<<

[CANCER RESEARCH 58. 2269-2277. June I. 1998] Perspectives in Cancer Research

Is 2-Methoxyestradiol an Endogenous Metabolite That Inhibits Mammary Carcinogenesis?

Bao Ting Zhu1 and Allan H. Conney2

Laboratory for Cancer Research, Department of Chemical Biology, College of Ptiannac\, Rutgers-The Stale University of New Jersey, Piscataway, New Jerse\ 08854-8020

Abstract or can be hydroxylated at various positions by NADPH-dependent present in and (2- or 4-hydroxyestradiol and 2- or 4-hydroxy- extrahepatic target cells (reviewed in Refs. 33 and 34). The chemi estrone) are chemically reactive estrogen metabolites that are O-methyl- cally reactive catechol estrogens (2- or 4-hydroxyestradiol and 2- or ated to less polar monomethyl ethers by catechol-O-methyltransferase, an 4-hydroxyestrone) can be metabolically O-methylated to monomethyl present in many tissues such as the liver, kidney, brain, placenta, ethers (structures shown in Fig. 1) by COMT,3 an enzyme present in uterus, and mammary gland. In the present report, we review recent studies on the antitumorigenic and antiangiogenic effects of exogenously large amounts in many organs and cells such as liver, kidney, brain, administered 2-methoxyestradiol in vitro and in vivo. We also discuss data placenta, uterus, mammary gland, and RBCs (35-42). Because of the that suggest that endogenous formation of 2-methoxyestradiol (and its ubiquitous distribution of COMT activity in mammalian tissues (35- 2-hydroxyestradiol precursor) may have a protective effect on estrogen- 42) and the very rapid metabolic O-methylation of catechol estrogens induced cancers in target organs. Although the molecular mechanism of as demonstrated both in vitro and in vivo (42-46), large amounts of action of 2-methoxyestradiol is not clear, we suggest that some unique monomethylated estrogen metabolites are produced. A previous study effects of 2-methoxyestradiol may be mediated by a specific intracellular reported that blood concentrations of unconjugated and conjugated effector or receptor that is refractory to the parent hormone, estradiol. 2-methoxyestrone are approximately 4000 and 6000 pg/ml, respec Additional research is needed to identify factors that regulate the meta tively, in pregnant women (47,48). which are over 50-fold higher than bolic formation and disposition of 2-methoxyestradiol in liver and in the concentration of estradiol. Interestingly, 2-methoxyestradiol and target cells and to evaluate the effects of modulating 2-methoxyestradiol formation on estrogen-induced carcinogenesis. 2-methoxyestrone have higher binding affinities for the - binding globulin in blood than does estradiol (48), which may be an important factor (in addition to rapid metabolic formation of me- Introduction thoxyestrogens) that contributes to their high circulating levels. Chronic administration of estrogens (such as estradiol and estrone) Most studies showed that methylated catechol estrogens have little results in tumor formation in target organs in several animal models or no binding affinities for the classical (<0.1% (1-7). Estrogens have also been increasingly implicated as an impor compared with estradiol). and they lack estrogenic activity in the tant etiological factor in the causation of certain types of human uterus (43, 49, 50). An important question is why animals and humans cancer (8-16). Chronic administration of estrogens to postmeno- make large amount of hormonally inactive yet highly lipophilic es pausal women is strongly associated with an increased risk of endo- trogen metabolites that probably have much longer half-lives than metrial cancer (8-12). Although the relationship between estrogen estradiol? Studies on the chemical reactivity and potential genotoxic- and breast cancer is not as strong as that between estrogen and ity of catechol estrogens (23, 25-27, 29-32) have led to the sugges endometrial cancer, there is mounting evidence associating elevated tion that metabolic O-methylation may be a rapid inactivation/detox- breast cancer risk with an increased total life-time exposure to estro ification pathway for these estrogen-derived , as is known for gens (13-16). Epidemiological studies indicate an increased risk of . However, there are several studies indicating that breast cancer in women with: (a) high serum levels of estrogens 2-methoxyestradiol may not be just an inactive metabolite of estradiol (comparison of women in the United States with those in Japan and but may have unique biological activities that are not shared with China); (b) prolonged exposure to estrogens (due to early menarche estradiol, 2-hydroxyestradiol, or 4-methoxyestradiol (51-57). and late menopause or long-term use of estrogen supplements); and In this report, we review data that point to the hypothesis that (c) obesity (a cause of increased formation of estrogen by enzymatic endogenously formed 2-methoxyestradiol may have a unique protec aromatization of in adipose cells) (reviewed in Refs. 15 and tive role against estrogen-induced carcinogenesis in estrogen target 16). Although the mechanism of estrogen-induced carcinogenesis is organs. We also propose that some unique actions of 2-methoxyestra still not clear, it is thought that the superpotent mitogenic action of diol may be mediated by a specific but unidentified intracellular estrogenic hormones (5, 14, 17-21), together with the potential geno- receptor or effector that is refractory to the parent hormone, estradiol. toxicity of some reactive estrogen metabolites (e.g.. 4- and 16a- Inhibitory Effect of 2-Methoxyestradiol on the Growth of hydroxylated estrogens; see Refs. 6 and 22-32), may play important Tumor Cells in Vitro and in Vivo roles in the induction of estrogen-associated cancers in target organs of animals and humans. 2-Methoxyestradiol inhibits the proliferation of many human can cer cell lines in vitro (54, 55, 57-61). Human breast cancer cell lines

Received 1/16/98; accepted 3/26/98. were particularly sensitive to the cytotoxic effect of 2-methoxyestra The costs of publication of this article were defrayed in pan by the payment of page diol and several synthetic analogues irrespective of the estrogen charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. receptor status of the cell lines studied (58). An earlier study showed 1To whom requests for reprints should be addressed, at Rutgers University, College of that the inhibitory effect of 2-methoxyestradiol (at a 10 nM concen- Pharmacy. Laboratory for Cancer Research, Department of Chemical Biology, 164 Frelinghuysen Road. Piscataway, NJ 08854-8020. Fax: (732) 445-0687; E-mail: baoting® 3 The abbreviations used are: COMT. calechol-O-methyllransferase; !7ß-HSD. 17fJ- rci.rutgers.edu. 2 William M. and Myrle W. Garbe Professor of Cancer and Leukemia Research. hydroxysteroid dehydrogenase. 2269

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research. 2-METHOXYESTRADIOL AS AN INHIBITOR OF BREAST CANCER

Fig. 1. The structures of monomethylated cat- echo] estradiol metabolites. Estradici is hydroxy- lated to catechols (2- and 4-hydroxyestradiol| largely by cytochrome P450 enzymes. These cat echols can be rapidly O-melhylated by COMT to form monomethoxy estradiol metabolites. 2-Me- thoxyestradiol and 4-methoxyestradiol are the H,C major monomethylated isomers formed from their respective catechol precursors. Although the structures for estrone and its catechol metab 2-OH-3-MeO-E2 olites are not shown, estrone can also be metab Catecholamines, olized to catechols and then to 0-melhylated , etc. catechols. Because COMT catalyzes the metab olism of a wide spectrum of endogenous and exogenous catechols. the enzymatic O-methyla- 17ß-Estradiol(E2) tion of estradiol or estrone catechols can be reg ulated by these non-estrogen catechols (e.g., CH30 hydroxylated . catecholamines, and COMT \ 4-MeO-E2 others). P450. cytochrome P450; 2-OH-E-,. 2-hy- droxyestradiol; 4-OH-E2. 4-hydroxyestradiol; 2-MeO-E2. 2-melhoxyestradiol; 2-OH-J-MeO- \ E2, 2-hydroxyestradiol ,1-methyl ether; 4-MeO- £2,4-methoxyestradiol; 4-OH-3-MeO-E2, 4-hy CH3O droxyestradiol 3-methyl ether. OH 4-OH-3-MeO-E2

