identified with an antibody raised against the estrogen receptor. Cancer Research 44, 1012-1018.
Thompson, E. W., Katz, D., Shima, T. B., Wakeling, A. E., Lippman, M. E. and Dickson, R. B. (1989). ICI 164,384, a pure antagonist of estrogen stimulated MCF-7 cell proliferation and invasiveness. Cancer Research 49, 6929-6934. Toko, T., Maksuo, K. I., Shibata, J., Wierzba, K., Nukatsuka, M., Takeda, S., Yamada, Y., Asao, T., Hirose, T. and Sato, B. (1992). Interaction of DP-TAT-59 an active metabolite of new triphenylethylene derivative (TAT-59) with estrogen receptors. Journal of Steroid Biochemisty and Molecular Biology 43, 507-514. Toko, T., Sigimoto, Y., Matsuo, K. I., Yamasaki, R., Takeda, S., Wierzba, K., Asao, Y. and Yamada, Y. (1990). TAT-59 a new triphenylethylene derivative with antitumor activity against hormone dependent tumors. European Journal of Cancer 26, 397-404. Tomas, E., Kauppila, A., Blanco, G., Apaja-Sarkkinen, M. and Laatikainen, T. (1995). Comparison between the effects of tamoxifen and toremifene on the uterus in postmenopausal breast cancer patients. Gynecology and Oncology 59,261-266. Tominga, T., Abe, O., Izuo, M. and Nomurga, U. T. (1990). A phase emifene. Breast Cancer Research and Treatment 16,27-27.
I study of tor-
Tonetti, D. A. and Jordan, V. C. (1995). Possible mechanisms in the tamoxifen-resistant breast cancer. Anticancer Drugs 9,498-507. Tonetti, D. A. and Jordan, V. C. (1996). The development of targeted prevent diseases in women. Molecular Medicine Today 2, 218-223.
emergence
of
antiestrogens
to
Tormey, D. C., Gray, R., Abeloff, M. D., Roseman, D. L., Gilchrist, K. W., Barylak, E. J., Stott, D. and Falkson, G. (1992). Adjuvant therapy with a doxarubicin regimen and long term tamoxifen in premenopausal breast cancer patients: A Eastern Cooperative Oncology Group Trial. Journal of Clinical Oncology 10, 1848-1855. Turken, S., Siris, E., Selding, E., Flaster, E., Hyman, G. and Lindsay, R. (1989). Effect of tamoxifen on spinal bone density in women with breast cancer. Journal of the National Cancer Institute 81, 1086-1088. Turner, R. T., Wakley, G. K., Hannon, K. S. and Bell, N. H. (1987). Tamoxifen prevents the skeletal effects of ovarian hormone deficiency in rats. Journal of Bone Mineral Research 2, 449-456. Turner, R. T., Wakley, G. K., Hannon, K. S. and Bell, N. H. (1988). Tamoxifen inhibits osteoclast mediated resorption of trabecular bone in ovarian hormone-deficient rats. Endocrinology 122, 1146- 1150.
Molecular Mechanisms and Future Uses of Antiestrogens Tyler, E. T., Olson, H. J. and Gotlieb, M. H. (1960). The induction antiestrogen. International Journal of Fertility 5, 429-431.
245 of ovulation
with an
Tzukerman, M. T., Esty, A., Santiso-Mere, D., Danelian, D., Parker, M. G., Stein, R. B., Pike, J. W. and McDonnell, D. P. (1994). Human estrogen receptor transactivational capacity is determined by both cellular and promoter context and mediated by two functionally distinct intramolecular regions. Molecular Endocrinology 8, 21-30. Valavaara, R. (1990). Phase II experience with toremifene in the treatment of ER-positive breast cancer of postmenopausal women. Cancer Investigations 8,275-276. Valavaara, R. and Pyrhonen, S. (1989). L ow-dose toremifene in the treatment of estrogenrec:eptor-positive advanced breast cancer in postmenopausal women. Current Therapy and Research 46, 966-973. Valavaara, R., Pyrhonen, S., Heikkinen, M., Rissanen, P., Blanco, G., Tholix, E., Nordman, E., Taskinen, D., Holsti, L. and Hajba, A. (1988). Toremifene, a new antiestrogenic treatment of advanced breast cancer Phase II study. European Journal of Cancer 24, 785-790. Van de Velde, P., Nique, F., Bouchoux, F., Bremaud, J., Hameau, M. C., Lucas, D., Moratille, C., Viet, S., Philibert, D. and Teutsch, G. (1994). RU 58,668 a new pure antiestrogen inducing a regression of human mammary carcinoma implanted in nude mice. Journal of Steroid Biochemistry and Molecular Biology 48, 187- 196. van Leeuwen, F. E., Benraadt, J., Coebergh, J. W. W., Kiemeney, L. A., Gimbrere, Otter, R., Shouten, L. J., Damriuis, R. A., Bontenbal, M. and Diedenhorst, (1994). Risk of endometrial cancer after tamoxifen treatment of breast Lancet 343,448-452.
C. H., F. W. cancer.
L., Hayes, D. F. and Gams, R. A. (1995). Vogel, C. L., Schoenfelder, J., Shermano, Wclrsening bone scan in the evaluation of antitumor response during hormonal therapy for breast cancer. Journal of Clinical Oncology 13, 1123-1328. Vogel, C. L., Shemano, I., Schoenfelder, J., Gams, R. A. and Green, M. R. (1993). Multicenter phase II efficacy trial of toremifene in tamoxifen refractory patients with advanced breast cancer. youma of Clinical Oncology 11, 345-350. von Angerer, E., Krebel, N., Kager, indoles as novel pure estrogen 2636-2640.
