Fernand Labrie, Van Luu-The, Claude Labrie, Alain Bélanger, Jacques Simard, Sheng-Xiang Lin, and Georges Pelletier


Molecular Endocrinology and Oncology Research Center, Laval University Medical Center (Centre Hospitalier de l’Université Laval) and Laval University, Quebec City, Quebec G1V 4G2, Canada.


Abbreviations: ADT-G, Androsterone glucuronide; AR, androgen receptor(s); DHEA, dehydroepiandrosterone; DHEA-S, DHEA sulfate; DHT, dihydrotestosterone; 3α-diol-G, androstane-3α,17β-diol glucuronide; 3β-diol-G, androstane-3β,17β-diol glucuronide; 5-diol, androst-5ene-3β,17β-diol; 4-dione, androstenedione; DMBA, dimethylbenz(a)anthracene; E1, estrone; E2,17β-estradiol; ERT, estrogen replacement therapy; HRT, hormone replacement therapy; HSD, hydroxysteroid dehydrogenase; MPA, medroxyprogesterone acetate; PRAP, prolactin receptor-associated protein.


Address all correspondence and requests for reprints to: Prof. Fernand Labrie, Molecular Endocrinology and Oncology Research Center, Laval University Medical Center (Centre Hospitalier de l'Université Laval), 2705 Laurier Boulevard, Quebec City, Quebec G1V 4G2, Canada. E-mail: femand.labrie@crchul.ulaval.ca.


Abstract

Serum androgens as well as their precursors and metabolites decrease from the age of 30-40 yr in women, thus suggesting that a more physiological hormone replacement therapy at menopause should contain an androgenic compound. It is important to consider, however, that most of the androgens in women, especially after menopause, are synthesized in peripheral intracrine tissues from the inactive precursors dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEA-S) of adrenal origin. Much progress in this new area of endocrine physiology called intracrinology has followed the cloning and characterization of most of the enzymes responsible for the transformation of DHEA and DHEA-S into androgens and estrogens in peripheral target tissues, where the locally produced sex steroids are exerting their action in the same cells in which their synthesis takes place without significant diffusion into the circulation, thus seriously limiting the interpretation of serum levels of active sex steroids. The sex steroids made in peripheral tissues are then inactivated locally into more water-soluble compounds that diffuse into the general circulation where they can be measured. In a series of animal models, androgens and DHEA have been found to inhibit breast cancer development and growth and to stimulate bone formation. In clinical studies, DHEA has been found to increase bone mineral density and to stimulate vaginal maturation without affecting the endometrium, while improving well-being and libido with no significant side effects. The advantage of DHEA over other androgenic compounds is that DHEA, at physiological doses, is converted into androgens and/or estrogens only in the specific intracrine target tissues that possess the appropriate physiological enzymatic machinery, thus limiting the action of the sex steroids to those tissues possessing the tissue-specific profile of expression of the genes responsible for their formation, while leaving the other tissues unaffected and thus minimizing the potential side effects observed with androgens or estrogens administered systemically. (Endocrine Reviews 24: 152-182, 2003)



I. Androgens and Their Role in Women


A. Introduction

The most widely recognized fact about menopause is that it is accompanied by a rapid arrest of estrogen secretion by the ovaries. The cessation of ovarian estrogen secretion is illustrated by the marked decline in circulating 17β-estradiol (E2) levels. This easily measurable change in circulating E2, coupled with the demonstrated benefits of estrogens on menopausal symptoms and bone resorption (1), has concentrated almost all of the efforts of hormone replacement therapy (HRT) on various forms of estrogens as well as combinations of estrogen and progestin to avoid the potentially harmful stimulatory effects of estrogens used alone on the endometrium, which can result in endometrial hyperplasia and cancer. It should be mentioned, however, that although progestins are well recognized to protect the endometrium, preclinical (2-4) and clinical (5-7) data strongly suggest that they have a negative impact on breast cancer. The recent data of the Women's Health Initiative Study show that the combination of Premarin and Provera (Prempro) causes a 26% increase in the risk of breast cancer at 5.2 yr of follow-up, thus seriously questioning the use of a progestin as part of HRT in postmenopausal women (8).


Despite the well known beneficial effects of estrogen therapy on menopausal symptoms (9-11) and their role in reducing bone loss and possibly coronary heart disease (12-17), compliance is low. The majority of women decide not to take estrogens or stop treatment early because of the fear of breast and uterine cancer (11) and of symptoms associated with this therapy, namely uterine bleeding, breast tenderness, and fluid retention.


The almost exclusive focus on the role of ovarian estrogens at menopause has removed the attention from the progressive and dramatic fall in circulating dehydroepiandrosterone (DHEA), which starts early at the age of 30-40 yr (18-23). Because DHEA is transformed into both androgens and estrogens in peripheral tissues, such a fall in the serum concentration of the steroid precursors DHEA and DHEA sulfate (DHEA-S) explains why postmenopausal women, as discussed later, are not only lacking estrogens but are also deprived from androgens. Moreover, women taking contraceptives or estrogen replacement therapy (ERT) have reduced ovarian androgen secretion attributable to inhibition of gonadotropin secretion, as well as reduced androgen bioavailability attibutable to increased SHBG levels (24).


B. Decrease of serum DHEA, androgens, and their metabolites with age

Until recently, because of assay difficulties, only a limited number of circulating adrenal and gonadal steroids have been measured during advancing age, thus limiting the evaluation of the relative role of different sources of sex steroids. This analysis is of special importance in postmenopausal women in whom the sex steroids of adrenal origin gain particular importance after the arrest of estrogen secretion by the ovaries at menopause (25). It is important to recall that in the 50to 60-yr-old age group, serum DHEA has already decreased by 70%, compared with the 20to 30-yr-old peak values (Ref. 23; Fig. 1). It is thus quite remarkable that most of the important decline in circulating DHEA, DHEA-S, androst-5-ene-3β,17β-diol (5-diol), 5-diol-G, androstenedione (4-dione), and the conjugated metabolites of androgens, namely androsterone glucuronide (ADT-G), androstane3a,17β-diol glucuronide (3α-diol-G), and androstane3β,17β-diol glucuronide (3β-diol-G), occurs between the age ranges of 20-30 and 50-60 yr, whereas relatively smaller changes occur after the age of 60 yr (23). It is important to realize, as illustrated in Fig. 2, not only that serum DHEA and DHEA-S decrease by 50% between the ages of 21 and 40 yr but also that a similar decrease is observed for serum testosterone (26). Such data could well suggest that HRT with androgens should start early at menopause to compensate for this early fall in the secretion of androgen precursors by the adrenals and the parallel decrease in serum testosterone.




Figure 1. Effect of age (20 -30 to 70 -80 yr old) on serum concentration of DHEA (A), DHEA-S (B), DHEA-fatty acid esters (DHEA-FA; C), and 5-diol (D) in women. A marked decline is shown in serum concentrations of adrenal C19 sex steroid precursors and conjugated androgen metabolites during aging (26). [Reproduced with permission from F. Labrie et al.: J Clin Endocrinol Metab 82:2396-2402, 1997 (23). © The Endocrine Society.]



Figure 2. Illustration of the 50% parallel decrease in serum DHEA, DHEA-S, and testosterone between the ages of 21 and 40 yr in normal women (26).


Using the serum concentrations of ADT-G, 3α-diol-G, and 3β-diol-G as estimates of total androgens, the average sum of the serum concentrations of these conjugated metabolites of dihydrotestosterone (DHT) are 37.5, 8.47, and 30.2 nM in men compared with 32.5, 4.28, and 17.3 nM in women (23). The average serum concentrations of ADT-G, 3α-diol-G, and 3β-diol-G, measured in women between the ages 20 and 80 yr are thus 86.6% (ADT-G), 50.5% (3α-diol-G), and 57.2% (3β-diol-G), compared with those found in men of the same age (20-80 yr; Table 1; Ref. 23). Although the metabolic clearance rates of the three main androgen metabolites are likely to show differences between men and women, an estimate of the relative amount of total androgens in women and men calculated on the basis of the sum of the serum concentrations of these three metabolites suggests that total androgen production in women is more than two thirds, or 71%, of that observed in men (23, 27). Such an approach is based on the knowledge that active androgens are inactivated to glucuronide derivatives before their diffusion from the intracellular compartment into the circulation where they can be measured as ADT-G, 3α-diol-G, and 3β-diol-G.


Table 1. Comparison of serum androgen metabolites (20 -80 yr of age; nM)α



Such data showing the presence of relatively high levels of androgens in normal women strongly suggest that the androgens play a major physiological role in women. The 44.5% fall that occurs in serum DHEA from 20 -30 yr of age to the age of 40 -50 yr in women could well explain the bone loss that precedes menopause (27-30). Age-related bone loss has been reported to begin in the fourth decade, and changes in bone turnover have been found well before menopause (28 30). In agreement with these findings, bone density was lower at all sites examined in women classified as perimenopausal compared with premenopausal (31). In fact, the changes in precursor androgen secretion by the adrenals precede by 10 -20 yr the detectable decrease in ovarian steroidogenesis that occurs abruptly at menopause (23). In fact, serum FSH increases in premenopausal women even before serum E2 shows a decline (32).


After the recognition that such a large proportion of androgens and estrogens in men and women originate from DHEA and DHEA-S of adrenal origin (25), we have studied the serum concentration of a large series of androgens and estrogens as well as their metabolites after percutaneous administration of DHEA in 60- to 70-yr-old men and women (27). We then observed that changes in serum DHEA within the physiological range of young adult men and women led only to small or nonsignificant changes in serum testosterone, DHT, or E2, whereas, on the other hand, the concentration of the conjugated metabolites of DHT were markedly increased (27). Such data clearly indicate the poor value of measurements of serum androgens and estrogens as parameters of total androgenic and estrogenic activities in men and women.


As well demonstrated in a long series of preclinical studies, supplementation with physiological amounts of exogenous DHEA permits the biosynthesis of androgens (essentially testosterone and DHT) and estrogens only in the target tissues that contain the specific steroidogenic enzymes (25, 33). The widespread tissue distribution of steroidogenic enzymes is illustrated in Table 2 (34). In fact, in 22 peripheral tissues of the monkey, steroid sulfatase, 3β-hydroxysteroid dehydrogenase (HSD), androgenic 17β-HSD, estrogenic 17β-HSD, aromatase, and 5α-reductase are all present in 114 of 132 (86%) possible sites. Genomic studies are in progress to determine the identity of all families of steroidogenic enzymes in the various peripheral target tissues.


Table 2. . Distribution of intracrine steroidogenic enzymes in the monkey



The active androgens and estrogens synthesized in peripheral target tissues exert their activity in the cells of origin, and very little diffusion of the active sex steroids occurs, thus resulting in very low levels in the circulation. In fact, the most striking effects of DHEA administration are seen on the circulating levels of the glucuronide derivatives of the metabolites of DHT, namely ADT-G, 3α-diol-G, and 3β-diol-G, these metabolites being produced locally in the peripheral intracrine tissues that possess the appropriate steroidogenic enzymes to synthesize DHT from the adrenal precursors DHEA and DHEA-S. These peripheral target tissues also contain the steroid-inactivating enzymes required to metabolize DHT into inactive and more water-soluble conjugates, especially glucuronide derivatives (25, 35). Such local biosynthesis and action of androgens in target tissues eliminates the exposure of other tissues to androgens and thus minimizes the risks of undesirable masculinizing or other androgen-related side effects. The same applies to estrogens, although we feel that a reliable parameter of total estrogen secretion (comparable to the glucuronides for androgens) has not yet been identified.


C. Androgens and bone physiology


1. Role of androgens and estrogens in bone physiology

A predominant role of androgens in bone physiology has already been suggested (36). In fact, both testosterone and DHT increased the transcription of α (I) procollagen mRNA in osteoblast-like osteosarcoma cells (37). Treatment with DHT has also been shown to stimulate endochondral bone development in the orchiectomized rat (38). Androgens stimulate osteoblast differentiation, these cells being known to contain androgen receptors (AR; Refs. 39 -41). Moreover, bone mineral density measured in the lumbar spine, femoral trochanter, and total body was increased more by estrogen plus testosterone implants than by E2 alone over a 24-month treatment period in postmenopausal women (42). In agreement with these data, biomarkers of bone formation were increased compared with estrogen alone when methyltestosterone was added to estrogen (43).


The essential role of androgens in bone mineralization is illustrated by the reduced bone mineral density in patients with the androgen insensitivity syndrome (44 -46). In such patients having an inactive AR, estrogens are unable to increase bone mineral density (44, 45). Thus, at doses of estrogen able to restore bone mineral density in hypogonadal women, estrogens could not exert a similar effect in patients with androgen insensitivity. Such data suggest that both estrogens and androgens are required to acquire normal bone mineral density. In fact, a correlation has been found between androgens and bone mineral density in premeno pausal women (31, 47).


In established osteoporosis, anabolic steroids have been reported to help prevent bone loss (48). Moreover, androgen therapy, as observed with nandrolone decanoate, increases vertebral bone mineral density in postmenopausal women (49). Similarly, sc E2 and testosterone implants have been found to be more efficient than oral estrogen in preventing osteoporosis in postmenopausal women (50). Although the difference has been attributed to the different routes of administration of the estrogen, the cause of the difference could well be the action of testosterone. Studies have convincingly shown that androgen plus estrogen was more efficient than estrogen in improving bone mineral density in postmenopausal women (42, 43, 50, 52-55).