tration) on the proliferation of MCF-7 human breast cancer cells was modulate estrogen-induced tumorigenesis in the breast and possibly not shared by estradiol or 2-hydroxyestradiol (55). At considerably other target organs. higher concentrations (>1 piM), 2-methoxyestradiol was highly cyto- Induction of kidney tumors in male Syrian hamsters by chronic toxic to several human cancer cell lines in culture, including HeLa treatment with estradiol or other natural or synthetic estrogens is a cells (a cell line derived from human cervical cancer). MCF-7 breast frequently used animal model for studying the mechanisms of estro cancer cells, Jurkat leukemia cells, and retinoic acid-differentiated gen-induced cancer (2, 6, 63). Recent studies showed that chronic SH-SY5Y neuroblastoma cells (54, 57-60). 2-Methoxyestradiol ap administration of quercetin. a commonly ingested dietary polyphenol pears to target actively dividing cells by disturbing cellular events that is a substrate and also a potent inhibitor of COMT (64, 65), associated with cell proliferation (54, 56). significantly increased the severity of estradiol-induced kidney tumor It is of considerable interest to note that oral administration of formation in male Syrian hamsters (64). Additional studies showed 2-methoxyestradiol (75 mg/kg body weight/day) inhibited the growth that the formation of 2- and 4-methoxyestradiol in vivo is inhibited by of transplanted Meth-A sarcoma and B16 melanoma cells in C3H chronic administration of quercetin (65). This inhibition of the met mice (57), and this treatment also inhibited the growth of a human abolic O-methylation of catechol estrogens is due to a combination of breast carcinoma cell line (MDA-MB-435, estrogen receptor nega three mechanisms (65): (a) a direct of COMT tive) in immunodeficient mice (61). by quercetin (which is a catechol and an excellent substrate for Inhibitory Effect of 2-Methoxyestradiol on Angiogenesis in Vitro COMT); (b) a noncompetitive inhibition due to increased levels of S-adenosyl-L-homocysteine in the target organ; and (c) a reduced and in Vivo availability in the target organ of S-adenosyl-i.-methionine, the sup 2-Methoxyestradiol has a potent inhibitory effect on the prolifera porting co-substrate for the enzymatic O-methylation of catechol tion of blood-vessel endothelial cells in vitro (ICJO, —¿100nM;Ref. estrogens. An inhibition of the COMT-catalyzed O-methylation of 57). Additional studies showed that 2-methoxyestradiol induces ap- catechol estrogens during quercetin administration decreases the pro optosis in cultured arterial endothelial cells in a time- and concentra duction of 2-methoxyestradiol in addition to causing an accumulation tion-dependent manner, and the migration of these vascular endothe of the reactive 2- and 4-hydroxyestradiol intermediates in target lial cells in vitro is also inhibited (62). Administration of this estradiol organs (65). 4-Hydroxyestradiol is a strong renal carcinogen in Syrian metabolite to animals strongly inhibits angiogenesis in vivo (57, 61, hamsters (63, 66), and this compound is believed to contribute to the 62). It is noteworthy that 2-methoxyestradiol is among the most potent carcinogenic action of estradiol (6). It will be of interest to determine endogenous nonprotein inhibitors of in vitro angiogenesis, and its whether other dietary polyphenols that are ingested in large amounts antiangiogenic effect is highly specific and is not shared by several by humans will also enhance estrogen-associated carcinogenesis as closely related structural analogues (57). The strong antiangiogenic well as to determine the relevance of these observations for humans. effect of 2-methoxyestradiol described here as well as its potent It is worth noting that renal carcinogenesis in Syrian hamsters cytostatic effect on the growth of cultured tumor cells (described chronically treated with (a nonsteroidal estrogen) is above) may both contribute to the inhibitory effect of 2-methoxyestra not enhanced by chronic coadministration of quercetin.4 Unlike the diol on tumor growth in animal models (57, 61). facile formation of methylated catechols from estradiol, catechol formation from diethylstilbestrol and the subsequent O-methylation of Decreased 2-Methoxyestradiol Formation Is Associated with an Increased Risk of Estrogen-induced Cancers its catechol intermediates are only very minor metabolic pathways (67, 68). Diethylstilbestrol can directly undergo metabolic redox Because 2-methoxyestradiol inhibits the proliferation of breast can cycling (without conversion to a catechol) to generate free radicals cer cells and angiogenesis in vitro and in vivo (described above), factors that alter the metabolic formation of 2-methoxyestradiol may 4 B. T. Zhu and J. G. Liehr. unpublished data. 2270

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research. 2-MhTHOXYKSTRADIOI. AS AN INHIBITOR OF BREAST CANCER and form reactive quinone/semiquinone intermediates (23, 25, 69), with a normal alíele.This observation is consistent with the proposed which is believed to contribute to diethylstilbestrol-induced carcino- antitumorigenic role of 2-methoxyestradiol and the carcinogenic role genesis. In addition, it was observed that 7.12-dimethylbenz(«|anthra- of 4-hydroxyestradiol in human mammary carcinogenesis. It will be cene-induced mammary tumors in rats (70) and azoxymethanol-in- of interest in future studies to determine the COMT enzyme activity duced colonie neoplasms in mice (71) were not increased (but and levels of 2-methoxyestradiol and 4-hydroxyestradiol in the breast significantly decreased) by chronic cotreatment with quercetin. The of the same subjects and to correlate these parameters with the breast inhibitory effects of quercetin on 2-methoxyestradiol formation in cancer risk in women with a normal versus a variant alíeleof the vitro and in vivo, and its stimulatory effect on estradiol-induced COMT gene. carcinogenesis but not diethylstilbestrol-, 7,l2-dimethylbenz[a)an- thracene-, or azoxymethane-induced carcinogenesis (described above) Increased Formation of 2-Hydroxyestradiol and 2- are consistent with the concept that selective stimulation of estradiol- Methoxyestradiol Is Associated with a Decreased Risk of induced carcinogenesis by quercetin is caused by a reduced formation Estrogen-induced Cancers of the antitumorigenic 2-methoxyestradiol. In accord with the inhibitory effect of dietary catechols such as The data discussed above suggest that decreased formation of quercetin and fisetin on COMT-catalyzed O-methylation of catechol 2-methoxyestradiol is associated with increased formation of estradi ol-induced tumors. There are also data (discussed below) suggesting estrogens (64, 65). endogenous catechols may also exert an inhibitory effect on the O-methylation of catechol estrogens. Recent studies have that increased formation of 2-hydroxyestradiol and 2-hydroxyestrone shown that very high concentrations of endogenous catecholamines (products of estrogen 2-hydroxylation ), which subse (substrates and inhibitors of catechol estrogen O-methylation) are quently leads to elevated formation of 2-methoxyestradiol and 2-me- present in male Syrian hamster kidney. CD-I mouse uterus, and thoxyestrone by COMT, is associated with a decreased risk of estro Fisher-344 rat pituitary (72). three target organs that develop estrogen- gen-associated tumors in target organs. induced tumors (1, 2. 4). The concentrations of catecholamines in The formation of spontaneous mammary tumors in female C3H/ these target tissues are up to 50-fold higher than those in nontarget OuJ mice, spontaneous uterine tumors in Oncomice or in Donryu rats, tissues of these animals or in tissues of other strains or species (72). and chemically induced mammary tumors in Sprague Dawley rats is Because catecholamines inhibit the enzymatic O-methylation of cat dependent on the presence of endogenous estrogens (5, 92-94). Stud echol estrogens in vitro at physiologically relevant concentrations ies during the past several years have shown that chronic treatment of (72), we believe that high tissue concentrations of catecholamines animals with indole-3-carbinol stimulates the 2-hydroxylation of es- may inhibit the metabolic formation of 2-methoxyestradiol in addition tradiol (95-97), and this treatment is associated with decreases in to elevating the concentrations of reactive catechol estrogen interme spontaneous mammary tumorigenesis in C3H/OuJ mice (95, 96), diates in target organs. spontaneous uterine tumorigenesis in Oncomice (98) or Donryu rats Earlier studies in female C3H/He mice showed that increased levels (99), and chemically induced mammary tumorigenesis in Sprague of environmental or manipulative stress markedly increased the inci Dawley rats (100). Recent studies in our laboratory showed that dence of spontaneous mammary cancers (73). Interestingly, epidemi chronic administration of 0.05% sodium phénobarbitalin the drinking ológica!studies in humans suggest a positive relationship between the water to female C3H/OuJ mice for 16 months very strongly inhibited degree of sustained emotional stress and the risk for developing breast the formation of spontaneous mammary tumors, and this inhibitory cancer (74-76). Because studies in animals and humans both indicate effect of phénobarbitalwas accompanied by a manyfold increase in that an increased level of stress is accompanied by an increased liver microsomal 2-hydroxylation of estradiol with little or no increase in estradiol 4- or I6a-hydroxylation.^ release of endogenous catecholamines, we suggest a unifying hypoth esis that sustained stress increases the levels of catecholamines. which Earlier studies in humans showed that cigarette smokers have then inhibit the enzymatic O-methylation of catechol estrogens, re enhanced 2-hydroxylation of estradiol in vivo (101-103) and some sulting in decreased formation of 2-methoxyestradiol and increased what lower serum and urinary levels of estradiol and estrone ( 104- accumulation of reactive catechol intermediates in target tissues. This 106). This stimulatory effect of cigarette smoking on estrogen 2-hy hypothesis is in accord with data indicating an association of sustained droxylation was associated with a decreased risk of uterine emotional or environmental stress with an increased breast cancer endometrial cancer (in addition to an increased incidence of osteopo risk. Decreased immune system function with sustained stress has also rosis) in female cigarette smokers (107-109). been postulated to play a role in the increased breast cancer risk Human cytochrome P450 1A1 as well as other P450 isoforms (e.g.. observed (77, 78). 1A2 and 3A4) possess strong catalytic activity for estrogen 2- Several early studies noted marked person-to-person variations in hydroxylation (33, 34). A recent preliminary study suggested that the COMT activity in RBCs (79-81). Additional studies showed that individuals with an Msp\ polymorphism in the CYPIAI gene had a the distribution of COMT activity in an American population has a low basal level of estrogen 2-hydroxylation. and treatment of these polymorphic bimodal pattern (82-84). Genetic analysis revealed that individuals with indole-3-carbinol failed to induce estradiol 2-hy human COMT is encoded by a single gene (a wild-type and a variant droxylation (110). Research on a possibly increased risk of breast alíele),and approximately one-fourth of the Caucasians studied in cancer in Caucasian women or in African-American women with herited a variant alíeleencoding a low activity form of the enzyme polymorphisms in the CYPIAI gene has resulted in inconsistent (82, 85-90). We anticipate that individuals who inherit the low conclusions (111, 112). activity form of the COMT gene will be at increased risk for estrogen- It was recently reported that African-American women have a associated breast cancer due to decreased formation of the antitumori lower rate of estrogen 2-hydroxylation than American Caucasian genic 2-methoxyestradiol in addition to retarded inactivation of cat women (113. 114). It is not known whether this difference in estrogen echol estrogen intermediates (particularly 4-hydroxyestradiol, which 2-hydroxylation contributes to the higher breast cancer mortality rate is hormonally active and is carcinogenic). A recent study by Lavigne observed in African-American women than in American Caucasian et al. (91) indicated that postmenopausal women with a variant alíele women (115). The relative roles of genetic and environmental factors coding for a low activity form of the COMT have an increased risk (odds ratio, 2.18) for developing breast cancer compared with women 5 Unpublished data. 2271

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research. 2-METHOXYESTRADIOL AS AN INHIBITOR OF BREAST CANCER