M. and Ganss, R. (1990). 1-(Aminoalkyl) Z-phenylantagonists. ‘Journal of Medicine and Chemisty 33,
von Sallman, L., Grimes, P. and Collins, E. (1963). Triparanol-induced Archives Ophthamology 70, 1128- 1136.
cataracts
in rats.
Wakeling, A. E. (1994). A new approach to breast cancer therapy-total estrogen ablation with pure antiestrogens. In: Long-tern2 Tamoxifen Treatment for Breast Cancer. (Jordan, V. C., ed.), pp. 219-234. University of Wisconsin Press, Madison. Wakeling, A. E. and Bowler, J. (1987). Steroidal ogy 112, R7-RlO.
pure antiestrogens.
Wakeling, A. E. and Bowler, J. (1988). Novel antiestrogens Journal of Steroid Biochemisty 31, 645-653.
without
Journal of Endocrinol-
partial agonist
activity.
V. C. Jordan and W. J. Gradishar
246
Wakeling, A. E. and Valcaccia, B. (1983). Antioestrogenic and antitumour activities series of non-steroidal antioestrogens. Journal of Endocrinology 99, 45.5-464.
of a
Wakeling, A. E., Dukes, M. and Bowler, J. (1991). A potent clinical potential. Cancer Research 51, 3867-3873.
with
Walpole, A. L. and Paterson, ii, 783-789.
E. (1949). Synthetic
oestrogens
specific pure antiestrogen
in mammary
Ward, H. W. C. (1973). Antioestrogen therapy for breast cancer: dose levels. British MedicalJournal i, 13-14.
cancer. Lancet
a trial of tamoxifen
at two
Ward, R. L., Morgan, G., Dalley, D. and Kelly, P. J. (1993). Tamoxifen reduces bone turnover and prevents lumbar spine and proximal femoral bone loss in early postmenopausal women. Bone Mineralogy 22, 87-94. Warri, A. M., Huovinon, R. L., Laine, A. M., Mairtikainen, P. M. and Harkonen, P. L. (1993). Apoptosis in toremifene growth inhibition of human breast cancer cells in vivo and in vitro. Journal of the National Cancer Institute 85, 1412-1418. Webster, L. K., Crinis, N. A., Stokes, K. H. and Bishop, J. F. (1991). High performance liquid chromatographic method for the determination of toremifene and its major human metabolites. J. Chromatogr. B. Biomed. Appl. 565, 482-487. Welshons, W. V. and Jordan, V. C. (1987). Adaption of estrogen-dependent MCF-7 cells to low estrogen (phenol red-free) culture. European Journal of Cancer 23, 1935-1939. White,
I. N. H., deMatteis, F., Davies, A., Smith, L. L., Crofton-Sleigh, C., Venitt, S., Hewer, A. and Philips, D. H. (1992). Genotoxic potential of tamoxifen and analogues in female Fischer 344/n rats, DBA/2 and C57BL/6 mice and in human MCL-5 cells. Carcinogenesis 13, 2197-2203.
Wiebe,
V. J., Benz, C., Shemano, I., Cadman, T. B. and DeGregorio, M. W. (1990). Pharmacokinetics of toremifene and its metabolites in patients with advanced breast cancer. Cancer Chemotherapy and Pharmacology 25, 247-25 1.
Williams, G. M., Iatropoulos, M. J., Djordjevic, M. V. and Kaltenberg, 0. P. (1993). The triphenylethylene drug tamoxifen is a strong liver carcinogen in the rat. Carcinogenesis 14, 315-317. Williamson, J. G. and Ellis, J. P. (1973). The induction of ovulation by tamoxifen. of Obstetrics and Gynaecology in the British Commonwealth 80, 844-847.
Journal
Winterfeld, G., Hauff, P., Grolich, M., Arnold, W., Fichtner, I. and Staab, H. J. (1992). Investigations of droloxifene and other hormonal manipulations on N-nitrosomethylurea-induced rat mammary tumours. Journal of Cancer Research and Clinical Oncology 119,91-96. Wiseman, H., Cannon, M., Astein, H. R. V. and Halliwell, B. (1993a). Tamoxifen inhibits lipid peroxidation in cardiac microsomes. Comparison with liver microsomes and potential relevance to the cardiovascular benefits associated with cancer prevention and treatment by tamoxifen. Biochemical Pharmacology 45, 1851-1855.
Molecular Mechanisms and Future Uses of Antiestrogens
247
Wiseman, H., Paganga, G., Rice-Evans, C. and Halliwell, B. (1993b). Protective actions of tamoxifen and 4-hydroxytamoxifen against oxidative damage to human low-density lipoproteins: a mechanism accounting for the cardioprotective action of tamoxifen? Biochemical Journal 292, 635-638. Witte, R. S., Pruitt, B., Tormey, D. C., Moss, S., Rose, D. P., Falkson, G., Carbone, P. P., Ramirez, G., Falkson, H. and Pretorius, F. J. (1986). A phase I/II investigation of trioxifene mesylate in advanced breast cancer. Clinical and endocrinological effects. Cancer 57, 34-39. Wolf, D. M., Langan-Fahey, S. M., Parker, C. J., McCague, R. and Jordan, V. C. (1993). Investigation on the mechanism of tamoxifen-stimulated breast tumor growth using nonisomerizable analogs of tamoxifen and metabolites. Journal of the National Cancer Institute 85, 806-812. Wosikowski, K., Kung, W., Hasmann, M., Loser, R. and Eppenberger, U. (1993). Inhibition of growth factor activated proliferation by antiestrogens and effects on early gene expression of MCF-7 cells. International Journal of Cancer 53,290-297. Yang, N. N., Venugopalan, M., Hardikar, S. and Glasebrook, A. (1996). Identification of an estrogen response element activated by metabolites of 17fl-estradiol and raloxifene. Science 273. 1222-1225.