Although androgens are gaining increasing support because of their unique actions in postmenopausal women, virilizing effects are observed with the use of supraphysiological doses of testosterone (56, 57). The availability of a compound such as DHEA, an inactive precursor that is transformed into active androgens only in specific target tissues, would be an important advantage over androgens exerting systemic effects in all tissues possessing AR.


D. Other roles of androgens in women


1. General

It is likely that the androgens produced from DHEA have other beneficial effects in postmenopausal women. The detailed benefits of androgens added to ERT or HRT have been described on general well-being, energy, mood, and general quality of life (58, 59). Improvements in the major psychological and psychosomatic symptoms, namely irritability, nervousness, memory, and insomnia, have been reported after addition of androgens to ERT (60). In addition, androgenic compounds have been found to be beneficial for the treatment of the mastalgia frequently caused by HRT (61). In fact, ERT may result in severe breast pain that may lead to discontinuation of therapy.


2. Libido and sexual satisfaction.

Loss of libido and/or sexual satisfaction are common in early postmenopause. The addition of androgens to HRT is known to have beneficial effects on these problems (42, 53, 57, 58, 6264). Moreover, a series of studies have shown the beneficial effects of androgens on libido in postmenopausal women (42,6567). In women who have undergone oophorectomy and hysterectomy, transdermal testosterone improves sexual function and psychological well-being (68). Similar findings have been observed with DHEA administered to women with adrenal insufficiency, this steroid being the most important precursor of androgens in postmenopausal women (69). On the other hand, mood and fatigue were significantly improved after DHEA replacement therapy in Addison's disease (70).


3. Hot flashes

The addition of androgens has been found to be effective in relieving hot flashes in women who had unsatisfactory results with estrogen alone (71). Androgen therapy is also successful in reducing hot flashes in hypogonadal men (72). In agreement with its transformation into androgens (27), DHEA has been found useful in reducing hot flashes (73, 74). In fact, marked improvements in the vasomotor symptoms were observed in early postmenopausal women who received 50 mg DHEA orally daily from an average score of 18.4 before treatment to a score of 4.5 at 6 months (74).


4. Cardiovascular function and lipids

There is also evidence that androgens may improve endothelium-dependent and -independent vasodilation in postmenopausal women (75). In fact, parenteral testosterone therapy improved brachial artery vasodilatation in postmenopausal women using longterm estrogen therapy. It is also of great interest that the addition of parenteral testosterone does not negate the favorable effects of estrogen on low-density lipoprotein cholesterol (76).



II. DHEA Is Predominantly Converted into Androgens in Women


A. Intracrinology

Man is unique, with some other primates, in having adrenals that secrete large amounts of the precursor steroids DHEA and DHEA-S, which are converted into 4-dione and then into potent androgens and/or estrogens in peripheral tissues (Refs. 25, 77, and 78; Fig. 3). Adrenal secretion of DHEA and DHEA-S increases during adrenarche in children at the age of 6 8 yr, and maximal values of circulating DHEA-S are reached between the ages of 20 and 30 yr.



Figure 3. Schematic representation of the role of ovarian and adrenal sources of sex steroids in premenopausal women. After menopause, the secretion of estradiol by the ovaries ceases, and almost 100% of sex steroids are made locally in peripheral target intracrine tissues.


Thereafter, serum DHEA and DHEA-S levels decrease markedly (Fig. 1; Refs. 18 and 20 -22). In fact, as mentioned earlier, at 70 yr of age, serum DHEA-S levels are decreased to approximately 20% of their peak values, whereas they can decrease by 95% by the age of 85-90 yr (22). The 70 -95% reduction in the formation of DHEA and DHEA-S by the adrenals during aging results in a dramatic reduction in the formation of androgens and estrogens in peripheral target tissues. Such a marked decrease in the formation of sex steroids in peripheral tissues could well be involved in the pathogenesis of diseases associated with aging.


It is thus remarkable that man, in addition to possessing very sophisticated endocrine and paracrine systems, has largely vested in sex steroid formation in peripheral tissues (25,27, 77,78). In fact, although the ovaries and testes are the exclusive sources of androgens and estrogens in lower mammals, the situation is very different in man and higher primates, where active sex steroids are in large part or wholly synthesized locally in peripheral tissues, thus providing target tissues with controls that adjust the formation and metabolism of sex steroids to local requirements. This situation is well illustrated in women by the absence of significant difference in the intracellular levels of E2 in breast cancer tissue between premenopausal and postmenopausal women (79). Because the postmenopausal ovary does not secrete estrogens, intratumoral E2 is necessarily made from adrenal precursor steroids (25).


Transformation of the adrenal precursor steroids DHEA-S and DHEA into androgens and/or estrogens in peripheral target tissues depends upon the level of expression of the various steroidogenic and metabolizing enzymes in each of these tissues. This sector of endocrinology that focuses on the intracellular hormone formation and action has been called intracrinology (Refs. 25 and 78; Fig. 4). This situation of a high secretion rate of adrenal precursor sex steroids in men and women is thus completely different from all animal models used in the laboratory, namely rats, mice, guinea pigs, and all others (except monkeys) in which the secretion of sex steroids takes place exclusively in the gonads (77, 80). A major problem that is at least partially responsible for the delayed progress in the recognition of the formation of a major proportion of sex steroids in peripheral tissues or intracrinology is the fact that the animal models usually used in the laboratory do not secrete significant amounts of adrenal precursor sex steroids, thus focusing all attention on the testes and ovaries as the exclusive sources of androgens and estrogens. The term intracrinology was thus coined (78) to describe the synthesis of active steroids in peripheral target tissues in which the action is exerted in the same cells where synthesis takes place without release of the active steroids in the extracellular space and general circulation (25).



Figure 4. Schematic representation of endocrine, paracrine, autocrine, and intracrine secretion. Classically, endocrine activity includes the hormones secreted in specialized glands, called endocrine glands, for release into the general circulation and transport to distant target cells. On the other hand, hormones released from one cell can influence neighboring cells (paracrine activity) or can exert a positive or negative action on the cell of origin (autocrine activity). Intracrine activity describes the formation of active hormones that exert their action in the same cells in which synthesis took place without release into the pericellular compartment. [Reprinted with permission from F. Labrie: Mol Cell Endocrinol 78:C113-C118, 1991 (25).]


Proof of the role of estrogen formation in peripheral intracrine tissues is particularly well illustrated in women by the important benefits on breast cancer observed in postmenopausal women treated by a series of aromatase inhibitors (81). Most convincingly, because the postmenopausal ovaries do not secrete estrogens, the recent observation that administration of the antiestrogen raloxifene for only 3 yr in postmenopausal women led to a 76% decrease in the incidence of breast cancer (82) is a clear demonstration of the role of extraovarian estrogens in the development and growth of breast cancer.


B. Structure of the human steroidogenic enzymes

As mentioned above, transformation of DHEA and DHEA-S into active androgens and/or estrogens in peripheral target tissues depends on the level of expression of the various steroidogenic and metabolizing enzymes in each cell type. Elucidation of the structure of most of the tissuespecific genes that encode the steroidogenic enzymes responsible for the transformation of DHEA and DHEA-S into androgens and/or estrogens has permitted rapid progress in this area (Refs. 33 and 83-86; Fig. 5). The major importance of DHEA and DHEA-S is illustrated by the finding that approximately 50% of total androgens in the prostate of adult men derive from these adrenal precursor steroids (77,87,88). Our best estimate of the intracrine formation of estrogens in peripheral tissues in women is in the order of 75% before menopause and close to 100% after menopause (25). Although testosterone of ovarian and adrenal origin can act directly in peripheral tissues, its transformation into estrogens requires the action of the peripheral or intracrine steroidogenic enzymes, especially aromatase (89).



Figure 5. Human steroidogenic enzymes in peripheral intracrine tissues.


Because the molecular structure of most of the key non-P-450-dependent enzymes required for sex steroid formation had not been elucidated, and knowing that local formation of sex steroids is most likely to play a major role in the control of activity of both normal and tumoral hormone-sensitive tissues, an important proportion of our research program and that of other groups has been devoted to this exciting and therapeutically promising area (33, 35, 84, 90 -92). The synthesis from DHEA of the most potent natural androgen, DHT, and of the most potent natural estrogen, E2, involves several enzymatic activities, namely 3β-HSD, 17β-HSD, 5α-reductase, and/or aromatase (Fig. 5).


1. Human 3β-HSD isoenzymes and their genes

Despite its essential role in the biosynthesis of all classes of hormonal steroids, the structure of the 3β-HSD/Δ54-isomerase gene family, hereafter called 3β-HSD, was only elucidated relatively recently (84, 93-96). The membrane-bound enzyme 3β-HSD catalyzes an essential step in the transformation of all 5-pregnen-3β-ol and 5-androsten-3β-ol steroids into the corresponding Δ4-3-keto-steroids, namely progesterone as well as the precursors of all androgens, estrogens, glucocorticoids, and mineralocorticoids.


Experiments performed using microsomes and purified enzymes show that 3β-HSD can catalyze the interconversion of 3β-hydroxy- and 3-keto-5α-androstane steroids (97). On the other hand, experiments performed under more physiological conditions (i.e., in intact transfected cells in culture without added cofactor) indicate that 3β-HSD catalyzes almost exclusively the oxidation of 3β-hydroxyinto 3-keto5α-androstane steroids (98). The reverse reductive reaction is catalyzed by another enzyme, namely 3(α→β)-hydroxysteroid epimerase [3(α→β)-HSE; Refs. 98 and 99] and type 7 17β-HSD (our unpublished data).


3β-HSD is found not only in the classical steroidogenic tissues (placenta, adrenal cortex, ovary, and testis) but also in several peripheral tissues, including the skin, adipose tissue, breast, lung, endometrium, prostate, liver, kidney, epididymis, and brain (34,84, 91, 100), thus catalyzing the first step in the intracrine transformation of DHEA into 4-dione, the precursor of both androgens and estrogens. The existence of multiple members of the 3β-HSD gene family offers the unique possibility of tissueand/or cell-specific expression of this enzymatic activity.


After purification of 3β-HSD from human placenta and development of antibodies against the enzyme in rabbits (101), we have isolated and characterized a first 3β-HSD cDNA type (93) and its corresponding gene (94). The second 3β-HSD cDNA type, which corresponds to the almost exclusive mRNA species expressed in the adrenals and gonads, was chronologically designated human type 2 3β-HSD (95). The structure of the corresponding human type 2 3β-HSD gene has also been elucidated (96). The human 3β-HSD genes corresponding to human cDNAs type 1 and 2 contain four exons and three introns within a total length of 7.7-7.8 kb. These genes were assigned by in situ hybridization to the p13.1 region of chromosome 1 and are closely linked to D1S514 located at 1-2 cM of the centromeric marker D1Z5 (102).


We have observed that mutations in the type 2 3β-HSD gene are responsible for classic 3β-HSD deficiency, a form of congenital adrenal hyperplasia that impairs steroidogenesis in both the adrenals and gonads (103-105). However, the absence of mutations in the type 1 gene provided the longawaited molecular explanation for the persistence of peripheral steroidogenesis in these 3β-HSD type 2-deficient patients, thus demonstrating the importance of peripheral sex steroid formation or intracrinology.


2. Human 17β-HSDs.

The 17β-HSDs are responsible for the formation and inactivation of all active androgens and estrogens. As discussed above for 3β-HSD, until recently, 17β-HSDs as well as almost all other dehydrogenases were considered to be reversible enzymes that catalyze the interconversion of substrates and products, mainly because the enzymatic activity was first characterized using tissue homogenates, subfractions, or purified proteins with added oxidized (NAD+, NADP+) or reduced (NADH, NADPH) cofactors. These exogenous cofactors drive the reaction in the oxidative or reductive direction depending on their oxidized or reduced state, respectively. However, using a more physiologically relevant method of enzymatic activity analysis, namely intact transfected cells in culture without the addition of exogenous cofactors, the transfected enzyme catalyzes the reaction in a unidirectional manner (85, 98, 99, 106, 107). These findings agree with the isolation of multiple types of 17β-HSDs in which approximately half catalyze the reductive reaction (types 1, 3, 5, and 7) and half catalyze the oxidative reaction (types 2, 4, 6, and 8).


a. Type 1 17β-HSD

The molecular structure of the human type 117β-HSD gene and mRNA, which encode a predicted protein of 327 amino acids, was the first of the 17β-HSDs to be elucidated (Refs. 108-111; Fig. 6). This enzyme is a member of the short-chain alcohol dehydrogenase superfamily. The type 1 17β-HSD enzyme is a cytosolic protein that exists in a homodimeric form that catalyzes predominantly the interconversion of estrone (E1) to E2 using NADP(H) as cofactor (112, 113).




Figure 6. Structure of the genes and mRNAs encoding human types 1-5, 7, and 8 17β-HSD and the corresponding proteins. aa, Amino acids. [Reproduced by permission of the Society for Endocrinology (33).]