Other hydroxylated estrogen metabolites

2-OH-E2 P45O P450 -*- 4-OH-E2,16a-OH-Ej, etc 2-Hydroxylatìon •¿â€¢c I Estrogen receptor-mediated Genotoxicity mitogenic stimulation

2-MeO-E2 Inhibition

Estrogen-induced carcinogenesis

Fig. 2. A diagram depicling the beneficial effect of estradici 2-hydroxylation and subsequent 2-methoxyestradiol formation on estrogen-induced carcinogenesis. The multiple pathways of oxidative estradici metabolism by NADPH-dependent cytochrome P450 enzymes leads to the formation of: Ui) hormonally active and chemically reactive metabolites (e.g., 4-hydroxyestradiol. 16a-hydroxyestronel: (hi 2-hydroxylated metabolites (e.g.. 2-hydroxyestradiol. 2-hydroxyestrone): and (c) other hydroxylated metabolites. Selectively increasing the 2-hydroxylation pathway of estradici metabolism will not only decrease the availability of the parent hormone, but it will also increase the subsequent formation of 2-methoxyestradiol. a nonpolar estradici metabolite that has potent growth-inhibitory and antiangiogenic actions in mammary tumors. P450, cytochrome P450; £,, 170-estradiol: 2-OW-£2.2-hydroxyestradiol; 4-OH-E^. 4-hydroxyestradiol; 16a-OH-E,. I6a-hydroxyestrone; 2-MeO-E,. 2-methoxyestradiol.

in explaining these differences in estrogen metabolism in African- estradiol or estrone (129-133) as well as the subsequent O-methyla American and Caucasian-American women have not been explored. tion of the catechol metabolites (39, 40). It is expected that large Consistent with observations associating high levels of estrogen amounts of 2-methoxyestradiol will be produced in these target tis 2-hydroxylation with a reduced cancer risk in animals and humans, sues. Because cultured human breast cancer cells (regardless of their earlier studies with a commonly used hamster kidney tumor model estrogen receptor status) are particularly sensitive to growth inhibition showed that 2-hydroxyestradiol and 2-hydroxyestrone are devoid of by 2-methoxyestradiol (58). increased formation of 2-methoxyestra carcinogenic activity at dose levels where estradici or 4-hydroxyestra diol by the breast is likely to be an important endogenous factor that diol gives rise to a 100% tumor incidence (63, 66). The lack of suppresses the development of estrogen-associated mammary cancers. carcinogenic activity of 2-hydroxylated estrogen metabolites is not There is a need to enhance our understanding of person-to-person completely understood. Although earlier studies suggested that 2- and differences in the formation of 2-methoxyestradiol and of factors that 4-hydroxyestradiol have similar chemical reactivity and genotoxicity influence the formation of 2-methoxyestradiol in normal human breast when tested in vitro (23, 25, 26), 2-hydroxyestradiol appears to have and in breast cancers. Selectively enhancing the endogenous forma a faster rate of metabolism by COMT-catalyzed O-methylation (44. tion of 2-methoxyestradiol in target cells may represent a new ap 72). a more rapid metabolic clearance in vivo (116, 117), and a weak proach for the prevention and treatment of mammary cancers. er hormonal activity in estrogen target organs (118-126) than 4- hydroxyestradiol. Moreover, the COMT-catalyzed O-methylation of Potential Physiological Functions of 2-Methoxyestrone 4-hydroxyestradiol is inhibited by 2-hydroxyestradiol, whereas the O-methylation of 2-hydroxyestradiol is not inhibited by 4-hy Although very high concentrations of 2-methoxyestrone are present droxyestradiol (44). It is likely that 4-hydroxyestradiol may accumu in the human circulation, this estrone metabolite only has about 1% of late in target cells because of inhibition of its O-methylation. Finally, the growth-inhibitory potency of 2-methoxyestradiol in cultured the presence of increased amounts of 2-hydroxylated estrogens will blood-vessel endothelial cells or fibroblasts (57). Because 17ß-HSDis subsequently lead to increased formation of 2-methoxyestradiol, and a group of intracellular isozymes catalyzing interconversions between the formation of this cytostatic and antiangiogenic metabolite may be estradiol and estrone (134, 135), it is expected that this enzyme an important reason for the lack of carcinogenicity of 2-hydroxylated will also catalyze interconversions between 2-methoxyestradiol and estrogens in animals and for the reduced cancer risk in animals and 2-methoxyestrone (Fig. 3). Accordingly, it is likely that 2-me humans associated with high levels of estrogen 2-hydroxylation. This thoxyestrone is an immediate metabolic precursor of 2-methoxyestra concept is illustrated in Fig. 2. diol. High levels of estradiol are present in estrogen target tissues, and 17ß-HSDactivity has a cell-specific localization (such as in the cancerous breast tissue contains particularly high levels of estradiol secretory glandular epithelium and proliferative endometrium of the and estrone (127, 128). Many breast cancers also contain high levels uterus: Ref. 136), and this enzyme activity is regulated by certain of enzymes that catalyze the formation of catechol estrogens from hormones during different stages of the normal menstrual cycle (137). 2272

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research. 2-METHOXYESTRADIOL AS AN INHIBITOR OF BREAST CANCER

17ß-HSD(oxidativeactivity) Fig. 3. Metabolic interconversions between estradici and estrone (or between 2-methoxyestradiol and 2-me- 17ß-HSD(reductiveactivity) thoxyestrone) catalyzed by 17ß-HSD.The 17j3-oxidative ac HO HO Estradici tivity of 17j3-HSD will convert estradici to estrone, whereas Estrone the 17/3-reductive activity of 17ß-HSDwill convert estrone to estradici. Although the 17ß-HSD-cataly/.ed interconversions HjC between estradici and estrone have been studied extensively, the postulated interconversions between 2-methoxyestradiol and 2-methoxyestrone have not been studied. CH3O 17ß-HSD(oxidativeactivity) 17ß-HSD(reductiveactivity) HO 2-Methoxyestradiol 2-Methoxyestrone

The uterine 17ß-HSD(type II) predominantly catalyzes the 17/3- active 16a-hydroxyestrone and loa-hydroxyestradiol) are carcino oxidative conversion of estradiol to estrone (anticipated conversion of genic, whereas the products of the 2-hydroxylation pathway (hormon 2-methoxyestradiol to 2-methoxyestrone; Refs. 134 and 138), whereas ally weak 2-hydroxyestrone and 2-hydroxyestradiol) are not carcino the 17ß-HSD(type I) present in breast tumors predominantly cata genic. According to this hypothesis, the beneficial effect of increased lyzes the 17ß-reductiveconversion of estrone to estradiol (anticipated estrogen 2-hydroxylation is because it decreases the pool of endoge conversion of 2-methoxyestrone to 2-methoxyestradiol; Refs. 139 and nous parent estrogen and thereby reduces the formation of 16a- 140). It is noteworthy that the 17/3-reductive activity of 17/3-HSD is hydroxyestrone and 16a-hydroxyestradiol. 16a-Hydroxyestrone, higher in mammary tumors than in normal breast that does not contain which is not only hormonally active but also chemically reactive (28, malignant cells (141-143). This increased 17ß-HSD(reductive activ 152, 153, 155), may play a role in estrogen-induced mammary cancer. ity) in mammary cancer cells not only provides these cells with a The concept of loa-hydroxylated estrogens as an important etiolog- favorable estrogenic environment for growth due to increased con ical factor in hormonal carcinogenesis, however, is weakened by the version of estrone to estradiol but may also increase the formation of very low carcinogenic activity of 16a-hydroxyestrone and loa-hy the antitumorigenic 2-methoxyestradiol from 2-methoxyestrone. The droxyestradiol () relative to estradiol or 4-hydroxyestradiol (63, amount of 2-methoxyestradiol formed in situ from 2-methoxyestrone 156). Moreover, some studies suggest that estrogen 16a-hydroxyla- would depend on the availability of 2-methoxyestrone and the level of tion is not markedly different among women with different risks for 17ß-HSD(reductive activity) in target cells. mammary cancer (114, 155, 157-159). To accommodate the 16a- Although long-term exposure to exogenously administered estra hydroxylation hypothesis with these experimental observations, em diol or estrone has been shown in several animal models to induce phasis has been shifted to the ratios of estrogen 2- to 16o-hydroxy- tumors in their target organs (1-7), surprisingly the very large amount lation as a biomarker for breast cancer risk (98, 152, 153, 155). This of endogenous estrogens present during human full-term is shift in emphasis tends to decrease the importance of the absolute associated with a decrease in the risk of human uterine and mammary extent of estrogen 16a-hydroxylation. cancers (144-147). It is possible that endogenous substances that are The recent findings of potent antitumorigenic activity for 2- produced during pregnancy possess protective activity against estro methoxyestradiol suggest a new explanation for the inverse relation gen-dependent tumor formation. Previous studies suggest that proges ship observed previously between the ratios of estrogen 2- to 16a- terone may be one of the inhibitory substances in animals and possibly hydroxylation and breast cancer risk. We believe a major reason that also in humans (148-151). Because human placenta produces large enhanced 2-hydroxylation of estradiol is associated with decreased amounts of estradiol and estrone, and this organ also contains high mammary carcinogenesis is because enhanced 2-hydroxylation in estrogen 2-hydroxylase and COMT activity, it is expected that large creases the formation of 2-methoxyestradiol, and this compound is a amounts of 2-methoxyestrogens will be produced by the placenta. potent antitumorigenic metabolite of estradiol (illustrated in Fig. 2). Consistent with these observations, a previous study (47) reported Possible Mechanisms of Action of 2-Methoxyestradiol that the blood concentration of unconjugated plus conjugated 2-methoxyestrone was —¿10,000pg/ml in pregnant women. Although The mechanism for the growth-inhibitory and antiangiogenic ef the level of 2-methoxyestradiol was not reported, it is likely that fects of 2-methoxyestradiol is not known. Observations indicating the substantial amounts are formed from 2-methoxyestrone in target cells ability of 2-methoxyestradiol to induce abnormal spindle formation in by the action of the reductive 17ß-HSD.The potent growth-inhibitory cultured cells (54), to interact with tubulin at the colchicine binding and antiangiogenic activity of 2-methoxyestradiol may protect against site (56), and to inhibit tubulin polymerization (56, 61 ) have led to the estrogen-induced carcinogenesis in target organs during human preg suggestion that the antitubulin activity of 2-methoxyestradiol may nancy. It will be of considerable interest to test this hypothesis. contribute in an important way to its cytotoxic effect. It should be noted, however, that the concentration of 2-methoxyestradiol or some Importance of 2-Methoxyestradiol for Understanding the Physiological Significance of the Estrogen 2-/16a-Hydroxylation of its synthetic analogues required for inhibition of tubulin polymer ization or colchicine binding to tubulin in vitro is considerably higher Ratio (low P.Mrange) than that required for inhibiting the growth of many Bradlow, Fishman, and their colleagues (98, 152-154) proposed different types of cancer cells in culture ( 10-100 nM; Refs. 56, 58, 59, that the extent of estrogen 16a-hydroxylation or the ratio of estrogen and 61). It is noteworthy that a recent study showed that 2-me 2- to 16a-hydroxylation is an important biomarker for the risk of thoxyestradiol at concentrations sufficient for causing complete met- breast cancer in animals and humans. The core of their hypothesis is aphase arrest in cultured cells does not inhibit the formation of mitotic that the products of the 16a-hydroxylation pathway (hormonally spindles (160). 2273

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research. 2-METHOXYKSTRA»10L AS AN INHIBITOR OF BREAST CANCER

In cultured H460 or A549 human lung cancer cells (both of which effect; (c) target cells that are particularly sensitive to growth inhibi contain the wild-type p53 gene), treatment with 5 JU.M2-methoxyestra- tion by 2-methoxyestradiol; (d) endogenous and exogenous fact diol increases the wild-type p53 protein levels and induces apoptosis ors that regulate the metabolic formation and disposition of 2- (161). However, 2-methoxyestradiol at the same concentration does methoxyestradiol in liver and in estrogen target cells; and (e) the not induce apoptosis in human lung cancer cells, either without the molecular mechanism(s) underlying the growth-inhibitory and anti- wild-type p53 gene or with a mutated p53 gene (161). These results angiogenic actions of 2-methoxyestradiol. Studies in these and related suggest that 2-methoxyestradiol at relatively high concentrations may areas will not only greatly enhance our understanding of the physio activate the wild-type p53-mediated apoptosis. logical roles of 2-methoxyestradiol, a nonpolar estradiol metabolite Treatment of MCF-7 human breast cancer cells with a low concen formed in large amounts in liver and in estrogen target cells, but these tration (10 nM) of 2-methoxyestradiol caused marked changes (an studies may also offer novel mechanistic insights and strategies for the increase or decrease) in the intracellular cyclic AMP concentration prevention and treatment of estrogen-associated cancers. depending on the phase of the cell cycle, whereas 10 nw estradici or 2-hydroxyestradiol had little or no effect (55). This change of cyclic AMP levels during 2-methoxyestradiol treatment was accompanied References by marked changes in the phosphorylation patterns of various cellular 1. Clifton. K. H., and Meyer. R. K. Mechanism of anterior pituitary tumor induction by proteins (55). A recent report indicated that treatment of MCF-7 cells oestrogen. Anal. Ree.. 125: 65-81. 1956. with 10 nM 2-methoxyestradiol selectively altered the expression of 2. Kirkman. H. Estrogen-induced tumors of the kidney in Syrian hamster. III. Growth characteristics in the Syrian hamster. Nail. Cancer Insi. Monogr.. /: 1-57. 1959. several kinases involved in cell cycle control (162, 163). These 3. Cults. J. H.. and Noble. R. L. F.slrone-induced mammary tumors in the rat. I. biochemical responses to very low concentrations of 2-methoxyestra Induction and behavior of tumors. Cancer Res., 24: 11 16-1 123. 1964. 4. Newbold. R. R.. Bullock. B. C.. and MacLachlan. J. A. Uterine adenocarcinoma in diol may be physiologically important, but more studies are needed. mice following developmental treatment with estrogen. Cancer Res.. 50: 7677- The high specificity and high potency of 2-methoxyestradiol as an 7681. 1990. inhibitor of angiogenesis and tumor cell growth are of great inter 5. Nandi. S.. Gu/ma. R. C.. and Yang. J. Hormones and mammary carcinogenesis in mice, rats, and humans: a unifying hypothesis. Proc. Nati. Acad. Sci. USA, 92: est and have led us to suggest that some of the unique effects of 3650-3657. 1995. 2-methoxyestradiol may be mediated by a specific receptor or intra 6. Yager. J. D., and Liehr. J. G. Molecular mechanisms of estrogen carcinogenesis. cellular effector (such as a transcriptional factor or an enzyme) that is Annu. Rev. Pharmacol. Toxicol.. 36: 203-232. 1996. 7. Li, J. J.. and Li. S. A. The effects of hormones on tumor induction, tu: J. G. Arcos. refractory to estradiol. Candidate receptors for 2-methoxyestradiol M. F. Argus, and Y. T. Woo (eds.). Chemical Induction of Cancer, pp. 396-449. include: (a) the so-called nuclear type II estrogen binding sites, which Boston. MA: Birkhauser. 1996. have a low apparent binding affinity for estradiol (164-166) and 8. Ziel. H. K.. and Finkle. W. D. Increased risk of endometrial carcinoma among users of . N. Engl. J. Med.. 29.1: 1167-1170. 1975. functionally may mediate a growth-inhibitory effect in estrogen- 9. Siiter. P. K.. Williams. J. E.. and Takaki. N. K. abnormalities in endometrial sensitive cells (167): (b) one or more of the multiple variants of the and breast carcinoma: a unifying hypothesis. J. Steroid. Biochem.. 7: 897-903. classical estrogen receptor, which are present in some estrogen target 1976. 10. Mack. T. M.. Pike. M. C., Henderson. B. E.. Pfeffer. R. I., Gerkins. V. R.. Arthur. tissues or cells (168, 169); (<•)anovel estrogen receptor that was M., and Brown. S. E. Estrogens and endometrial cancer in a retirement community. N. Engl. J. Med.. 294: 1262-1267. 1976. recently found to be expressed in rat prostate and ovary (170); and (d) 11. McDonald. T. W., Annegers. J. F.. O'Fallow. W. M., Dockerty, M. B.. Malkasian. certain member(s) of the nuclear orphan receptor family (171, 172). G. D.. Jr.. and Karland. L. T. Exogenous estrogens and endometrial carcinoma: case Whatever the structural identity of the putative intracellular effector control and incidence study. Am. J. Obstet. GynecoL 127: 572-580. 1977. or receptor for 2-methoxyestradiol. we believe that the interaction of 12. Grady. D.. and Ernster. V. L. Endometrial Cancer. In: D. Schottenfeld and J. F. Fraumeni. Jr. (eds.). Cancer Epidemiology and Prevention. Ed. 2. pp. 1058-1089. 2-methoxyestradiol with its specific effector or receptor in target cells Oxford: Oxford University Press. 1996. is an initial step leading to the expression of its growth-inhibitory 13. Jick. H.. Walker, A. M.. Watkins. R. N., D'Ewart. D. C.. Hunter. J. R.. Danford, A., Madsen. S., Diñan.B. J.. and Rothman. K. J. Replacement estrogens and breast effects in sensitive cells. An understanding of the mechanism of action cancer. Am. J. Epidcmiol.. 112: 586-594. 1980. of 2-methoxyestradiol should greatly enhance our understanding of its 14. Pike. M. C.. Spicer. D. V., Dahmoush. L., and Press, M. F. Estrogens, progcstagens. unique physiological and antitumorigenic functions. normal breast cell proliferation, and breast cancer risk. Epidemiol. Rev., 15: 17-35, 1993. 15. Kelsey. J. L.. and Gammon. M. D. The epidemiology of breast cancer. CA Cancer Concluding Remarks J. Clin.. 41: 146-165. 1991. 16. Feigelson. H. S.. and Henderson. B. E. Estrogens and breast cancer. Carcinogenesis We have described recent studies on the antitumorigenic and anti- (Lond.l. 17: 2279-2284. 1996. 17. Silver. M. A quantitative analysis of the role of oestrogen in mammary development angiogenic effects of exogenously administered 2-methoxyestradiol in in the rat. J. Endocrino!.. IO: 17-34. 1953. vitro and in vivo. We have also discussed data which suggest that 18. Martin. L.. Finn. C. A., and Trinder. G. Hypertrophy and hyperplasia in the mouse endogenously formed 2-methoxyestradiol may have a protective ef uterus after estrogen treatment: an autoradiogruphic study. J. Endocrinol., 56: 133-141. 1973. fect against estrogen-induced cancers in target organs. Although the 19. Banerjee. M. R. Responses of mammary cells to hormones. Int. Rev. Cytol.. 4: 1-97, molecular mechanisms of action of 2-methoxyestradiol are not 1976. known, a hypothesis is proposed that some unique effects of 2- 20. Quarmby. V. E.. and Korach, K. S. Influence of 17/3-estradiol on the pattern of cell division in the uterus. Endocrinology. /14: 694-702. 1984. methoxyestradiol may be mediated by its interaction with a specific 21. Holland. M. B.. and Roy. D. Estrogen-induced cell proliferation and differentiation cellular receptor or effector that is refractory to estradiol. in the mammary gland of the female Noble rat. Carcinogenesis (Lond.). 16: 1955- Although the data reviewed here collectively suggest that 2- 1961. 1995. 22. Bradlow. H. L.. Hershcopf, R. J.. Martucci. C. P.. and Fishman. J. Estradiol methoxyestradiol may be an important endogenously formed estrogen I6o-hydroxylation in the mouse correlates with mammary tumor incidence and metabolite that protects cells from estrogen-induced carcinogenesis, it presence of mammary tumor virus: a possible model for the hormonal etiology of breast cancer in humans. Proc. Nati. Acad. Sci. USA. 82: 6295-6299. 1985. should be pointed out that there are still many important unresolved 23. Liehr. J. G.. Ulubelen. A. A., and Strobcl. H. W. Cytochrome P-450-mediated redox questions about this estradiol metabolite that require more research. It cycling of estrogens. J. Biol. Chem., 261: 16865-16870. 1986. 24. Swaneck, G. E.. and Fishman, J. Covalent binding of the endogenous estrogen will be important to find out: (a) the concentrations of endogenously 16a-hydroxyestronc to estradiol receptor in human breast cancer cells: character formed 2-methoxyestradiol in the circulation and in estrogen target ization and intranuclear locali/ation. Proc. Nati. Acad. Sci. USA. 85: 7831-7835. tissues or cells under different physiological or pathophysiological 1988. 25. Liehr, J. G. Genotoxic effects of estrogens. Mutât.Res.. 238: 269-276, 1990. conditions; (b) the effective circulating or intracellular concentrations 26. Liehr. J. G., and Roy. D. Free radical generation by redox cycling of estrogens. Free of 2-methoxyestradiol that are needed to exert an antitumorigenic Radical Biol. Med.. 8: 415-423. 1990. 2274