To perform the structure-function analysis of type 1 17β-HSD, the protein was rapidly purified from the placenta, thus yielding a highly active preparation (113, 114). The protein was also overproduced in baculovirus, and crystals were obtained (115). This crystallization led to the elucidation of the three-dimensional structure of human type 1 17β-HSD (116), thus achieving the first x-ray structure determination of a mammalian steroidogenic enzyme. The structure of type 1 17β-HSD from human placenta was determined at 2.2-A resolution by a combination of isomorphous replacement (with a single mercury derivative) and molecular replacement techniques.


b. Type 2 17β-HSD

The structure of a cDNA encoding a second type of 17β-HSD cDNA was then reported (117,118). This cDNA encodes a predicted protein of 387 amino acids with a molecular weight of 42,782 (Fig. 6). This protein is most likely associated with the membranes of the endoplasmic reticulum. The enzyme catalyzes the conversion of E2 to E1, testosterone to 4-dione, and 5-diol to DHEA. This enzyme, chronologically designated type 2 17β-HSD, is also a member of the short-chain alcohol dehydrogenase superfamily, but it shares only about 20% sequence identity with the type 1 17β-HSD cytoplasmic enzyme (109). This enzyme uses NAD(H) as a cofactor (117) and is less specific than type 1 17β-HSD, both estrogens and androgens acting as substrates. This enzyme inactivates the estrogens and androgens made after the reductive action of type 2,3, and 5 17β-HSDs.


c. Type 3 17β-HSD

A third type of human 17β-HSD cDNA encoding a predicted protein of 310 amino acids with a molecular weight of 34,513 was then characterized (119). Type 3 17β-HSD, a microsomal isozyme, using NADP(H) as a cofactor, is expressed predominantly in the testes, where it synthesizes testosterone from 4-dione. This enzyme, which shares 23% sequence identity with the two other 17β-HSD enzymes, is the site of the mutations responsible for male pseudohermaphroditism resulting from 17β-HSD deficiency (119).


d. Type 4 17β-HSD

Human type 4 17β-HSD is a 736-aminoacid protein of molecular mass 80 kDa that can transform E2 to E1 and 5-diol to DHEA (120, 121). The human type 4 17β-HSD mRNA is expressed in virtually all human tissues examined by Northern blot, including the liver, heart, prostate, testis, lung, skeletal muscle, kidney, pancreas, thymus, ovary, intestine, placenta, and several human breast cancer cell lines. This enzyme possibly plays a role in the inactivation of estrogens in a large series of peripheral tissues, although its activity is low and its importance in steroid formation in the human remains to be established. Indeed, mutations in type 4 17β-HSD gene lead to a fatal form of Zellweger syndrome (122).


e. Type 5 17β-HSD

Although type 3 17β-HSD synthesizes testosterone from 4-dione in the Leydig cells of the testes, thus providing approximately 50% of the total amount of androgens in men, the same enzymatic reaction is catalyzed in the peripheral target tissues in both men and women as well as in the ovary by a different enzyme, namely type 5 17β-HSD (106). This enzyme is highly homologous with types 1 and 3 3α-HSD as well as 20α-HSD (106) and thus belongs to the aldo-keto reductase family.


In the postmenopausal ovary, hypertrophied stromal cells are localized mainly at the periphery and hilus (123). These stromal cells contain both 3β-HSD and type 5 17β-HSD, thus permitting the transformation of DHEA into 4-dione and then into testosterone. The amount of stromal hyperplasia in postmenopausal ovaries is correlated with the ovarian vein levels of 4-dione and testosterone (124). These hyperplastic stromal cells are thus responsible for the synthesis of 4-dione and testosterone in the postmenopausal ovary.


Type 5 17β-HSD is not only expressed in the ovary but is also present in a large series of peripheral tissues including the mammary gland. The epithelium lining the acini and ducts of the mammary gland is composed of two layers, an inner epithelial layer and an outer discontinuous layer of myoepithelial cells. By immunocytochemistry, 3β-HSD is seen in the epithelial cells of acini and ducts as well as in stromal fibroblasts (Fig. 7A). Immunostaining is also observed in the walls of blood vessels, including the endothelial cells. In the positive cells, the labeling is mainly cytoplasmic. No significant labeling could be detected in the myoepithelial cells. As shown in Fig. 7B, immunostaining for type 5 17β-HSD gives results almost superimposable to those obtained for 3β-HSD, the cytoplasmic labeling being observed in both epithelial and stromal cells and blood vessel walls (125). Studies performed at the electron microscopic level revealed that in sections stained for 3β-HSD or type 5 17β-HSD, labeling was not associated with any specific membranebound organelles in the different reactive cell types (126).



Figure 7. Human mammary gland immunostained for 3β-HSD (A) and type 5 17β-HSD (B). Staining can be observed in the secretory epithelial cells of acini (A). Stromal cells (arrows) and capillaries (arrowheads) are also labeled. Magnification, X430.


f. Type 6 17-HSD

Using a rat prostate cDNA obtained by expression cloning, Biswas and Russell (127) have isolated cDNA clones that metabolize 3α-diol. Among the many clones obtained, one type, named type 6 17β-HSD, catalyzes selectively the oxidation of 3α-diol to androsterone. The transformation of other C19-steroids, namely DHT to androstanedione and testosterone to 4-dione, also occurs but at an approximately 50- to 100-fold lower rate.


Type 6 17β-HSD shares 65% homology with rat type 1 retinol dehydrogenase and thus belongs to the retinol dehydrogenase family. The human counterpart has not yet been described, and its role remains to be established.


g. Type 717β-HSD

Type 7 17β-HSD was first cloned from a rat corpus luteum cDNA library and was identified as prolactin receptor-associated protein (PRAP; Ref. 128). With the use of expression cloning of a mouse mammary epithelial (HC11) cell cDNA library, a clone that shares 89% identity with rat PRAP and catalyzes selectively the transformation of E1 to E2 has been isolated (129). After transfection into HEK-293 cells, Nokelainen et al. (129) also found that rat PRAP catalyzes efficiently and selectively the transformation of E1 to E2, whereas the transformation of C19 steroids was much weaker.


Human type 7 17β-HSD cDNA is 1.5-kb long and encodes a protein of 37 kDa or 341 amino acids (130). With the use of RT-PCR, this enzyme is detected in the ovary, breast, placenta, testis, prostate, and liver. Comparison with other 17β-HSDs indicates that it shares less than 20% identity, a typical percentage for the other members of the 17β-HSD family. The human type 7 17β-HSD gene spans 21.8 kb and consists of nine exons and eight introns. The gene is assigned to human chromosome bands 10p11.2 (130). It is noteworthy that type 5 17β-HSD is also mapped to human chromosome 10 (bands 10p15→14). The importance of this enzyme remains to be established.


h. Type 8 17β-HSD

Type 8 17β-HSD is also known as the product of the Ke6 gene, which is found in the HLA region (131). This area is well known to contain genes encoding the human major histocompatibility complex. This complex is thought to be involved in polycystic kidney disease because aberrant gene expression has been found in two different models of polycystic kidney disease mice (132). Recently, Fomitcheva et al. (133) have found that the overproduced protein fused with GST catalyzes efficiently the transformation of E2 to E1. The transformation of testosterone to 4-dione is about 25% of that of E2 into E1. Using HEK-293 cells stably transfected with human type 8 17β-HSD, we have shown recently that this enzyme selectively converts E2 to E1, the transformation of E1 as well as of androgen substrates being negligible (134).


3. Human 5α-reductase isoenzymes

The enzyme 5α-reductase catalyzes the 5α-reduction of 4-dione, testosterone, and other 4-ene-3-keto-steroids to the corresponding 5α-dihydro-3keto-steroids. The best known role of this enzyme is the transformation of testosterone into DHT, the most potent androgen, which is responsible for the differentiation of the male external genitalia and prostate as well as virilization at puberty. The major impact of 5α-reductase in men, however, is its role in prostate cancer and benign prostatic hyperplasia. Two types of human steroid 5α-reductases, chronologically identified as type 1 and type 2, were isolated from human prostatic cDNA libraries (135, 136). The structure of the human type 1 5α-reductase gene was first elucidated (137). This gene is not responsible for 5α-reductase deficiency and is relatively insensitive to the inhibitor finasteride (136). Type 2 5α-reductase, on the other hand, is the isozyme responsible for male pseudohermaphroditism from 5α-reductase deficiency and is sensitive to finasteride (136, 138).


Considering the crucial role of type 2 5α-reductase, we have elucidated the structure of its corresponding gene (83). The type 2 5α-reductase gene contains five exons and four introns and shows splicing sites identical to those of the type 1 gene. Its coding region shares 57% homology with that of the type 1 5α-reductase gene. Type 1 5α-reductase is the predominant form expressed in human skin (139).


C. Women produce about two thirds of the androgens synthesized in men


1. Decline in serum androgen precursors and metabolites occurs well before menopause

To gain a better knowledge of the role of DHEA and DHEA-S transformation in both men and women, we have analyzed the serum levels of 18 conjugated C21and C19-steroids (23). The data obtained show a dramatic decline in the circulating levels of DHEA, DHEA-S, 5-diol, and 5-diol fatty acid esters between the ages of 20 and 80 yr (Fig. 1). As mentioned earlier, in the 50to 60-yr-old group, serum DHEA has already decreased by 70% from its 20to 30-yr-old peak values in women (Fig. 1). It should be added that between the ages of 21 and 40 yr, mean serum testosterone in normal women decreases from approximately 1.3 to 0.61 nM (Ref. 26; Fig. 2). A parallel decrease is observed for serum DHEA and DHEA-S, thus suggesting the role of DHEA in the progressive decline in serum testosterone between the ages of 21 and 40 yr in normal women.


The serum concentrations of the conjugated metabolites of DHT, namely ADT-G, 3α-diol-G, and 3β-diol-G, are the most reliable parameters of the total androgen pool in women, whereas serum testosterone is mostly a measure of direct secretion of testosterone by the ovaries and/or adrenals. In fact, although the vast majority of testosterone and DHT is synthesized in the peripheral tissues in women, only a small proportion, estimated at 10-15% of the intracellular content of these androgens, diffuses out of the intracellular compartment without prior metabolism and can be measured as active androgen in the circulation. This is because testosterone and DHT, instead of being almost quantitatively released in the circulation, are rapidly glucuronidated into ADT-G, 3α-diol-G, and 3β-diol-G (Fig. 8). Because the individual glucuronosyltransferases responsible for the inactivation of androgens in the human mammary gland have not yet been identified, the human prostate is used as an example of the types of glucuronosyltransferases involved (140, 141). These metabolites are much more water soluble than DHT and thus easily diffuse into the general circulation where they can be measured en route for their elimination mainly by the kidneys (Figs. 9 and 10). The serum concentration of the aboveindicated conjugated androgen metabolites decreases by 47.5-72.7% between the 20- to 30- and 70- to 80-yr age groups in women, thus suggesting a parallel decrease in the total androgen pool with age (23).



Figure 8. Enzymes involved in the peripheral metabolism or inactivation of androgens in peripheral tissues.



Figure 9. Distribution in women of the active androgens testosterone and DHT, the sex steroid precursor DHEA, and the main metabolites of androgens (ADT-G, 3α-diol-G, and 3β-diol-G) in the circulation, and in peripheral intracrine tissues. The height of the bars is proportional to the concentration of each steroid or its derivatives in individual compartments (336).



Figure 10. Schematic representation of the secretion of DHEA, DHEA-S, and 4-dione by the adrenals and E2,4-dione, and testosterone by the ovaries as well as the intracellular metabolism of these steroids in the peripheral intracrine tissues. Especially after menopause, the level of androgens active in peripheral tissues is best estimated by the serum concentration of the metabolites of DHT, namely ADT-G, 3α-diol-G, and 3β-diol-G.


As assessed by measurement of the circulating levels of these conjugated metabolites of DHT, it can be estimated that women produce approximately 71% or two thirds of the total androgens synthesized in men (Table 1); in women, most of these androgens originate from the transformation of DHEA and DHEA-S into testosterone and DHT in peripheral intracrine tissues. Such an estimate of the androgen pools in men and women based on the serum concentration of androgen metabolites can be influenced by possible differences in the metabolic clearance rates of these metabolites in men and women.


2. Plasma sex steroid levels are not a valid parameter of the intracellular situation in women

Proof that changes of the intracellular concentration of sex steroids cannot be estimated by the measurement of testosterone and E2 in the circulation has been obtained in a study performed in postmenopausal women (23). This study analyzed in detail the serum concentrations of the active androgens and estrogens, as well as a series of free and conjugated forms of their precursors and metabolites, after daily application for 2 wk of a 10-ml 20% DHEA solution on the skin to avoid first passage of DHEA through the liver.


After daily administration of a single dose of DHEA percutaneously, serum DHEA, DHEA-S, and DHEA fatty acid esters increased approximately 175%, 130%, and 250% above control, respectively (Fig. 11), whereas serum 4-dione and testosterone increased by about 100% and 50% over control, respectively (Fig. 12). In parallel with the changes in serum DHEA, DHEA-S, and DHEA fatty acids, the most important effects (Fig. 13) were seen on the glucuronidated metabolites of ADT, 3α-diol, and 3β-diol. In fact, treatment with DHEA caused an increase in serum ADT-G, 3α-diol-G, and 3βdiol-G of approximately 125% (Fig. 13A), 140% (Fig. 13B), and 120% (Fig. 13C), respectively. No significant effect was observed on serum E1, E2, or DHT.



Figure 11. Effect of daily percutaneous administration of a 10 ml 20% solution of DHEA in 50% ethanol-50% propylene glycol for 2 wk in 60- to 70-yr-old women on serum levels of DHEA (A), DHEA-S (B), and DHEA-fatty acid esters (C; Ref. 26).



Figure 12. Effect of daily percutaneous administration of 10 ml 20% solution of DHEA in 50% ethanol-50% propylene glycol for 2 wk in 60- to 70-yr-old women on serum levels of 4-dione (A) and testosterone (B; Ref. 26).



Figure 13. Effect of daily percutaneous administration of 10 ml 20% solution of DHEA in 50% ethanol-50% propylene glycol for 2 wk in 60- to 70-yr-old women on serum levels of ADT-G (A), 3α-diol-G (B), and 3β-diol-G (C; Ref. 26).