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research. :-METHOXYI-.SlRAl>IOL AS AN INHIBITOR OF BREAST CANCER

27. Nutter, L. M., Ngo. E. O., and Abul-Hajj. Y. J. Characterization of DNA damage inhibits angiogenesis and suppresses tumor growth. Nature (Lond.). 368: 237-239. induced by 3,4-estrone-ri-quinone in human cells. J. Biol. Chem., 266: 16380- 1994. 16386, 1991. 58. Cushman. M.. He, H. M.. Katzenellenhogcn. J. A.. Lin. C. M.. and Hamel. E. 28. Telang, N. T., Sato. A.. Wong. G. Y.. Osborne. M. P.. and Bradlow. H. L. Induction Synthesis, antitubulin and antimitotic activity, and cytotoxicity of analogs of 2-me- by estrogen metabolite 16a-hydroxyestrone of genotoxic damage and aberrant thoxyestradiol. an endogenous mammalian metabolite of estradiol that inhibits proliferation in mouse mammary epithelial cells. J. Nati. Cancer Inst.. H4: 634-638. lubulin polymerization by binding to the colchicine binding site. J. Med. Chem.. 38: 1992. 2041-2049. 1995. 29. Han. X.. and Liehr. J. G. 8-Hydroxylation of guanine bases in kidney and liver DNA 59. Cushman. M.. He. H. M.. Kat/enellenbogen. J. A.. Vanna. R. K.. Hamel. E., Lin. of hamsters treated with estradiol: role of free radicals in estrogen-induced carci- C. M., Ram. S., and Sachdeva. Y. P. Synthesis of analogs of 2-methoxyestradiol nogenesis. Cancer Res., 54: 5515-5517. 1994. with enhanced inhibitory effects on tubulin polymerization and cancer cell growth. 30. Han. X.. and Liehr, J. G. DNA single-strand breaks in kidneys of Syrian hamsters J. Med. Chem.. 40: 2323-2334. 1997. treated with steroidal estrogens: hormone-induced free radical damage preceding 60. Nakagawa-Yagi. Y.. Ogane. N.. Inoki. Y.. and Kitoh. N. The endogenous estrogen renal malignancy. Carcinogenesis (Lond.). 15: 997-1000, 1994. metabolite 2-methoxyestradiol induces apoplotic neuronal cell death I'Mritrn. Life 31. Nutter, L. M.. Wu, Y-Y.. Ngo. E. O.. Sierra. E. E.. Gutierrez, P. L.. and Abul-Hajj. Sci., 58: 1461-1467. 1996. Y. J. An u-quinone form of estrogen produces free radicals in human breast cancer 61. Klauber. N.. Parangi. S.. Flynn. E.. Hamel. E., and D'Amato. R. J. Inhibition of cells: correlation with DNA damage. Chem. Res. Toxicol., 7: 23-28. 1994. angiogenesis and breast cancer in mice by the microtubule inhibitors 2-me- 32. Cavalieri. E. I... Stack. D. E.. Devanesan, P. D., Todorovic. R.. Dwivedy, I.. thoxyestradiol and taxol. Cancer Res.. 57: 81-86. 1997. Higginbotham. S.. Johansson. S. L., Patii, K. D.. Gross. M. L.. Gooden. J. K.. 62. Yue. T. L., Wang. X.. Louden, C. S., Gupta. S.. Pillarisetti. K.. Gu. J. L.. Hart, T. K.. Ramanathan. R.. Cerny. R. L., and Rogan. E. G. Molecular origin of cancer: catechol Lysko. P. G.. and Feuerstein. G. Z. 2-Methoxyestradiol. an endogenous estrogen estrogen-3,4-quinones as endogenous tumor initiators. Proc. Nati. Acad. Sci. USA, metabolite, induces apoplosis in endothelial cells and inhibits angiogenesis: possible 94: 10937-10942, 1997. role for stress-activated protein kinase signaling pathway and Fas expression. Mol. 33. Manucci. C. P., and Fishman. J. P450 enzymes of estrogen metabolism. Pharmacol. Pharmacol.. 51: 951-962. 1997. Then. 57: 237-257. 1993. 63. Li. J. J.. and Li. S. A. Estrogen Carcinogenesis in Syrian hamster tissues: role of 34. Zhu. B. T.. and Conney. A. H. Functional role of estrogen metabolism in target cells: metabolism. Fed. Proc.. 46: 1858-1863. 1987. review and perspectives. Carcinogenesis (Lond.), 19: 1-27. 1998. 64. Zhu. B. T.. and Liehr. J. G. Quercetin increases the severity of eslradiol-induced 35. Axelrod. J.. and Tomchick. R. En/ymatic O-methylation of epinephrine and other tumorigenesis in hamster kidney. Toxicol. Appi. Pharmacol.. 125: 149-158. 1994. catechols. J. Biol. Chem.. 233: 702-705. 1958. 65. Zhu. B. T.. and Liehr. J. G. Inhibition of catechol O-methyltransferase-cataly/.ed 36. Axelrod. J.. Alhers. W.. and Clemente. C. D. Distribution of catechol-O-methyl- O-methylation of 2- and 4-hydroxyestradiol by quercetin: possible role in estradiol- transferase in the nervous system and other tissues. J. Ncurochem.. 5: 68-71. 1959. induced tumorigenesis. J. Biol. Chem.. 271: 1357-1363. 1996. 37. Guldberg, H. C.. and Marsden. C. A. Catechol-O-methyltransferase. Pharmacolog- 66. Liehr. J. G.. Fang. W. F.. Sirbasku. D. A., and Ulubelen. A. A. Carcinogenicity of ical aspects and physiological role. Pharmacol. Rev.. 27: 135-206, 1975. catechol estrogens in Syrian hamsters. J. Steroid. Biochem.. 24: 353-356, 1986. 38. Inoue. K.. Tice. L. W.. and Creveling. C. R. Immunocylochemical localization of 67. Metzler. M. The metabolism of dielhylstilbestrol. CRC Crii. Rev. Biochem.. 10: catechol-O-methyltransferase. In: E. Usdin. N. Weiner, and M. B. H. Youdim (eds.). 171-212. 1981. Structure and [-'unction of Monoamine Enzymes, pp. 835-859. New York: Marcel 68. Metzler. M. Biochemical toxicology of diethylstilhestrol. In: E. Hodgson, J. R. Dekker. 1977. Bend, and R. M. Philpot (eds.). Reviews in Biochemical Toxicology. Vol. 6, pp. 39. Assicot. M.. Contesso. G., and Bohuon. C. Catechol-O-methyltransferase in human 191-220. New York: Elsevier. 1984. breast cancers. Eur. J. Cancer. 13: 961-966. 1977. 69. (jladek. A., and Liehr. J. G. Mechanism of genotoxicity of diethylstilbestrol in vivti. 40. Amin. A. M.. Creveling, C. R.. and Lowe. M. C. Immunohistochemical localization J. Biol. Chem.. 264: 16847-16852. 1989. of catechol methyltransferase in normal and cancerous breast tissues of mice and 70. Verma. A. K.. Johnson, J. A.. Gould. M. N.. and Tanner. M. A. Inhibition of rats. J. Nati. Cancer Inst.. 70: 337-342. 1983. 7.12-dimethylhenz|i/|anthracene- and A'-nilrosomclhylurca-induced rat mammary 41. Thakker. D. R.. and Creveling. C. R. O-Methylation. In: G. J. Mulder (ed.). cancer by dietary flavonol quercetin. Cancer Res.. 48: 5754-5758. 1988. Conjugation Reactions in , pp. 193-232. London: Taylor & 71. Deschner. E. E.. Ruperto. J.. Wong. G.. and Newmark. H. L. Quercetin and rulin as Francis, Ltd., 1990. inhibitors ofazoxymethanol-induced colonie neoplasia. Carcinogenesis (Lond.). 12: 42. Guldberg, H. C.. and Marsden, C. A. Catechol-O-methyltransferase. Pharmacolog 1193-1196. 1991. ical aspects and physiological role. Pharmacol. Rev., 27: 135-206. 1975. 72. Zhu. B. T.. and Liehr. J. G. Inhibition of the catechol-O-methyltransferase-catalyzed 43. Ball. P.. and Knuppen. R. Catecholoestrogens (2- and 4-hydroxyoestrogens): chem O-methylalion of 2- and 4-hydroxyestradiol by catecholamines: implications for the istry, biogenesis, metabolism, occurrence and physiological significance. Acta mechanism of estrogen-induced Carcinogenesis. Arch. Biochem. Biophys., 304: Endocrino!., 232: 1-127. 1980. 248-256. 1993. 44. Roy. D.. Weisz. J., and Liehr, J. G. The O-methylation of 4-hydroxyestradiol is 73. Riley, V. Mouse mammary tumors: alteration of incidence as apparent function of inhibited by 2-hydroxyestradiol: implications for estrogen-induced Carcinogenesis. stress. Science (Washington DC). 189: 465-467, 1975. Carcinogenesis (Lond.). //: 459-462. 1990. 74. Cooper. C. L. (ed.) Stress and Breast Cancer. New York: John Wiley and Sons. 45. Zhu, B. T., Roy. D.. and Liehr. J. G. The carcinogenic activity of ethinyl estrogens 1988. is determined by both their hormonal characteristics and their conversion to catechol 75. Faragher. E. B.. and Cooper. C. L. Type A stress prone behavior and breast cancer. metabolites. Endocrinology. ¡32:577-583, 1993. Psychol. Med.. 20: 663-670. 1990. 46. Zhu. B. T., Bui, Q. D.. Weisz. J.. and Liehr. J. G. Conversion of estrone to 2- and 76. Forsen. A. Psychosocial stress as a risk for breast cancer. Psychother. Psychosom.. 4-hydroxyestrone by hamster kidney and liver microsomes: implications for the 55: 176-185. 1991. mechanism of estrogen-induced Carcinogenesis. Endocrinology, 135: 1772-1779. 77. Green. S.. and Watson. M. Towards a psychobiological model of cancer: psycho 1994. logical considerations. Soc. Sci. Med.. 20: 773-777, 1985. 47. Ball, P., and Knuppen. R. Formation, metabolism, and physiologic importance of 78. Aragona. M.. Muscatello. M. R.. Losi. E.. Panetta. S.. La Torre. F.. Pastura, G.. catecholestrogens. Am. J. Obstet. Gynecol.. 163: 2163-2170. 1990. Bertolani. S.. and Mesiti. M. Lymphocyte number and stress parameter modifica 48. Dunn. J. F. Transport of estrogens in human plasma. In: G. R. Merriam and M. B. tions in untreated breast cancer patients with depressive mood and previous life Lipsett (eds.). Catechol Estrogens, pp. 167-176. New York: Raven Press. 1983. stress. J. Exp. Ther. Oncol., /: 354-360, 1996. 49. Fishman. J., and Martucci. C. P. Biological properties of I6a-hydroxyestrone: 79. Cohn. C. K., Dunner. D. L.. and Axelrod. J. Reduced calechol-O-methyltranslerase implications in estrogen physiology and pathophysiology. J. Clin. Endocrinol. activity in red blood cells of women with primary affective disorder. Science Metab.. 51: 611-615. 1980. (Washington DC). 170: 1323-1324. 1970. 50. Merriam. G. R.. MacLusky. N. J.. Picard. M. K., and Naftolin. F. Comparative 80. Weinshilboum. R. M.. Raymond. F. A.. Elveback. L. R.. and Weidman. W. H. properties of the catechol estrogens. I. Methylation by catechol-O-methyltransferase Correlation of erythrocyle catechol-O-methyltransferase activity between siblings. and binding to cytosol estrogen receptors. . 36: 1-11. 1980. Nature (Lond.). 252: 490-491. 1974. 51. Gordon. S. Cantrall, E. W.. Cekleniak. W. P., Albers. H. J.. Mauer. S.. Stolar. S. M.. 81. Weinshilhoum. R. M., and Raymond. F. A. Inheritance of low erythrocyte catechol- and Bernstein. S. Steroid and lipid metabolism. The hypocholesteremic effect of O-methyltransferase activity in man. Am. J. Hum. Genet.. 29: 125-135, 1977. estrogen metabolites. Steroids. 4: 787-791. 1964. 82. Scanlon. P. D.. Raymond. F. A., and Weinshilboum. R. M. Calechol-O-niethyltrans- 52. Martucci, C. Induction of rat plasma renin substrate by 2-methoxyestrone. The ferase: thermolahile enzyme in erythrocytes of subjects homozygous for alíelefor Endocrine Society 64th Annual Meeting, abstract 889. 1982. low activity. Science (Washington DC), 203: 63-65. 1979. 53. Katayama. S.. and Fishman. J. 2-Hydroxyestrone suppresses and 2-methoxyestrone 83. Weinshilboum. R. Melhyltransferase pharmacogenetics. Pharmacol. Ther.. 43: 77- augments the preovulatory prolactin surge in the cycling rat. Endocrinology. /10: 90. 1989. 1448-1450. 1982. 84. Boudikova. B.. Szumlanski. C.. Maidak. B.. and Weinshilboum. R. Human liver 54. Seegers, J. C.. Aveling, M-L.. van Aswegen. C. H.. Cross. M.. Koch, F.. and Joubert, catechol-O-methyllransferase pharmacogenetics. Clin. Pharmacol. Ther., 4S: 381- W. S. The cytotoxic effects of estradiol-17/3. catecholestradiols and methoxyestra- 389. 1990. diols on dividing MCF-7 and HeLa cells. J. Steroid Biochem., 32: 797-809, 1989. 85. Lundstrom. K.. Saiminen. M.. Jalanko. A., Savolainen, R.. and Ulmanen. I. Chining 55. Lottering, M-L., Haag, M., and Seegers, J. C. Effects of 17ß-estradiolmetabolites on and characterization of human placental catcchol-O-methyltransferase cDNA. DNA cell cycle events in MCF-7 cells. Cancer Res., 52: 5926-5932. 1992. Cell Biol., 10: 181-189. 1991. 56. D'Amato. R. J., Lin. C. M.. Flynn, E.. Folkman. J.. and Hamel, E. 2-Methoxyestra- 86. Bertocci. Miggiano, V., Parada. M. D.. Dembic, Z.. Lahm. H-W.. and Malherbe. P. diol. an endogenous mammalian metabolite, inhibits tubulin polymerization by Human catechol-O-methyltransferase: cloning and expression of the membrane- interacting at Ihc colchicinc Nile. Proc. Nail. Acad. Sci. USA. "/.- 3Q64-.1968. 1994. associaled form. Proc. Nati. Acad. Sei. USA. XX: 1416-1420. 1991. 57. Fotsis, T.. Zhang. Y., Pepper, M. S.. Adlercreutz. H.. Montesano. R.. Nawrolh. P. P.. 87. Tenhunen. J„Salmincn. M.. Lundstrom. K.. Kiviluolo, T., Savolainen, R., and and Schweigerer. L. The endogenous oestrogen metabolite 2-methoxyoestradiol Ulmanen. I. Genomic organization of the human calechol-O-methyltransferase gene 2275