The present data show that elevations in serum DHEA within the physiological range found in young adult women led to only small or even no significant changes in serum testosterone, DHT, or E2, whereas, by contrast, the concentrations of the conjugated metabolites of DHT are markedly elevated, in parallel with the changes in serum DHEA, DHEA-S, and 5-diol. Such data obtained in normal postmenopausal women offer unique proof that the serum levels of androgens and estrogens are poor indicators of total androgenic and estrogenic activities in women. In fact, as mentioned earlier, serum testosterone and E2 reflect almost exclusively the contribution of the small and direct sex steroid secretion by the ovaries and/or adrenals.


The 50% increase in serum testosterone of approximately 0.8 nM (from 1.5-2.3 nM) observed in women during DHEA treatment corresponds to a much larger increase of approximately 20 nM in serum DHEA. These data are in agreement with the information obtained in men after medical or surgical castration in which the serum levels of testosterone decreased from 15 nM to about 1.5 nM after elimination of testicular androgens. Thus, after castration, the serum levels of testosterone in 60- to 70-yr-old men became comparable to those observed in intact postmenopausal women. The 1.5 nM serum testosterone remaining after castration in men originates essentially from adrenal DHEA (77, 87). The present data thus offer an independent measure of the amount of testosterone that diffuses into the circulation from the androgens synthesized from DHEA and DHEA-S in peripheral intracrine tissues (25).


In a recent study, daily oral administration of 50 mg DHEA had no significant effect on serum testosterone or DHT, whereas DHEA and ADT-G were increased to a similar extent (80 -90%; Ref. 142). In another study, predosing serum levels of DHEA-S in postmenopausal women were increased from 0.55 pg/ml to about 1.4 pg/ml (143) after daily oral administration of 25 mg DHEA for 6 months. Serum DHEA and testosterone levels, however, measured 23 h after the last administration of DHEA, were not changed significantly. Similarly, the 50-mg/d oral dose of DHEA was found to lead to serum androgen levels in the premenopausal range (144).


Our data obtained after percutaneous administration of DHEA in normal postmenopausal women offer the first direct analysis of the correlation between the serum levels of DHEA and DHEA-S with the serum concentration of active androgens and estrogens and their corresponding glucuronidated and sulfated metabolites. It can be concluded that measurements of serum testosterone and E2 mainly reflect ovarian and/or adrenal steroid secretion, whereas the major contribution of the adrenals is not accurately represented in the circulating levels of active sex steroids. The present data clearly demonstrate that DHEA and DHEA-S are converted in a series of intracrine tissues into the active androgens and/or estrogens that exert their biological effects at their site of synthesis. These steroids are then metabolized in the same cells into inactive glucuronidated and sulfated metabolites, which finally diffuse in the extracellular compartment and can be measured in the circulation. Measurement of the conjugated metabolites of androgens is the only approach that permits an accurate estimate of the total androgen pool in women. It is likely that a similar situation exists for estrogens, although a precise evaluation of the pharmacokinetics of estrogen metabolism and identification of their metabolites remains to be completed.


3. Contribution of the postmenopausal ovary to serum 4-dione and testosterone

It is well recognized that the postmenopausal ovary is a steroid-secreting gland (145,146). In fact, the postmenopausal ovary is well known to secrete testosterone, and most authors agree that it also secretes some 4-dione (147, 148). In fact, a correlation has been observed between the degree of ovarian stromal hyperplasia and the secretion of androgens by the ovary (124, 149). Moreover, lowering serum gonadotropins with a GnRH agonist has been shown to result in decreased serum androgen levels, thus indicating that the stromal cells of the ovary are under gonadotropin control (150,151). In agreement with these data, receptors for LH and FSH have been described in the ovarian stromal cells.


It should be mentioned that Couzinet et al. (152) have reported that the postmenopausal ovary does not contribute significantly to serum androgen levels. This observation is unique and, if confirmed, will bring even more emphasis on the importance of the adrenals in sex steroid physiology after menopause.


Despite the above-described limitations of the interpretation of serum levels of sex steroids, it is of interest to provide the best available estimate of the contribution of the ovaries and adrenals to the serum levels of 4-dione and testosterone. The majority of studies show declining levels of serum testosterone and 4-dione with age (149, 153-156). Testosterone concentration in the ovarian venous blood is 15 times higher than in peripheral blood (147). In fact, the production of testosterone by the ovary has been estimated to decrease from 250 to 180 μg/d after menopause (157).


As illustrated in Fig. 14A, although the ovaries and adrenals contribute about equally to the serum levels of 4-dione in premenopausal women (158, 159), the contribution of the ovaries decreases to about 20% after menopause (158, 159), despite a progressive fall in the contribution of the adrenals through transformation of declining amounts of DHEA into 4-dione, thus leading to lower total serum concentration of 4-dione after menopause. Similarly, the serum levels of testosterone in premenopausal women originate in approximately equal amounts from the ovaries and adrenals (Refs. 158 and 159; Fig. 15). Peripheral serum testosterone decreases by 50% after ovariectomy in postmenopausal women, thus indicating that the approximately equal contribution of the ovaries and adrenals to serum testosterone remains after menopause. In another study, human chorionic gonadotrophin stimulation and dexamethasone suppression tests in postmenopausal women have suggested that the ovary contributes about 50% of testosterone and 30% of 4-dione in the peripheral circulation (160).



Figure 14. Contribution of the ovaries and adrenals to the serum levels of 4dione in pre- and postmenopausal women, respectively (158, 159).



Figure 15. Contribution of the ovaries and adrenals to the serum levels of testosterone in pre- and postmenopausal women, respectively (158, 159).


To take into account the low degree of diffusion of the active androgens synthesized intracellularly from adrenal DHEA in peripheral target tissues, we estimate that the serum levels of testosterone should be multiplied by about 10 to compare with the testosterone of direct ovarian and adrenal origins. In other words, as mentioned above, only about 10% of intracellular testosterone synthesized from DHEA leaks into the general circulation. The remaining 90% of locally produced testosterone is mostly converted locally into DHT, which is then converted into ADT-G, 3α-diol-G, and 3β-diol-G (Figs. 8-10). Some testosterone and DHT are also glucuronidated and are found in the circulation as Testo-G and DHT-G. One can thus estimate, as illustrated schematically in Fig. 16, that after menopause the contribution of the ovaries to the intracellular concentration of testosterone is only about 10%. This estimate is based on the observation that serum levels of testosterone are reduced by 90% from 15.0 nM to about 1.5 nM after castration in men, whereas the intraprostatic concentration of DHT is reduced only by 50% to about 2.5 ng/g tissue or about 7.5 nM (77,80). Thus, whereas 7.5 nM intratissular DHT of testicular origin corresponds to 13.5 nM serum testosterone, 1.5 nM serum testosterone of adrenal origin corresponds to the same 7.5 nM intratissular DHT, thus requiring a multiplication factor of 9 to compensate for the poor diffusion of testosterone synthesized intracellularly from DHEA compared with the efficacy of entry of circulating testosterone in the prostatic tissue. Such calculations are in agreement with other data showing that serum ADT-G levels reflect essentially adrenal androgen secretion (161). In fact, Giagulli et al. (161) have concluded that DHEA-S accounts for 70 80% of serum ADT-G levels.



Figure 16. Schematic representation of the contribution of the ovaries and adrenals to the serum and intratissular concentrations of testosterone. DHEA is transformed in a series of peripheral intracrine tissues into testosterone, which acts locally on the AR directly or after transformation into the more active androgen DHT. Only a small fraction (estimated at 10%) of the active androgens diffuse into the extracellular space and reach the general circulation, whereas the majority of testosterone and DHT is inactivated by glucuronosyltransferases and released as ADT-G, 3α-diol-G, 3β-diol-G, Testo-G, and DHT-G. These are estimates based on the steroid measurements performed in prostatic tissue of intact and castrated men (77, 80).



III. Androgens Inhibit Breast Cancer


Androgens have been suspected for many decades of being estrogen antagonists and have been used to treat or prevent estrogen-sensitive mammary cancer (162, 163).


A. Clinical data

Estrogens have long been known to play a predominant role in the development and growth of human breast cancer (164 -166). On the other hand, well recognized observations have shown that androgens such as testosterone propionate (162, 167-169), fluoxymesterone (170, 171), and calusterone (172) used in the adjuvant therapy of breast cancer have an efficacy comparable to that achieved with other types of endocrine manipulations (165, 169, 173, 174).


Most importantly, a higher response rate and a longer time to disease progression have been observed when androgens were combined with an antiestrogen, compared with an antiestrogen alone (171, 175). The benefits of combined treatment with fluoxymesterone and tamoxifen vs. tamoxifen alone were observed in postmenopausal women with metastatic breast cancer (175), both in terms of response rate and time to progression of disease.


As summarized later, such additive inhibitory effects of an antiestrogen and androgen on breast cancer have been clearly demonstrated in a series of experimental models. The above-mentioned clinical data are also well supported by the observation of a synergistic effect of DHEA and of the pure antiestrogen EM-800 on prevention of the development of dimethylbenz(a)anthracene (DMBA)-induced mammary tumors in the rat (176). Moreover, the almost exclusive androgenic component in the action of DHEA on the histomorphology and structure of the rat mammary gland has recently been shown (177), thus supporting such an inhibitory effect of DHEA.


It should also be mentioned that androgens have been shown to induce an objective remission after failure of antiestrogen therapy and hypophysectomy. These clinical observations indicate that the benefits obtained with androgen therapy in breast cancer cannot be due solely to a suppression of pituitary gonadotropin secretion but must result, at least in part, from a direct effect on tumor growth in women. The role of androgens as direct inhibitors of breast cancer growth is well supported by the presence of AR in a large proportion of human breast cancers (178-181). In fact, in primary breast cancer, AR has been found in 54% of premenopausal and 48% of postmenopausal patients (180, 182). The presence of AR has also been described in MCF-7 cells (183, 184).


The overwhelming clinical evidence for tumor regression observed in 20 -50% of pre- and postmenopausal breast cancer patients treated with various androgens (173) favors the view that naturally occurring androgens might constitute an as yet overlooked, direct inhibitory control of mammary cancer cell growth. It is thus reasonable to suggest that an imbalance between androgenic and estrogenic influences could modify the overall growth rate of breast tumors in much the same way as that suggested for progestins in estrogen target tissues (185). There is also genetic evidence in agreement with a protective role of androgens against breast cancer (186, 187). Interestingly, the observation that an increased response rate can be obtained by combining androgens and an antiestrogen therapy in breast cancer patients (171,175) is in agreement with our observations summarized later that the mechanisms of the inhibition exerted by the two types of agents are different, whereas their effects, at least in part, are additive.


In this context, it has been found that Western women having a low excretion of adrenal androgenic metabolites respond more poorly to endocrine therapy and have a shorter survival time (188 -190). Possibly because of the small number of cancer cases in many studies, the methodology used, the low predictive value of measurements of serum sex steroid levels, and the association in case-control studies between serum androgen levels and breast cancer risk have led to contradictory data. Thus, subnormal levels of serum androgens have been found in women with increased risk of breast cancer (191-193), whereas opposite data have also been reported (194 -197).


It is of interest that suppression of androgens in men is associated with breast growth (198). Moreover, mutations in AR have been linked with breast cancer in men (199).


It should be added that treatment of ovariectomized monkeys with testosterone decreased by about 40% the stimulation of mammary epithelial proliferation induced by E2 (200). It is possible that part of the increased risk of breast cancer in BRCA-1 mutant patients is associated with the decreased efficiency of the mutated BRCA-1 gene to interact with the AR (201). It is also pertinent to mention that female athletes and transsexuals taking androgens show atrophy of mammary gland epithelial tissue (202, 203).


B. Preclinical data

Lacassagne (204) first observed in 1936 that treatment of mice with testosterone propionate delayed the occurrence of E1-stimulated mammary tumors. In DMBA-induced tumors, high doses of DHT (0.5-4.0 mg/d) for several weeks caused the regression of 60% of established tumors (163). Similar effects were observed with testosterone propionate (205) and dromostanolone propionate (206, 207).


In support of the early clinical data mentioned above, our previous studies have clearly demonstrated that androgens exert a direct inhibitory effect on the proliferation of human breast cancer cells (208-213). In fact, the first demonstration of a potent and direct inhibitory effect of androgens on human breast cancer growth was obtained in the estrogensensitive human breast cancer cell line ZR-75-1 (208). In that study, as shown in Fig. 17A, DHT not only completely blocked the stimulatory effect of E2 on cell proliferation but also reduced cell growth in the absence of estrogens. At low cell density (Fig. 17B), it can be seen that DHT completely prevented breast cancer cell growth.



Figure 17. Time course of the effect of DHT and/or E2 on the proliferation of ZR-75-1 cells. A, Cells were plated at 1 X 104 cells/2.0-cm2 well; 48 h later (zero time), 1 nM E2 (⚫), 10 nM DHT (□), or both steroids (■) were added, and cell numbers were determined at the indicated time intervals. Control cells received the ethanol vehicle only. B, Same as A, except that the initial density was 5.0 X 103 cells/2.0-cm2 well (208).


DHT has been shown to be formed from testosterone and 4-dione in human breast cancer tissue both in vitro in tissue pieces and in vivo (214). Such data indicate the presence of 5α-reductaseinbreastcancertissue,anenzymethoughttobe specific for androgen-dependent tissues. In ZR-75-1 cells, concentrations of DHT in the incubation medium similar to the plasma levels found in normal women (215–217) and breast cancer patients (Ref. 218; 0.3–0.7 nm) are potent inhibitors of the mitogenic effect of E2 and even inhibit growth in the absence of estrogens (208). Furthermore, testosterone, at concentrations observed in adult women (1–3nm; Refs. 215–218), is also a potent inhibitor of cell growth. 4-Dione also led to significant growth inhibition in ZR-75-1 cells, although the active concentrations (IC50, 15 nm) are in the upper range of the plasma concentrations (1–10nm)foundin women (215–218).