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research. 2-METHOXYESTRADIOL AS AN INHIBITOR OF BREAST CANCER

and its expression from two disiine! promoters. Eur. J. Biochem., 223: 1049-1059. 115. Miller. B. A., Ries, L. A. G., Hankey, B. F., Kosary, C. L.. Harris, A., Devesa. S. S., 1994. and Edwards, B. K. (eds.). SEER Cancer Statistics Review: 1973-1990. NIH 88. Lachman. H. M., Papólos. D. F., Saito. T.. Yu. Y-M, Szumlanski, C. L.. and Publication No. 93-2789. Bethesda. MD: National Cancer Institute, 1993. Weinshilboum, R. M. Human catechol-O-methyltransferase pharmacogenetics: de- 116. Lipsett, M. B.. Merriam, G. R., Kono. S., Brandon. D. D.. Pfeiffer. D. G.. and scription of a functional polymorphism and its potential application to neuropsychi Loriaux. D. L. Metabolic clearance of catechol estrogens. In: G. R. Merriam and atrie disorders. Pharmacogenetics, 6: 243-250. 1996. M. B. Lipsett (eds.). Catechol Estrogens, pp. 105-114. New York: Raven Press, 89. Chen, C. H., Lee, Y. R., Liu, M. Y., Wei, F. C, Koong, F. J., Hwu. H. G., and Hsiao, 1983. K. J. Identification of a Ugll polymorphism of catechol-O-methyltransferase 117. Emons. G.. Merriam. R.. Pfeiffer. D.. Loriaux. D. L.. Ball, P., and Knuppen. R. (COM7Ìgene and association study with schizophrenia. Am. J. Med. Genet.. 67: Metabolism of exogenous 4- and 2-hydroxyestradiol in human male. J. Steroid 556-559, 19%. Biochem., 28: 499-504, 1987. 90. Karayiogou. M., Altemus. M.. Galke. B. L.. Goldman. D.. Murphy. D. L.. Ott. J., and 118. Davies, I. J., Naftolin, F.. Ryan. K. J., Fishman. J.. and Sin, J. The affinity of Gogos. J. A. Genotype determining low activity catechol-O-methyltransferase ac catechol estrogens for estrogen receptors in the pituitary and anterior hypothalamus tivity as a risk factor for obsessive-compulsive disorder. Proc. Nati. Acad. Sci. USA, of the rat. Endocrinology, 97: 554-557, 1975. 94: 4572-4575. 1997. 119. Martucci. C.. and Fishman. J. Uterine estrogen receptor binding of catecholestrogens 91. Lavigne, J. A., Helzlsouer, K. J.. Huang. H-Y.. Strickland. P. T., Bell. D. A., Seimin, and of [l,3.5(10)estratriene-3,15a,16a.l7/3-tetrol]. Steroids, 27: 325-333. O., Watson, M. A.. Hoffman, S.. Comstock, G. W., and Yager, J. D. An association 1976. between the alíelecoding for a low activity variant of catechol-O-methyltransferase 120. Martucci. C., and Fishman. J. Impact of continuously administered catechol estro and the risk for breast cancer. Cancer Res., 57: 5493-5497. 1997. gens on uterine growth and LH secretion. Endocrinology. 105: 1288-1292. 1979. 92. Welsch. C. W. Host factors affecting the growth of carcinogen-induced rat mam- 121. Fishman. J. Biological action of catecholestrogens. J. Endocr.. 85: 59P-65P. 1981. mary carcinoma: a review and tribute to Charles Brenlon Huggins. Cancer Res.. 45: 122. Schneider. J.. Huh. M. M.. Bradlow, H. L., and Fishman, J. action of 3415-3443. 1985. 2-hydroxyestrone on MCF-7 human breast cancer cells. J. Biol. Chem., 259: 93. Jordan. V. C.. Lababidi. M. K.. and Mirecki, D. M. Anti-oestrogenic and anti-tumour 4840-4845. 1984. properties of prolonged therapy in C3H/OuJ mice. Eur. J. Cancer. 26: 123. Vandewalle, B., and Lefebvre. J. Opposite effects of estrogen and catecholestrogen 718-721, 1990. on hormone-sensitive breast cancer cell growth and differentiation. Mol. Cell. 94. Jordan. V. C.. Lababidi. M. K.. and Langan-Fahey, S. Suppression of mouse Endocrinol.. 61: 239-246, 1989. mammary tumorigenesis by long-term tamoxifen therapy. J. Nati. Cancer Inst., 83: 124. van Aswegen. C. H.. Purdy. R. H., and Wittliff, J. L. Binding of 2-hydroxyestradiol 492-496, 1991. and 4-hydroxyestradiol to estrogen receptor human breast cancers. J. Steroid Bio 95. Osbome, M. P., Telang, N. T.. Kaur. S., and Bradlow. H. L. Influence of chemo- chem., 32: 485-492. 1989. prevenlive agents on estradiol metabolism and mammary preneoplasia in the C3H 125. Schütze.N.. Vollmer. G.. Tiemann. I., Geiger. M., and Knuppen, R. Catecholestro mouse. Steroids. 55: 114-119. 1990. gens are MCF-7 cell estrogen agonists. J. Steroid Biochem. Mol. Biol., 46: 781-789. 96. Bradlow, H. L.. Michnovicz. J. J.. Telang. N. T.. and Osborne. M. P. Effects of 1993. dietary indole-3-carbinol on estradiol metabolism and spontaneous mammary tu- 126. Schütze.N.. Vollmer. G.. and Knuppen. R. Catecholestrogens are agonists of mors in mice. Carcinogenesis (Lond.). 12: 1571-1574. 1991. estrogen receptor-dependent gene expression in MCF-7 cells. J. Steroid Biochem. 97. Jellinck. P. H., Michnovicz. J. J.. and Bradlow, H. L. Influence of indole-3-carbinol Mol. Biol., 48: 453-461, 1994. on the hepatic microsomal formation of catechol estrogens. Steroids, 56: 446-450. 127. van Landeghemm. A. A.. Poortman. J.. Nabuurs, M.. and Thijssen, J. H. H. 1991. Endogenous concentration and subcellular distribution of estrogens in normal and 98. Bradlow. H. L.. Sepkovic. D. W.. Telang. N. T., and Osborne, M. P. Indole-3- malignant human breast tissue. Cancer Res.. 45: 2900-2906. 1985. carbinol: a novel approach to breast cancer prevention. Ann. NY Acad. Sci., 768: 128. Samen. R. J. Determinants of tissue oestradiol levels in human breast cancer. Cancer 180-200, 1995. Surv., 5: 597-616, 1986. 99. Kojima. T., Tanaka. T., and Mori. H. Chemoprevention of spontaneous endomelrial 129. Hoffman. A. R., Paul, S. M.. and Axelrod. J. Catecholestrogen synthesis and cancer in female Donryu rats by dietary indole-3-carbinol. Cancer Res., 54: 1446- metabolism by human breast tumors in vitro. Cancer Res., 39: 4584-4587. 1979. 1449, 1994. 130. MacLusky, N. J.. Naftolin. F.. Krey. L. C.. and Franks. S. The catechol estrogen. J. 100. Grubbs, C. J., Steele, V. E., Casebolt. T.. Juliana, M. M., Elo, I., Whitaker. L. M.. Steroid Biochem., 15: 111-124. 1981. Dragnev. K. H.. Kelloff. G. J., and Lubet. R. L. Chemoprevention of chemically- 131. Levin, M.. Weisz, J., Bui, Q. D., and Santen. R. J. Peroxidatic catecholestrogen induced mammary Carcinogenesis by indole-3-carbinol. Anticancer Res., 15: 709- production by human breast cancer tissue in vitro. J. Steroid Biochem., 28: 513-520, 716, 1995. 1987. 101. Michnovicz, J. J., Hershcopf, R. J., Naganuma. H., Bradlow. H. L., and Fishman. 132. Abul-Hajj. Y. J.. Thijssen. J. H., and Blankenstein. M. A. Metabolism of estradiol by J. Increased 2-hydroxylation of estradiol as a possible mechanism for the anti- human breast cancer. Eur. J. Cancer Clin. Oncol., 24: 1171-1178, 1988. estrogenic effect of cigarette smoking. N. Engl. J. Med.. 315: 1305-1309. 1986. 133. Liehr. J. G., and Ricci. M. J. 4-Hydroxylation of estrogens as a marker of human 102. Michnovicz. J. J.. Naganuma. H.. Hershcopf. R. J.. Bradlow, H. L.. and Fishman. mammary tumors. Proc. Nati. Acad. Sci. USA. 93: 3294-3296. 1996. J. Increased urinary calechol estrogen excretion in female smokers. Steroids. 52: 134. Williamson. D. G. The biochemistry of the 17-hydroxysteroid dehydrogenases. In: 69-83. 1988. R. Hobkirk (ed.). Steroid Biochemistry, Vol. I, pp. 83-110. Boca Raton. FL: CRC 103. Michnovicz, J. J., Hershcopf, R. J., Haley. N. J., Bradlow, H. L., and Fishman, J. Press, 1979. Cigarette smoking alters hepatic estrogen metabolism in men: implications for 135. Poutanen, M.. Isomaa. V.. Peltoketo. H., and Vihko. R. Regulation of oestrogen atherosclerosis. Metab. Clin. Exp. 38: 537-541. 1989. action: role of 17ß-hydroxysteroid dehydrogenases. Ann. Med.. 27: 675-682, 1995. 104. MacMahon. B., Trichopoulos. D.. Cole, P., and Brown. J. Cigarette smoking and 136. Tseng, L.. and Gurpide, E. Changes in the in vitro metabolism of estradiol during the urinary estrogens. N. Engl. J. Med.. 307: 1062-1065. 1982. menstrual cycle. Am. J. Obstet. Gynecol., 114: 1002-1008, 1972. 105. Jensen, J., Christiansen. C.. and Rodbro. P. Cigarette smoking, serum estrogens, and 137. Tseng, L.. and Gurpide. E. Induction of human endometrial estradiol dehydrogenase bone loss during hormone-replacement therapy early after menopause. N. Engl. by progestins. Endocrinology, 97: 825-833. 1975. J. Med.. 313: 973-975, 1985. 138. Hobkirk. R., and Glasier, M. A. Generation of estradiol within the pregnant guinea 106. Friedman. A. J.. Ravnikar. V. A., and Barbieri. R. L. Serum steroid hormone profiles pig uterine compartment with special reference to the myometrium. J. Steroid in postmenopausal smokers and nonsmokers. Fértil.Steril.. 47: 398-401. 1987. Biochem. Mol. Biol., 44: 291-297, 1993. 107. Baron, J. A. Smoking and estrogen-related disease. Am. J. Epidemiol.. 119: 9—22. 139. Poutanen. M.. Isomaa, V.. Peltokelo. H., and Vihko, R. Role of 17ß-hydroxysteroid 1984. dehydrogenase type I in endocrine and intracrine estradiol . J. Steroid 108. Lesko, S. M.. Rosenberg. L.. Kaufman. D. W., Heimlich, S. P.. Miller. D. R., Strom. Biochem. Mol. Biol.. 55: 525-532, 1995. B., Schollenleid. D.. Rosenshein, N. B.. Knapp. R. C., Lewis, J., and Shapiro, S. 140. Luu-The. V., Zhang, Y.. Poirier, D., and Labrie. F. Characteristics of human types Cigarette smoking and the risk of endometrial cancer. N. Engl. J. Med., 313: 1, 2 and 3 17ß-hydroxysteroid dehydrogenase activities: Oxidation/reduction and 593-596. 1985. inhibition. J. Steroid Biochem. Mol. Biol.. 55: 581-587, 1995. 109. Baron, J. A.. Vecchia. C. L., and Levi. F. The antiestrogenic effect of cigarette 141. Bonney, R. C., Reed. M. J.. Davidson. K.. Beranek, P. A., and James. V. H. T. The smoking in women. Am. J. Obstet. Gynecol., /62: 502-514, 1990. relationship between 17j3-hydroxysteroid dehydrogenase activity and oestrogen 110. Taioli, E., Trachman. J., Chen, X.. Sepkovic, D. W.. Bradlow, H. L., and Toniolo. concentrations in human breast tumours and in normal breast tissue. Clin. Endocri P. Correlation of estradiol 2-hydroxylation and breast cancer risk in patients carrying nol., 19: 727-739, 1983. (he MSP-1 polymorphism of CYP4501A1. Proc. Am. Assoc. Cancer Res., 37: 249, 142. McNeill, J. M., Reed. M. J., Beranek. P. A.. Bonney. R. C.. Ghilchik. M. W.. 1996. Robinson, D. J.. and James, V. H. A comparison of the in vivo uptake and 111. Taioli. E.. Trachman, J., Chen. X.. Toniolo. P., and Garte. S. J. A CYP1A1 restriction metabolism of [^HJoeslrone and [3H)oestradiol by normal breast and breast tumour fragment length polymorphism is associated with breast cancer in African-American tissue in postmenopausal women. Int. J. Cancer. 38: 193-196. 1986. women. Cancer Res., 55: 3757-3758. 1995. 143. Vermeulen. A., Deslypere, J. P.. Pavidaens, R.. Leclevq. G.. Roy. F., and Henson, 112. Bailey. L. R.. Roodi, N.. Verrier, C. S.. Yee. C. J.. Dupont. W. D., and Pari, F. F. J. C. Aromatase, 17ß-hydroxysteroid dehydrogenase and intratissular sex hormone Breast cancer and CYPIAI. GSTMI. and GSTT1 polymorphisms: evidence of a lack concentrations in cancerous and normal glandular breast tissue in post-menopausal of association in Caucasians and African Americans. Cancer Res.. 58: 65-70. 1988. women. Eur. J. Cancer Clin. Oncol.. 32: 515-522. 1986. 113. Taioli. E.. Garte, S., Trachman. J.. Garbers. S.. Sepkovic. D. W., Osbome. M. P., 144. Logan, W. P. D. Marriage and childbearing in relation to cancer of the breast and Mehl. S.. and Bradlow. H. L. Ethnic differences in estrogen metabolism in healthy uterus. Lancet. 2: 1199-1202. 1953. women. J. Nati. Cancer Inst., 88: 617, 1996. 145. Joly, D. J., Lilienfeld, A. M., Diamond, E. L.. and Brass, I. D. J. An epidemiologie 114. Coker, A. L., Crane, M. M.. Sticca. R. P.. and Sepkovic, D. W. R. Ethnic differences study of the relationship of reproductive experience to cancer of the ovary. Am. J. in estrogen metabolism in healthy women. J. Nati. Cancer Inst.. 89: 89-90. 1997. Epidemiol., 99: 190-209. 1974. 2276

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research. 2-METHOXYESTRAOIOL AS AN INHIBITOK OH BKI ASI ( AMÕ.K

146. Pathak. D. R.. Speizer. F. E.. Wille»,W. C.. Rosner. B., and Lipnick. R. J. Parity and 160. Attalla. H.. Makela. T. P.. Adlercreutz. H.. and Andcrsson. L. C. 2-Methoxyestradiol breast cancer risk: possible effect on age at diagnosis. Int. J. Cancer. 37: 21-25. arrests cells in mitosis without depolymerizing tubulin. Bioehem. Biophys. Res. 1986. Commun.. 22S: 467-473, 1996. 147. Kvale, G.. Heuch. I., and Nüssen,S. Reproductive factors and cancers of the breast 161. Mukhopadhyay. T.. and Roth. J. A. Induction of apoptosis in human lung cancer and genital organs. Are the different cancer sites similarly affected ? Cancer Detect. cells after wild-type p53 activation by methoxyestradiol. Oncogene. 14: 379-384, Prevent.. /5: 369-379. 1991. 1997. 148. Sherman. B. M.. and Korenman. S. G. Inadequate corpus lulcum function: a 162. Loitering. M-L.. de Kock. M., Viljoen. T. C., Grobler, C. J. S., and Seegers, J. C. pathophysiological interpretation of human breast cancer epidemiology. Cancer I7ß-Estradiol metabolites affect some regulators of the MCF-7 cell cycle. Cancer (Phila.). 33: 1306-1311. 1974. Lett., 110: 181-186, 1996. 149. Horwitz, K. B., Wei, L. U Sedlacek. S. M.. and d'Arville, C. N. Progestin action 163. Seegers. J. C.. Loitering. M-L.. and De Kock. M. The endogenous metabolites and receptor structure in human breast cancer: a review. Recent Prog. •¿y-linolenicacid(GLAl and 2-methoxyestradiol (2ME) inhibit cell cycle progression Hormone Res., 41: 249-316, 1985. and cause apoptosis induction selectively in tumor cells. Proc. Am. Assoc. Cancer 150. Mauvais-Jan is. P.. Kuttenn. F.. and Gompel. A. Antiestrogen action of progesterone Res., 38: 627. 1997. in breast tissue. Hormone Res.. 28: 212-218. 1987. 164. Markaverich. B. M.. and Clark. J. H. Two binding sites for estradici in rat uterine 151. Key, T. J. A., and Pike. M. C. The role of estrogens and progestagens in the nuclei: relationship to uterotropic response. Endocrinology. 105: 1458-1462. 1979. epidemiology and prevention of breast cancer. Eur. J. Clin. Oncol.. 24: 29-43. 165. Markaverich. B. M.. Williams. M.. Unchurch. S.. and Clark, J. H. Heterogeneity of 1988. nuclear estrogen-binding sites in the rat uterus: a simple method for the quanlitation of 152. Bradlow. H. L., Hershcopf. R. J., and Fishman, J. Oestradiol 16a-hydroxylase: a risk type I and type 11sites by |%H|estradiol exchange. Endcx-rinology. 109: 62-69. 1987. marker for breast cancer. Cancer Surv.. 5: 573-583, 1986. 166. Markaverich. B. M.. Gregory. R. R.. Alejandro. M. A.. Clark. J. H.. Johnson, G. A., 153. Fishman, J., Osbome, M. P.. and Telang. N. T. The role of estrogen in mammary and Middledilch. B. S. Methyl /7-hydroxyphenyllactale: an inhibitor of cell prolif carcinogenesis. Ann. NY Acad. Sci., 768: 91-100. 1995. eration and an endogenous for nuclear type II binding sites. J. Biol. Chem.. 154. Kabat. G. C.. Chang. C. J.. Sparano, J. A.. Sepkovic. D. W.. Hu. X-P.. Khalil. A.. 263: 7203-7210, 1988. Rosenblatt. R.. and Bradlow, H. L. Urinary estrogen metabolites and breast cancer: 167. Xu, X., and Thomas. M. L. Biophasic actions of estrogen on colon cancer cell a case-control study. Cancer Epidemiol. Biomarkers Prevent.. 6: 505-509. 1997. growth: possible mediation bv high- and low-affinity binding sites. Endocrine. 3: 155. Bradlow, H. L.. Telang. N. T.. Sepkovic. D. W.. and Osborne. M. P. 2-Hy- 661-665, 1995. droxyestrone: the "good" estrogen. J. Endocrino!., 750: S259-S265, 1996. 168. Fuqua, A., Fitzgerald, S. D., Chamness. G. C.. Tandon, A. K.. McDonnell. D. P., 156. Li. J. J.. Li, S. A.. Oberley, T. D.. and Parsons. J. A. Carcinogenic activities of Nawaz. Z.. O'Malley. B. W.. and McGuirc. W. L. Variant human breast tumor various steroidal and nonsteroidal estrogens in the hamster kidney: relation to estrogen receptor with constitutive Iranscriptional activity. Cancer Res.. 5/: 105- hormonal activity and cell proliferation. Cancer Res.. 55: 4347-4351. 1995. 109. 1991. 157. Lemon. H. M., Heidel. J. W.. and Rodriguez-Sierra. J. F. Increased catechol estrogen 169. Pfeffer, U., Fecarotta. E., and Vidali. G. Coexpression of multiple estrogen receptor metabolism as a risk factor for nonfamilial breast cancer. Cancer (Phila.). 69: variant messenger RNAs in normal and neoplastic breast tissues and MCF-7 cells. 457-465, 1992. Cancer Res.. 55: 2158-2165. 1995. 158. Adlercreutz, H.. Gorbach. S. L.. Goldin. B. R.. Woods. M. N., Dwyer, J. T., and 170. Kuiper, G. G., Enmark, E., Pelto-Huikko. M.. Nilsson. S.. and Gustafsson, J-A. Hämäläinen.E.Estrogen metabolism and excretion in Oriental and Caucasian Cloning of a novel estrogen receptor expressed in rat prostate and ovary. Prix:. Nail. women. J. Nati. Cancer Inst.. X6: 1076-1082, 1994. Acad. Sci. USA. 93: 5925-5930. 1996. 159. Adlercreutz. H.. Gorbach. S. L.. Goldin, B. R.. Woods. M. N.. Dwyer. J. T.. 171. Laudet. V.. Hanni, C., Coll. J.. Calzellis. F.. and Stehelin. D. Evolution of the Höckerstedt, K.. Wähälä,K..Hase. T.. Hämäläinen.E.,and Fotsis. T. Diet and nuclear receptor gene superfamily. EMBO J.. //: 1003-1012. 1992. urinary estrogen profile in various populations. A preliminary report. Polycyclic 172. Soonljens, C. D.. Rafter. J. J.. and Gustafsson. J-À.Ligands for orphan receptors? J. Aromatic Compounds. 6: 261-273. 1994. Endocrinol.. ISO: S241-S257. 1996.

2277

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research. Is 2-Methoxyestradiol an Endogenous Estrogen Metabolite That Inhibits Mammary Carcinogenesis?

Bao Ting Zhu and Allan H. Conney

Cancer Res 1998;58:2269-2277.

Updated version Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/58/11/2269

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/58/11/2269. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1998 American Association for Cancer Research.