Several lines of evidence show that the potent growthinhibitory effect of androgens observed in ZR-71-1 cells is mediated through their specific interaction with the AR. First, the potency of DHT and testosterone to induce antiproliferative effects (IC50, ~0.10 and 0.50 nM, respectively) is in agreement with their relative binding affinity for androgen specific binding sites in intact ZR-75-1 cells as well as in other human breast cancer cells (219, 220). Such values compare well with the potency of DHT to specifically stimulate the secretion of the Zn-α2-glycoprotein (221) and the GCDFP-15 glycoprotein (221, 222) in T47-D human breast cancer cells. The ability of 4-dione to induce an antiproliferative effect (IC50, ~15 nM) most likely results from its metabolic transformation into testosterone and DHT (223-225) than from its direct interaction with the AR (KD, ~200 nM). Secondly, the antiandrogen OH-flutamide competitively reversed the effect of DHT and 4-dione with an apparent dissociation constant (Ki, ~110 nM) consistent with its known affinity for the AR (226, 227).


Because the benefits of combined treatment with an androgen and an antiestrogen have already been observed in women with breast cancer, (171,175), in agreement with the in vitro data mentioned above (208-212), a more precise understanding of the mechanisms of action of androgens and antiestrogens in breast cancer cells becomes important. After a 12-d incubation of ZR-75-1 cells in the presence of 0.1 nM E2 in phenol red-free medium, cell number was increased 2.8-fold above control (P < 0.01; Fig. 18A). The addition of 1 nM DHT, on the other hand, caused a 78% blockade (P < 0.01) of E2-induced ZR-75-1 cell growth, whereas the pure steroidal antiestrogen EM-139 (228), on the other hand, not only completely reversed the effect of E2 but further inhibited cell number by 30% below control values (P < 0.01; Fig. 18B). It can also be seen in Fig. 4B that, in the absence of E2, EM-139 and DHT alone caused 21% (P < 0.01) and 43% (P < 0.01) inhibitions of basal cell growth, respectively. It can also be seen in Fig. 18B that the inhibitory effect of DHT is completely prevented by the addition of the pure antiandrogen OH-flutamide. Most interestingly, in another study, it was found that the growth-inhibitory effect of DHT is clearly additive to that induced by maximally effective concentra tions of the antiestrogen LY156758, thus indicating an action mediated by a mechanism different from interaction with the estrogen receptor (ER; 229). Accordingly, the evidence obtained leaves little doubt that the antiproliferative effect of androgens does not result from competition for binding to the ER, but rather is caused by an AR-mediated mechanism that is additive to blockade of the ER by an antiestrogen.



Figure 18. A, Time course ofthe effect of 0.1 nM E2, 1 nM DHT + E2, 0.3 μM EM-139 + E2, or control medium on the proliferation of ZR-75-1 cells during a 12-d incubation period. B, Time course of the effect of 1 nM DHT, 0.3 μM EM-139, DHT + EM-139, DHT + 0.3 μM OH-FLU, or control medium on the proliferation of ZR-75-1 cells. Three days after plating at an initial density of 5 X 105 cells/10 cm2 per well, cells were incubated with the indicated concentrations of the compounds with medium changes every 48 h for the indicated time periods. At the end of the indicated incubation periods, cell number was determined with a Coulter counter. Data are expressed as means ± SEM of quadruplicate wells (263).


After our demonstration of the inhibitory effect of DHT and antiestrogens on ZR-75-1 cell proliferation in vitro (208 212, 229), we extended our study in vivo to ovariectomized athymic mice using the same human breast cancer cells to more closely mimic the clinical situation in women. We thus examined the effect of DHT on tumor growth stimulated by physiological doses of E2 administered by SILASTIC-brand (Dow Corning Corp., Midland, MI) implants.


As illustrated in Fig. 19, E2 caused a progressive increase in total tumor area from 100% (which corresponds to an average of 0.23 ± 0.08 cm2) at the start of the experiment to 226 ± 31% after 100 d of treatment. Treatment with DHT, on the other hand, not only completely reversed the stimulatory effect of E2 on tumor growth but also decreased total tumor area to 48 ± 10% of its original size. The androgen DHT is thus a potent inhibitor of the stimulatory effect of E2 on ZR-75-1 human breast carcinoma growth in in vivo athymic mice. Similar inhibitory effects on E2-stimulated tumor growth were achieved with medroxyprogesterone acetate [MPA (Provera); Ref. 230], a compound having progestational, androgenic, and glucocorticoid activities (231). Because ovariectomized animals supplemented by exogenous estrogen were used in these studies, such data provide further support for a direct inhibitory action of androgens at the tumor cell level under in vivo conditions, thus adding to the well known inhibitory effect androgens exerted on the pituitary gonadal axis in intact women (232).



Figure 19. Effect of 100-d treatment of ovariectomized athymic mice with Silastic brand implants of E2 (1:3000, E2/cholesterol, wt/wt) alone or in combination with SILASTIC-brand implants of DHT (1:5, DHT/cholesterol, wt/wt) on average total ZR-75-1 tumor area in nude mice. Results are expressed as percentage of pretreatment values (means ± SEM of 11 tumors in the E2 group, 9 tumors in the E2 group, and 9 tumors in the E2 + DHT group; Ref. 230).


Considering the potential importance of androgens in breast cancer therapy, and to better understand the molecular mechanisms responsible for the antagonism between androgens and estrogens, we have investigated the effect of androgens on ER expression in the ZR-75-1 human carcinoma cell line. The specific uptake of [3H]E2 in intact ZR-75-1 cell monolayers was decreased by as much as 88% after a 10-d preincubation with increasing concentrations of DHT (Fig. 20). A half-maximal effect of DHT on [3H]E2 uptake was observed at 70 pM (209). Preincubation with dexamethasone and R5020 (100 nM each) had no effect on the specific uptake of [3H]E2 (data not shown). The addition of hydroxyflutamide, a nonsteroidal antiandrogen devoid of agonistic activity and with no significant affinity for receptors other than the AR (226,227) competitively reversed inhibition of [3H]E2 specific uptake by DHT. The inhibition constant (Ki) value for the reversal of DHT action by hydroxyflutamide was estimated at 39 nM (233), in agreement with the affinity of the antagonist for the AR (227). Thus, the primary site of action of DHT on [3H]E2-specific binding was clearly consistent with a specific interaction with the AR, rather than a direct activation and processing of the ER by DHT (234-239). Similar results were observed on progesterone receptor levels, thus showing a direct inhibitory effect of DHT in human breast cancer cells (209).



Figure 20. Effect of preincubation with increasing concentrations of DHT on [3H]E2-specific binding in ZR-75-1 human breast cancer cells, hydroxylapatite exchange assay of [3H]E2-specific binding of cytosol and nuclear (cytosol + nuclear = total) extracts obtained from ZR75-1 cells preincubated for 11 d with the indicated concentrations of DHT. E2 specific uptake of [3H]E2 in intact ZR-75-1 cells preincubated for 10 d with the indicated concentrations of DHT alone (Ο, control) or in the presence of 3 μM antiandrogen hydroxyflutamide (⚫, OHFLU). Values are given as means ± SE from triplicate determinations (209).


This study showed for the first time that androgens strongly suppress ER content in the human breast cancer cell line ZR-75-1, as measured by radioligand binding and anti-ER monoclonal antibodies. Similar inhibitory effects were observed on the levels of ER mRNA measured by ribonuclease protection assay (209). The androgenic effect was observed at subnanomolar concentrations of the nonaromatizable androgen DHT, regardless of the presence of estrogens, and was competitively reversed by the antiandrogen hydroxyflutamide. Such data on ER expression provide an explanation for at least part of the antiestrogenic effects of androgens on breast cancer cell growth and provide an explanation for the observations showing that the specific inhibitory effects of androgen therapy are additive to the standard treatment limited to blockade of estrogens by antiestrogens (229). Another possible clue to the mechanism of action of DHT in breast cancer cells is provided by the observation that androgens and estrogens exert opposite effects on progesterone receptor levels (240).


The data summarized above clearly support the hypothesis that at least part of the antagonism observed between the action of androgens and estrogens in breast cancer cells (208, 211, 215, 240) may be explained by the heterologous downregulation of the ER by an AR-mediated mechanism. The concentration of DHT needed to exert a half-maximal suppression of ER binding activity (0.07-0.1 nM) is lower than the concentration known to induce binding and nuclear retention of the ER (215, 234). Moreover, the inhibitory effect of DHT on ER content was competitively reversed by the antiandrogen hydroxyflutamide (226, 227). Such data clearly show that AR mediates the down-regulation of the ER by DHT observed in ZR-75-1 cells.


The effect of androgens on ZR-75-1 cell proliferation, however, cannot be solely explained by the suppression of ER expression, because androgens still exert very potent inhibitory effects on growth in the absence of estrogens, even after prolonged periods of estrogen deprivation before exposure to androgens (208, 211). Moreover, the antiproliferative activity of androgens in estrogen-deprived ZR-75-1 cells is more pronounced and is additive to that exerted by antiestrogens (208, 241).


Down-regulation of ER expression by androgens might be of crucial importance in their physiological mode of action, i.e., when estrogens are simultaneously present in normal as well as cancerous mammary gland tissue. In the specific case of human breast cancer, endogenous androgens may reduce the tumor cell sensitivity to estrogens by decreasing ER levels. Thus, in normal breast tissue, endogenous as well as locally produced androgens are likely to contribute to the regulation of the level of ER, thus modulating the sensitivity to estrogens. This inhibitory effect of androgens on intracellular ER concentrations may be expected to leave the relative effectiveness of the competitive blockade of estrogen action by antiestrogens unaffected, while decreasing the efficiency of any residual estrogenic stimulation of cell growth.


In agreement with the in vitro data, Dauvois et al. (242) have shown that constant release of the androgen DHT in ovariectomized rats bearing DMBA-induced mammary carcinoma caused a marked inhibition of tumor growth induced by E2 (Fig. 21). That DHT acts through interaction with the AR in DMBA-induced mammary carcinoma is well supported by the finding that simultaneous treatment with the antiandrogen flutamide completely prevented DHT action. Such data demonstrated, for the first time, that androgens are potent inhibitors of DMBA-induced mammary carcinoma growth by an action independent from inhibition of gonadotropin secretion and suggested an action exerted directly at the tumor level, thus further supporting in vitro data obtained with the human ZR-75-1 breast cancer cell line (208, 209).



Figure 21. Effect of 28-d treatment of ovariectomized (OVX) rats with Silastic brand implants of E2, DHT, E2 + DHT, or E2 + DHT + twice daily injections of flutamide (FLU) on average total DMBA-induced mammary tumor area in the rat. Results are expressed as percentage of pretreatment values as means ± SEM of 22-26 tumors per group. **, P < 0.01 OVX rats treated with the indicated steroid vs. OVX animals at the same time interval (242).


It should be mentioned that in vivo studies have demonstrated that controlled release of low-dose MPA, a compound having androgenic activity, also exerts a potent inhibitory effect on the development and growth of DMBA-induced mammary carcinoma in the rat (243, 244). MPA has in fact been clearly shown to exert androgenic inhibitory effects on the growth of human breast cancer cells in vitro (209, 231, 245), thus suggesting the role of AR in the beneficial effects of MPA in breast cancer in women (246, 247). As previously described, MPA is a compound having a complex series of activities, namely progestational, glucocorticoid, and androgenic (231). Despite the beneficial androgenic effects of this compound observed on human breast cancer cells in vitro (209, 231, 245) and in clinical studies (246, 247), the recent results of the Women's Health Initiative Study (8) clearly indicate that this compound is not recommended for longterm use in normal women where the stimulatory progestational component could well be predominant. A recent study in rats has shown that the addition of methyltestosterone inhibits the marked proliferation of the mammary gland epithelium induced by a low-dose oral contraceptive (248).



IV. DHEA Inhibits Breast Cancer


A. Preclinical studies


1. Introduction

ТLabrie and colleagues (78, 249) first demonstrated that DHEA possesses relatively potent androgenic activity and stimulates androgen-dependent gene expression in the rat ventral prostate. As mentioned earlier, the first androgen successfully used in the treatment of advanced breast cancer was testosterone propionate (250). Many studies subsequently confirmed the beneficial effects of androgens on breast cancer (165, 167-174, 251, 252). Moreover, in vitro studies have provided the first demonstration of the direct antiproliferative activity of androgens on the growth of human mammary carcinoma cells using the cell line ZR75-1 as model (208,253). Interestingly, Poulin et al. (208) have found that the inhibitory effect of androgens on the growth of ZR-75-1 human breast carcinoma cells is additive to that of an antiestrogen. The additive inhibitory effects of an androgen and an antiestrogen on the growth of human breast carcinoma cell line ZR-75-1 have also been observed in vivo in nude mice (230).


2. Inhibitory effect of DHEA on breast cancer

a. Prevention of mammary tumor development by DHEA

As described above, the human adrenals secrete large amounts of the precursor steroids DHEA and DHEA-S, both of which are converted into androgens in target intracrine tissues (25, 35, 78, 92, 249, 254, 255). To investigate the possibility that DHEA and its metabolites could have a preventive effect on the development of mammary carcinoma, we have studied the effect of increasing circulating levels of DHEA constantly released from Silastic brand implants on the development of mammary carcinoma induced by DMBA in the rat. The DMBA-induced mammary carcinoma in the rat has been widely used as a model of hormone-sensitive breast cancer in women (242, 256, 257).


Treatment with increasing doses of DHEA delivered constantly by SILASTIC-brand implants of increasing length and number caused a progressive inhibition of tumor development (258). It is of interest to see that tumor size in the group of animals treated with the highest dose (6 X 3.0-cm long implants) of DHEA was similar to that found in ovariectomized animals (Fig. 22), thus showing a complete blockade of estrogen action by DHEA. Such data clearly demonstrate that circulating levels of the precursor adrenal steroid DHEA comparable to those observed in normal adult premenopausal women (259) exert a potent inhibitory effect on the development of mammary carcinoma induced by DMBA in the rat. It is of special interest to see that serum levels of DHEA of 7.09 ± 0.64 nM and 17.5 ± 1.1 nM led to a dramatic inhibition of tumor development to 22% and 11% of animals bearing mammary carcinoma compared with 68% in control intact animals. At the highest dose of DHEA used, which corresponds to serum DHEA values of 27.2 ± 2.2 nM, the incidence of tumors was reduced to only 3.8%. It should be mentioned that the serum DHEA levels in normal 20to 30-yr-old women ranges between 8.3 and 17.3 nM (259).



Figure 22. Effect of increasing doses of DHEA constantly released from SILASTIC-brand implants and administered 7 d before the intragastric administration of 20 mg of DMBA in intact 50to 52-d-old female rats on average tumor area (cm2) per rat at the indicated time intervals (258).


With the previous knowledge of the potent inhibitory effect of androgens on the growth of human breast cancer as well as on the development and growth of DMBA-induced mammary carcinoma in the rat, it is reasonable to suggest that the present data showing a potent inhibitory effect of DHEA on the development of DMBA-induced mammary carcinoma can be at least partially explained by the androgenic action of the steroids synthesized by the enzymes present in the peripheral target tissues, an action exerted through intracrinology. Although the rat adrenals do not secrete significant amounts of DHEA (80), the enzymes required for the formation of androgens and estrogens are expressed in rat peripheral tissues (260, 261). Such data also suggest a potential use of DHEA as a physiological approach for the prevention of breast cancer in women.


b. Inhibitory effects of DHEA on the growth of human breast cancer xenografts

Because, as mentioned above, androgens have been clearly demonstrated to inhibit the growth of human breast cancer in women as well as in laboratory studies in vitro (167-170,172,175,208 -213,230,242,262-264) and DHEA is predominantly transformed into androgens in the mammary gland, we have studied the possibility that DHEA could inhibit the growth of the human ZR-75-1 breast cancer cell line in vivo in nude mice. To avoid the inhibitory effects of DHEA-derived steroids on gonadotropin secretion, we have used ovariectomized animals supplemented with E1.


As illustrated in Fig. 23, the size of the ZR-75-1 tumors increased by 9.4-fold over a 291-d period (9.5 months) in ovariectomized nude mice supplemented with E1; in contrast, in control ovariectomized mice that received the vehicle alone, tumor size decreased to 36.9% of the initial value during the course of the study (265). On the other hand, treatment with increasing doses of percutaneous DHEA caused a progressive inhibition of E1-stimulated ZR-75-1 tumor growth. Inhibitions of 50.4%, 76.8%, and 80.0% were achieved at 9.5 months of treatment with the daily doses of DHEA of 0.3,1.0, or 3.0 mg per animal, respectively (Fig. 23). In agreement with the decrease in total tumor load, treatment with DHEA led to a marked decrease in the average weight of the tumors remaining at the end of the experiment. To our knowledge, these data provide the first demonstration of the inhibitory effect of DHEA on the growth of human breast cancer xenografts in nude mice.



Figure 23. Effect of increasing doses of DHEA (a total dose of 0.3, 1.0, or 3.0 mg) administered percutaneously in two doses daily on average ZR-75-1 tumor size in ovariectomized nude mice supplemented with 0.5 μg E1 daily. Ovariectomized mice receiving the vehicle alone were used as additional controls. The initial tumor size was taken as 100%. DHEA (0.3, 1.0, or 3.0 mg per animal/d) was administered percutaneously on the dorsal skin in a 0.02-ml solution of 50% ethanol-50% propylene glycol. [Reproduced by permission of Oxford University Press (265).]


In the ovariectomized mouse, exogenous DHEA represents the only source of sex steroids in peripheral tissues, including the mammary gland. Moreover, by itself, DHEA does not possess any significant androgenic or estrogenic activity, its activity being dependent upon its transformation into androgens and/or estrogens in peripheral target intracrine tissues (25). Consequently, the inhibition of tumor growth seen after DHEA treatment in ovariectomized animals results from its intracrine in situ conversion into androgens in the mammary gland (25, 35, 78, 92, 255). In fact, we have recently shown that DHEA exerts an almost exclusively androgenic effect in the rat mammary gland (177). Moreover, DHEA is well known to be converted into androgens, and treatment with DHEA is known to induce androgen-sensitive gene expression in the rat ventral prostate (78, 249). Taken together, these data strongly suggest that DHEA exerts its inhibition of breast cancer development and growth through its conversion to androgens and activation of the AR.


One proposed mechanism for the inhibitory action of DHEA has been the antagonism of DHEA-derived 5-diol on the ER (79, 236, 266). In fact, DHEA can be converted in vivo and in vitro into the weak estrogen 5-diol, which interacts with the ER and can exert weak estrogen-like effects independent from aromatase (253,267-269). That this hypothesis of competition with 5-diol is most unlikely to apply is supported by the observation that increasing doses of diethylstilbestrol, a highly potent estrogen, do not interfere with the inhibitory effect of DHEA on human breast cancer MCF-7 cell proliferation (270). The argument is made even stronger by the finding that tamoxifen did not interfere with the antiproliferative action of DHEA. Moreover, despite the fact that human ZR-75-1 breast cancer cells do not express 3β-HSD and are thus unable to synthesize androgens, thus explaining the stimulatory effect of DHEA on the growth of these cells under in vitro conditions (253), DHEA exerts an inhibitory effect on the growth of the same cancer cells under in vivo conditions in nude mice, the androgens originating from neighboring or distant cells that possess the required mechanisms to transform DHEA into androgens in sufficient amounts to affect other cells after diffusion from their site of synthesis (271).


A group of researchers have reported that DHEA is inhibitory on breast cancer growth in the presence of estrogens, whereas it can be stimulatory on experimental models in which estrogens are absent (197, 270). It should be mentioned, however, that an absence of estrogens does not exist in women where comparable levels of E2 are found in breast cancer tissue in pre- and postmenopausal women (272). In fact, such a hypothetical situation of an absence of estrogens does not exist in normal women, even after menopause.


Although DHT exerts a potent inhibitory effect on breast cancer cell proliferation in ZR-75-1 human breast cancer cells (208, 210), DHT has not always been found to inhibit the growth of MCF-7 cells. The lack of inhibitory action of DHT in some MCF-7 cell lines is most likely due to the presence of a high level of 3α-HSD activity in these cells, thus preventing DHT from exerting its inhibitory effect before its transformation into 3β-diol, a compound having intrinsic estrogenic activity (our unpublished data; and Ref. 273). That the inhibitory effect of DHEA on breast cancer MCF-7 cell growth is due to interaction with AR is supported by the finding that the antiandrogen flutamide reversed the inhibitory effect of DHEA on MCF-7 human breast cancer cell proliferation, whereas the antiestrogen tamoxifen had no effect (274).


c. Additive inhibitory effects of DHEA and the antiestrogen EM-652 on the growth of DMBA-induced mammary tumors

Because antiestrogens (230, 275-278) as well as DHEA (258) can independently inhibit the development of DMBAinduced mammary carcinoma, we have studied the potential benefits of combining the new antiestrogen EM-800 with DHEA on the development of mammary carcinoma induced by DMBA in the rat. As illustrated in Fig. 24, 95% of control animals developed palpable mammary tumors by 279 d after DMBA administration. Treatment with DHEA or EM-800 alone partially prevented the development of DMBAinduced mammary carcinoma, the incidence being thus reduced to 57% (P < 0.01) and 38% (P < 0.01), respectively. Interestingly, combination of the two compounds led to a significantly greater inhibitory effect than that achieved by each compound administered alone (P < 0.01 vs. DHEA or EM-800 alone). In fact, the only two tumors that developed in the group of animals treated with both compounds disappeared before the end of the experiment (279).



Figure 24. Effect oftreatment with DHEA (10 mg, percutaneously, once daily) or EM-800 (75 μg, orally, once daily), alone or in combination for 9 months, on the incidence of DMBA-induced mammary carcinoma in the rat throughout the 279-d observation period. Data are expressed as percentage of the total number of animals in each group (279).


Such data obtained in vivo support our previous findings that the inhibitory effects of androgens and antiestrogens on mammary carcinoma are exerted at least in part by different mechanisms and that the combination of an androgenic compound with a pure antiestrogen has improved efficacy compared with each compound used alone in the prevention and treatment of breast cancer in women. The antagonism between androgens and estrogens on breast cancer growth is illustrated schematically in Fig. 25. DHEA, secondary to its predominant transformation into androgens in mammary gland tissue, exerts an inhibitory effect on mammary carcinoma development and growth, an effect that counteracts and can even completely neutralize the stimulatory effect of estrogens.



Figure 25. Antagonism between the inhibitory effects of androgens and DHEA and the stimulatory effects of estrogens on breast cancer proliferation.


It should be mentioned that recent data suggest that progestins exert a negative impact on breast cancer (2-5), with recent reports indicating an increased risk of the disease in women (6, 7). It is important to indicate that the absence of a stimulatory effect of DHEA on the human endometrium (73, 280) eliminates the need to administer a progestin to neutralize the stimulatory effect of estrogens in this tissue.


Although the above-mentioned data demonstrate the direct inhibitory effects of androgens and DHEA on breast cancer growth, it is likely that endogenous androgens and DHEA play an important physiological role in the control of normal breast tissue growth and function and that this antagonism between androgens and estrogens is also operative in breast cancer.


B. Epidemiological studies

Epidemiological studies have generally observed a protective effect of DHEA on breast cancer, especially in Western women (191, 193, 281, 282). In fact, low serum DHEA levels have been associated with breast cancer in women (281), whereas women with breast cancer were found to have low urinary levels of androsterone and etiocholanolone, two metabolites of DHEA (283, 284). Moreover, women with primary operable breast cancer had urinary levels of 11-deoxy17-ketosteroids (derived mainly from DHEA-S and DHEA) lower than normal, thus suggesting that a low secretion rate of DHEA and DHEA-S could precede the development of breast cancer. It might be relevant to mention that treatment with DHEA markedly delayed the appearance of breast tumors in C3H mice that were genetically bred to develop breast cancer (285).


C. DHEA and other cancers

A series of studies performed in experimental animals have shown the anticarcinogenic activity of DHEA (258, 286, 287). In fact, DHEA has been found to inhibit progression of the cell cycle of pancreatic, breast, and colon cancer cells (274, 288, 289). Moreover, a series of epidemiological studies suggest an inhibitory effect of DHEA on various types of cancers. These epidemiological data pertain to breast cancer (281), prostate cancer (290), and ovarian cancer (291).



V. Rationale for the Use of DHEA as a Source of Androgens in Postmenopausal Women


A. Tissue-specific androgenic and/or estrogenic activity of DHEA

The use of DHEA is based on the recent progress achieved in our understanding of sex steroid physiology in men and women (23, 25, 27, 33, 84, 86, 90, 92, 261, 280, 292, 294) and the recognition that women, at menopause, are not only deprived from estrogens because of a rapid loss of ovarian activity but also have been deprived from androgens for a longer period because of a progressive decrease of serum DHEA levels starting quite a few years before menopause. In fact, as mentioned earlier, normal women produce androgens in amounts equivalent to two thirds of the total amount of androgens synthesized in men (Ref. 26; Table 1). Consequently, the pool of androgens in women decreases progressively from the age of 30 yr in parallel with the decrease in the serum concentration of DHEA and DHEA-S (23). It thus appears logical to include an androgenic component to HRT at periand postmenopause, thus maintaining a physiological balance between estrogens and androgens in each cell and tissue, a goal that can only be achieved by the local formation of androgens and estrogens in peripheral tissues from the steroid precursor DHEA.


An additional reason to use DHEA, the physiological precursor of androgenic steroids, is the recent finding that estrogen therapy, by increasing the concentration of SHBG, which reduces free testosterone, may accelerate lean mass loss among postmenopausal women receiving ERT (295).


We feel that the increased understanding of androgen and estrogen formation and action in peripheral target tissues, called intracrinology (23,25,27,33,35,84,86,90,92,261,280, 292, 294), as well as our recent observations indicating the predominant role of androgens in the prevention of bone loss after ovariectomy in the rat (296) and the observation of a similar situation in postmenopausal women (280) have paved the way for timely and potentially highly significant progress in the field of sex steroid replacement therapy and protection of women's health during aging. Such a possibility is well supported by our observations and that of others of a series of beneficial effects of DHEA in postmenopausal women (73, 74, 280, 297-300).


B. Benefits of DHEA in postmenopausal women

A series of clinical studies have consistently shown beneficial effects of DHEA on physical and psychological wellbeing as well as on bone mineral density (73, 74, 298, 299, 301-306). DHEA replacement in Addison's disease is associated with an improvement in psychological well-being, mood, and fatigue (70). Most importantly, all these benefits, including improved libido, have been obtained without significant side effect (73, 74).


The 70 -95% reduction in the formation of DHEA and DHEA-S by the adrenals during aging results in a dramatic reduction in the formation of androgens and estrogens in peripheral target tissues, which could well be involved in the pathogenesis of age-related diseases such as insulin resistance (307, 308) and obesity (309 -311). In fact, DHEA has been found to improve glucose tolerance (312). Moreover, DHEA has been shown to have immunomodulatory effects in vitro (313) and in vivo in fungal and viral diseases (314), including HIV (315), and a stimulatory effect of DHEA on the immune system has been described in postmenopausal women (316).


As mentioned above, osteoporosis is a major problem among aging women, causing morbidity and mortality mainly through increased fracture rates (317). The use of ERT requires the addition of progestins to counteract the endometrial proliferation induced by estrogens, whereas both estrogens and progestins could increase the risk of breast cancer (5, 318). To avoid the limitations of ERT or HRT, we have studied the effect of 12 months of DHEA administration to 60- to 70-yr-old women on bone mineral density, parameters of bone formation and turnover, serum lipids, glucose and insulin, adipose tissue mass, muscular mass, energy, and well-being, as well as on vaginal and endometrial histology (280, 297). DHEA was administered percutaneously to avoid first passage of the steroid precursor through the liver.


We have thus evaluated the effect of chronic replacement therapy with a 10% DHEA cream applied once daily for 12 months in 60- to 70-yr-old women (n = 15). Anthropometric measurements showed no change in body weight but a 9.8% decrease in sc skin fold thickness at 12 months (P < 0.05; Ref. 297). Bone mass density was increased by 2.3% at the hip, 3.75% at the hip Ward's triangle, and 2.2% at the lumbar spine level (all P < 0.05; Ref. 280). These changes in bone mineral density were accompanied by significant decreases at 12 months of 38% and 22% in urinary hydroxyproline and in plasma bone alkaline phosphatase, respectively (all P < 0.05). An increase of 135% over control (P < 0.05) in plasma osteocalcin was concomitantly observed, thus suggesting increased bone formation in agreement with our preclinical data (296). Such data are in agreement with the finding that the remaining adrenal androgens play an essential role in the maintenance of bone mass in postmenopausal women with Addison's disease (319).


Testosterone administration to elderly men increases the fractional synthetic rate of muscle protein as well as muscle strength (320). The decline of testosterone and DHEA (23,26, 321, 322) with age could be at least partially responsible for sarcopenia in older men and women. In fact, an age-related loss of muscle mass has been observed in women, this loss being particularly important at menopause (323, 324). Loss of muscle mass, especially in the lower extremities, could well increase the risk for fall-related injuries, fractures, and significant loss of independence and quality of life (325, 326).


Measurements of midthigh fat and muscle areas by computed tomography have shown a 3.8% decrease (P < 0.05) of femoral fat and a 3.5% increase (P < 0.05) in femoral muscular area at 12 months of treatment with DHEA (297). There was no significant change in abdominal fat measurements. These changes in body fat and muscular surface areas were associated with a 12% decrease (P < 0.05) of fasting plasma glucose and a 17% decrease (P < 0.05) in fasting plasma insulin levels. Treatment with DHEA had no undesirable effect on the lipid or lipoprotein profile. In fact, there was an overall trend for a 3-10% decrease in total cholesterol. Plasma triglycerides were not affected.


The index of sebum secretion was 79% increased after 12 months of DHEA therapy, with a return to pretreatment values 3 months after cessation of treatment. DHEA administration stimulated vaginal epithelium maturation in 8 of 10 women who had a maturation value of zero at the onset of therapy, whereas a stimulatory effect was also seen in the three women who had an intermediate vaginal maturation before therapy. Most importantly, the estrogenic stimulatory effect observed in the vagina was not found in the endometrium, which remained completely atrophic in all women after 12 months of DHEA treatment (280).


As mentioned above, at the daily 50-mg dose orally, DHEA administered to women with adrenal insufficiency led to significant improvements in well-being, mood, and sexuality in subjects of both sexes (69, 70). Similarly, DHEA treatment in glucocorticoid-treated patients with systemic lupus erythematosus (327, 328) led to significant improvement in overall performance and activity. On the other hand, scores of activity of daily living were improved by DHEA in patients with myotonic dystrophy (329), whereas no change was observed in healthy elderly men (330). A significant improvement in mood and well-being was observed in patients with major depression (331) and midlife asthenia (332), whereas no effect was detected in perimenopausal women (333).


The data obtained after administration of DHEA clearly indicate the beneficial effects of DHEA therapy in postmenopausal women through its transformation into androgens and/or estrogens in specific intracrine target tissues without significant side effects. The absence of stimulation of the endometrium by DHEA eliminates the need for progestin replacement therapy, thus avoiding the fear of progestininduced breast cancer added to the well known stimulatory effect of estrogens. The observed stimulatory effect of DHEA on bone mineral density and the increase in serum osteocalcin, a marker of bone formation, are of particular interest for the prevention and treatment of osteoporosis and indicate a unique activity of DHEA on bone physiology, namely a stimulation of bone formation, whereas ERT and HRT can only reduce the rate of bone loss.


The first studies with DHEA used supraphysiological doses of the compound going up to 800-1600 mg/d (298,309, 334). The oral daily dose of 50 mg, however, has been found as the one providing physiological concentrations of androgens and estrogens (73, 74, 300, 335). We have also determined that the serum levels of DHEA using a 10% cream (280) were comparable to the ones obtained after daily oral administration of 100 mg of DHEA (our unpublished data).


The known specificity of the effect of DHEA in women is summarized in Table 3. Although bone formation, inhibition of mammary gland proliferation, stimulation of sebaceous glands, muscle mass increase, and improved libido are attributed to the formation of androgens in the corresponding target tissues, the decreased insulin resistance and vaginal maturation are best explained by the local formation of estrogens. Most importantly, at physiological replacement doses, DHEA does not stimulate the endometrium, thus removing the need to use a progestin to counteract the stimulation of the endometrium by estrogen. In summary, at physiological replacement doses, DHEA has been found in clinical studies to induce a series of beneficial effects closely associated with the protection of women's health, whereas no negative effects have been observed.


Table 3. Tissue-specific androgenic and estrogenic effects of DHEA





References


Christiansen C, Christensen MS, Larsen NE, Transbol IB 1982 Pathophysiological mechanisms of estrogen effect on bone metabolism. Dose-response relationships in early postmenopausal women. J Clin Endocrinol Metab 55:1124-1130 

Horwitz KB 1992 The molecular biology of RU486. Is there a role for antiprogestins in the treatment of breast cancer? Endocr Rev 13:146-163 

Musgrove ES, Lee CS, Sutherland RL 1991 Progestins both stimulate and inhibit breast cancer cell cycle progression while increasing expression of transforming growth factor a, epidermal growth factor receptor, c-fos, and c-myc genes. Mol Cell Biol 11:5032-5043 

Clarke CL, Sutherland RL 1990 Progestin regulation of cellular proliferation. Endocr Rev 11:266-301 

Colditz GA, Hankinson SE, Hunter DJ, Willett WC, Manson JE, Stampfer MJ, Hennekens C, Rosner B, Speizer FE 1995 The use of estrogens and progestins and the risk of breast cancer in postmenopausal women. N Engl J Med 332:1589-1593 

Ross RK, Paganini-Hill A, Wan PC, Pike MC 2000 Effect of hormone replacement therapy on breast cancer risk: estrogen vs. estrogen plus progestin. J Natl Cancer Inst 92:328-332 

Magnusson C, Baron JA, Correia N, Bergstrom R, Adami HO, Persson I 1999 Breast-cancer risk following long-term oestrogenand oestrogen-progestin-replacement therapy. Int J Cancer 81:339-344 

Women's Health Initiative 2002 Risks and benefits of estrogen plus progestin in healthy postmenopausal women. JAMA 288:321-333 

Lomax P, Schonbaum E1993 Postmenopausal hot flushes and their management. Pharmacol Ther 57:347-358 

Greendale GA, Judd HL 1993 The menopause: health implications and clinical management. J Am Geriatr Soc 41:426-436 

Grady D, Rubin SM, Petitti DB, Fox CS, Black D, Ettinger B, Ernster VL, Cummings SR 1992 Hormone therapy to prevent disease and prolong life in postmenopausal women. Ann Intern Med 117:1016-1037 

Lobo RA 1991 Effects of hormonal replacement on lipids and lipoproteins in postmenopausal women. J Clin Endocrinol Metab 73:925-930 

Harris ST, Genant HK, Baylink DJ, Gallagher JC, Karp SK, McConnell MA, Green EM, Stoll RW 1991 The effects of estrone (Ogen) on spinal bone density of postmenopausal women. Arch Intern Med 151:1980-1984 

Stampfer MJ, Colditz GA, Willett WC, Manson JE, Rosner B, Speizer FE, Hennekens CH 1991 Postmenopausal estrogen therapy and cardiovascular disease. Ten-year follow-up from the nurses' health study. N Engl J Med 325:756-762 

Barrett-Connor E, Bush TL 1991 Estrogen and coronary heart disease in women. JAMA 265:1861-1867 

Lindsay R 1993 Hormone replacement therapy for prevention and treatment of osteoporosis. Am J Med 95:37S-39S 

Field CS, Ory SJ, Wahner HW, Herrmann RR, Judd HL, Riggs BL 1993 Preventive effects of transdermal 17-ß estradiol on osteoporotic changes after surgical menopause: a two-year placebocontrolled trial. Am J Obstet Gynecol 168:114-121 

Vermeulen A, Deslypene JP, Schelfhout W, Verdonck L, Rubens R 1982 Adrenocortical function in old age: response to acute adrenocorticotropin stimulation. J Clin Endocrinol Metab 54:187-191 

Vermeulen A, Verdonck L 1976 Radioimmunoassays of 17ß-hydroxy-5α-androstan-3-one, 4-androstene-3,17-dione, dehydroepiandrosterone, 17ß-hydroxyprogesterone and progesterone and its application to human male plasma. J Steroid Biochem 7:1-10 

Orentreich N, Brind JL, Rizer RL, Vogelman JH 1984 Age changes and sex differences in serum dehydroepiandrosterone sulfate concentrations throughout adulthood. J Clin Endocrinol Metab 59: 551-555 

Belanger A, Candas B, Dupont A, Cusan L, Diamond P, Gomez JL, Labrie F 1994 Changes in serum concentrations of conjugated and unconjugated steroids in 40to 80-year-old men. J Clin Endocrinol Metab 79:1086-1090 

Migeon CJ, Keller AR, Lawrence B, Shepart II TH 1957 Dehydroepiandrosterone and androsterone levels in human plasma. Effect of age and sex: day-to-day and diurnal variations. J Clin Endocrinol Metab 17:1051-1062 

Labrie F, Belanger A, Cusan L, Gomez JL, Candas B 1997 Marked decline in serum concentrations of adrenal C19 sex steroid precursors and conjugated androgen metabolites during aging. J Clin Endocrinol Metab 82:2396-2402 

Lobo RA 2001 Androgens in postmenopausal women: production, possible role, and replacement options. Obstet Gynecol Surv 56: 361-376 

Labrie F 1991 Intracrinology. Mol Cell Endocrinol 78:C113-C118 

Zumoff B, Strain GW, Miller LK, Rosner W 1995 Twenty-fourhour mean plasma testosterone concentration declines with age in normal premenopausal women. J Clin Endocrinol Metab 80:1429-1430 

Labrie F, Belanger A, Cusan L, Candas B 1997 Physiological changes in dehydroepiandrosterone are not reflected by serum levels of active androgens and estrogens but of their metabolites: intracrinology. J Clin Endocrinol Metab 82:2403-2409 

Mazess RB 1982 On aging bone loss. Clin Orthop 165:239-252 

Riggs BL, Wahner HW, Dunn WL, Mazess RB, Offord KP, Melton LJ 1981 Differential changes in bone mineral density of the appendicular and axial skeleton with aging: relationship to spinal osteoporosis. J Clin Invest 67:328-335 

Johnston Jr CC, Hui SL, Witt RM, Appledorn R, Baker RS, Longcope C 1985 Early menopausal changes in bone mass and sex steroids. J Clin Endocrinol Metab 61:905-911 

Steinberg KK, Freni-Titulaer LW, DePuey EG, Miller DT, Sgoutas DS, Coralli CH, Phillips DL, Rogers TN, Clark RV 1989 Sex steroids and bone density in premenopausal and perimenopausal women. J Clin Endocrinol Metab 69:533-539 

Burger HG, Dudley EC, Hopper JL, Groome N, Guthrie JR, Green A, Dennerstein L 1999 Prospectively measured levels of serum follicle-stimulating hormone, estradiol, and the dimeric inhibins during the menopausal transition in a population-based cohort of women. J Clin Endocrinol Metab 84:4025-4030 

Labrie F, Luu-The V, Lin S-X, Simard J, Labrie C, El-Alfy M, Pelletier G, Belanger A 2000 Intracrinology: role of the family of 17β-hydroxysteroid dehydrogenases in human physiology and disease. J Mol Endocrinol 25:1-16 

Martel C, Melner MH, Gagne D, Simard J, Labrie F 1994 Widespread tissue distribution of steroid sulfatase, 3β-hydroxysteroid dehydrogenase/Δ54 isomerase (3β-HSD), 17β-HSD 5α-reductase and aromatase activities in the rhesus monkey. Mol Cell Endocrinol 104:103-111 

Labrie F, Simard J, Luu-The V, Belanger A, Pelletier G, Morel Y, Mebarki F, Sanchez R, Durocher F, Turgeon C, Labrie Y, Rheaume E, Labrie C, Lachance Y 1996 The 3β-hydroxysteroid dehydrogenase/isomerase gene family: lessons from type II 3β-HSD congenital deficiency. In: Hansson V, Levy FO, Tasken K, eds. Signal transduction in testicular cells. Ernst Schering Research Foundation Workshop. Berlin, Heidelberg, New York: SpringerVerlag; 185-218 

Chesnut CH, Ivey JL, Gruber HE, Matthews M, Nelp WB, Sisom K, Baylink DJ 1983 Stanozolol in postmenopausal osteoporosis: therapeutic efficacy and possible mechanisms of action. Metabolism 32:571-580 

Benz DJ, Haussler MR, Thomas MA, Speelman B, Komm BS 1991 High-affinity androgen binding and androgenic regulation of a1(I)procollagen and transforming growth factor- steady state messenger ribonucleic acid levels in human osteoblast-like osteosarcoma cells. Endocrinology 128:2723-2730 

Kapur SP, Reddi AH 1989 Influence of testosterone and dihydrotestosterone on bone-matrix induced endochondral bone formation. Calcif Tissue Int 44:108-113 

Abu EO, Horner A, Kusec V, Triffit JT, Compston JE 1997 The localization of androgen receptors in human bone. J Clin Endocrinol Metab 82:3493-3497 

Colvard DS, Eriksen EF, Keeting PE, Wilson EM, Lubahn DB, French FS, Spelsberg TC 1989 Identification of androgen receptors in normal human osteoblast-like cell. Proc Natl Acad Sci USA 86:854-857 

Kasperk CH, Wergedal JE, Farley JR, Linkhart TA, Turner RT, Baylink DJ 1989 Androgens directly stimulate proliferation of bone cells in vitro. Endocrinology 124:1576-1578 

Davis SR, McCloud P, Strauss BJ, Burger H 1995 Testosterone enhances estradiol's effects on postmenopausal density and sexuality. Maturitas 21:227-236 

Raisz LG, Wiita B, Artis A, Bowen A, Schwartz S, Trahiotis M, Shoukri K, Smith J 1996 Comparison of the effects of estrogen alone and estrogen plus androgen on biochemical markers of bone formation and resorption in postmenopausal women. J Clin Endocrinol Metab 81:37-43 

Munnuz-Torres M, Jodar E, Quesada M, Escobar-Jimenez F 1995 Bone mass in androgen-insensitivity syndrome: response to hormonal replacement therapy. Calcif Tissue Int 57:94-96 

Soule SG, Conway G, Prelevic GM, Prentice M, Ginsburg J, Jacobs HS 1995 Osteopenia as a feature of the androgen insensitivity syndrome. Clin Endocrinol (Oxf) 43:671-675 

Bertelloni S, Baroncelli GI, Federico G,Cappa M, Lala R, Saggese G 1998 Altered bone mineral density in patients with complete androgen insensitivity syndrome. Horm Res 50:309-314 

Slemenda C, Longcope C, Peacock M, Hui S, Johnston CC 1996 Sex steroids, bone mass, and bone loss. A prospective study of pre-, peri-, and postmenopausal women. J Clin Invest 97:14-21 

Hennernan PM, Wallach S 1957 The role of androgens and estrogens and their metabolic effects. A review of the prolonged use of estrogens and androgens in postmenopausal and senile osteoporosis. AMA Arch Intern Med 100:715-723 

Need AG, Horowitz M, Bridges A, Morris HA, Nordin BE 1989 Effects of nandrolone decanoate and antiresorptive therapy on vertebral density in osteoporotic postmenopausal women. Arch Intern Med 149:57-60 

Savvas M, Studd JWW, Fogelman I, Dooley M, Montgomery J, Murby B 1988 Skeletal effects of oral oestrogen compared with subcutaneous oestrogen and testosterone in postmenopausal women. Br Med J 297:331-333 

Deleted in proof Barrett-Connor E, Young R, Notelovitz M, Sullivan J, Wiita B, Yang HM, Nolan J 1999 A two-year, double-blind comparison of estrogen-androgen and conjugated estrogens in surgically menopausal women. J Reprod Med 44:1012-1020 

Castelo-Branco C 2000 Comparative effects of estrogens plus androgens and tibolone on bone, lipid pattern and sexuality in postmenopausal women. Maturitas 34:161-168 

Davis SR 1999 Androgen replacement in women: a commentary. J Clin Endocrinol Metab 84:1886-1891 

Davis SR, Burger HG 1996 Androgens and postmenopausal women. J Clin Endocrinol Metab 81:2759-2763 

Studd JW, Collins WP, Chakravarti S, Newton JR, Oram D, Parsons A 1987 Estradiol and testosterone implants in treatment of psychosexual problems in postmenopausal woman. Br J Obstet Gynecol 84:314-315 

Burger HG, Hailes J, Menelaus M, Nelson J, Hudson B, Balazs N 1984 The management of persistent menopausal symptoms with oestradiol-testosterone implants: clinical, lipid and hormonal results. Maturitas 6:351-358 

Sherwin BB 1988 Affective changes with estrogen and androgen replacement therapy in surgically menopausal women. J Affect Disord 14:177-187 

Sherwin BB, Gelfand MM 1985 Differential symptom response to parenteral estrogen and/or androgen administration in the surgical menopause. Am J Obstet Gynecol 151:153-160 

Watts NB, Notelovitz M, Timmons MC, Addison WA, Wiita B, Downey LJ 1995 Comparison of oral estrogens and estrogens plus androgen on bone mineral density, menopausal symptoms, and lipid-lipoprotein profiles in surgical menopause. Obstet Gynecol 85:529-537 

Pye JK, Mansel RE, Hughes LE 1985 Clinical experience of drug treatments for mastalgia. Lancet 2:373-377 

Leiblum S, Bachmann G, Kemmann E, Colburn D, Swartzman L 1983 Vaginal atrophy in the postmenopausal women. The importance of sexual activity and hormones. JAMA 249:2195-2198 

Sherwin BB, Gelfand MM 1987 The role of androgen in the maintenance of sexual functioning in oophorectomized women. Psychosom Med 49:397-409 

BagatellCJ,BremnerWJ 1997Androgensandbehaviorinmenand women. Endocrinologist 7:97-102 

Davis SR, Tran J 2001 Testosterone influences libido and well being in women. Trends Endocrinol Metab 12:33-37 

Davis SR 2000 Androgens and female sexuality. J Gend Specif Med 3:36-40 

Davis SR 1999 The therapeutic use of androgens in women. J Steroid Biochem Mol Biol 69:177-184 

Shifren JL, Braunstein GD, Simon JA, Casson PR, Buster JE, Redmond GP, Burki RE, Ginsburg ES, Rosen RC, Leiblum SR, Caramelli KE, Mazer NA 2000 Transdermal testosterone treatment in women with impaired sexual function after oophorectomy. N Engl J Med 343:682-688 

Arlt W, Callies F, van Vlijmen JC, Koehler I, Reincke M, Bidlingmaier M, Huebler D, Oettel M, Ernst M, Schulte HM, Allolio B 1999 Dehydroepiandrosterone replacement in women with adrenal insufficiency. N Engl J Med 341:1013-1020 

Hunt PJ, Gurnell EM, Huppert FA, Richards C, Prevost AT, Wass JA, Herbert J, Chatterjee VK 2000 Improvement in mood and fatigue after dehydroepiandrosterone replacement in Addison's disease in a randomized, double blind trial. J Clin Endocrinol Metab 85:4650-4656 

Sherwin BB, Gelfand MM 1984 Effects of parenteral administration of estrogen and androgen on plasma hormone levels and hot flushes in the surgical menopause. Am J Obstet Gynecol 148:552-557 

De Fazio J, Meldrum DR, Winer JH, Judd HL 1984 Direct action of androgen on hot flushes in the human male. Maturitas 6:3-8 

Baulieu EE, Thomas G, Legrain S, Lahlou N, Roger M, Debuire B, Faucounau V, Girard L, Hervy MP, Latour F, Leaud MC, Mokrane A, Pitti-Ferrandi H, Trivalle C, de Lacharriere O, Nouveau S, Rakoto-Arison B, Souberbielle JC, Raison J, Le Bouc Y, Raynaud A, Girerd X, Forette F 2000 Dehydroepiandrosterone (DHEA), DHEA sulfate, and aging: contribution of the DHEAge Study to a sociobiomedical issue. Proc Natl Acad Sci USA 97:4279-4284 

Stomati M, Monteleone P, Casarosa E, Quirici B, Puccetti S, Bernardi F, Genazzani AD, Rovati L, Luisi M, Genazzani AR 2000 Six-months oral dehydroepiandrosterone supplementation in early and late postmenopause. Gynecol Endocrinol 14:342-363 

Worboys S, Kotsopoulos D, Teede H, McGrath B, Davis SR 2001 Evidence that parenteral testosterone therapy may improve endothelium-dependent and -independent vasodilation in postmenopausal women already receiving estrogen. J Clin Endocrinol Metab 86:158-161 

Davis SR, Walker KZ, Strauss BJ 2000 Effects of estradiol with and without testosterone on body composition and relationships with lipids in postmenopausal women. Menopause 7:395-401 

Labrie F, Dupont A, Be langer A1985 Complete androgen blockade for the treatment of prostate cancer. In: de Vita VT, Hellman S, Rosenberg SA, eds. Important advances in oncology. Philadelphia: J.B. Lippincott; 193-217 

Labrie C, Belanger A, Labrie F 1988 Androgenic activity of dehydroepiandrosterone and androstenedione in the rat ventral prostate. Endocrinology 123:1412-1417 

Thijssen JH, van Landeghem AA, Poortman J 1986 Uptake and concentration of steroid hormones in mammary tissues. Ann NY Acad Sci 464:106-116 

Belanger B, Belanger A, Labrie F, Dupont A, Cusan L, Monfette G 1989 Comparison of residual C-19 steroids in plasma and prostatic tissue of human, rat and guinea pig after castration: unique importance of extratesticular androgens in men. J Steroid Biochem 32:695-698 

Buzdar AU, Smith R, Vogel C, Bonomi P, Keller AM, Favis G, Mulagha M, Cooper J 1996 Fadrozole HCL (CGS-16949A) versus megestrol acetate treatment of post-menopausal patients with metastatic breast carcinoma: results of two randomized double-blind controlled multiinstitutional trials. Cancer 77:2503-2513 

Cummings SR, Eckert S, Krueger KA, Grady D, Powles TJ, Cauley JA, Norton L, Nickelsen T, Bjarnason NH, Morrow M, Lippman ME, Black D, Glusman JE, Costa A, Jordan VC 1999 The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation. JAMA 281:2189-2197 

Labrie F, Sugimoto Y, Luu-The V, Simard J, Lachance Y, Bachvarov D, Leblanc G, Durocher F, Paquet N 1992 Structure of human type II 5α-reductase. Endocrinology 131:1571-1573 

Labrie F, Simard J, Luu-The V, Belanger A, Pelletier G 1992 Structure, function and tissue-specific gene expression of 3β-hydroxysteroid dehydrogenase/5-ene-4-ene isomerase enzymes in classical and peripheral intracrine steroidogenic tissues. J Steroid Biochem Mol Biol 43:805-826 

Luu-The V, Zhang Y, Poirier D, Labrie F 1995 Characteristics of human types 1, 2 and 3 17β-hydroxysteroid dehydrogenase activities: oxidation-reduction and inhibition. J Steroid Biochem Mol Biol 55:581-587 

Labrie Y, Durocher F, Lachance Y, Turgeon C, Simard J, Labrie C, Labrie F 1995 The human type II 17β-hydroxysteroid dehydrogenase gene encodes two alternatively-spliced messenger RNA species. DNA Cell Biol 14:849 861 

Belanger A, Brochu M, Cliche J 1986 Levels of plasma steroid glucuronides in intact and castrated men with prostatic cancer. J Clin Endocrinol Metab 62:812-815 

Labrie F, Belanger A, Dupont A, Luu-The V, Simard J, Labrie C 1993 Science behind total androgen blockade: from gene to combination therapy. Clin Invest Med 16:475-492 

Simpson ER, Davis SR 2001 Aromatase and the regulation of estrogen biosynthesis: some new perspectives. Endocrinology 142: 4589-4594 

Labrie F, Simard J, Luu-The V, Pelletier G, Belghmi K, Belanger A 1994 Structure, regulation and role of 3β-hydroxysteroid dehydrogenase, 17β-hydroxysteroid dehydrogenase and aromatase enzymes in formation of sex steroids in classical and peripheral intracrine tissues. Bailliere's Clin Endocrinol Metab 8:451-474 

Pelletier G, Dupont E, Simard J, Luu-The V, Belanger A, Labrie F1992 Ontogeny and subcellular localization of 3β-hydroxysteroid dehydrogenase (3β-HSD) in the human and rat adrenal, ovary and testis. J Steroid Biochem Mol Biol 43:451-467 

Labrie F, Luu-The V, Lin SX, Labrie C, Simard J, Breton R, Be langer A 1997 The key role of 17β-HSDs in sex steroid biology. Steroids 62:148-158 

Luu-The V, Lachance Y, Labrie C, Leblanc G, Thomas JL, Strickler RC, Labrie F 1989 Full length cDNA structure and deduced amino acid sequence of human 3β-hydroxy-5-ene steroid dehydrogenase. Mol Endocrinol 3:1310-1312 

Lachance Y, Luu-The V, Labrie C, Simard J, Dumont M, de Launoit Y, Guerin S, Leblanc G, Labrie F 1990 Characterization of human 3β-hydroxysteroid dehydrogenase/Δ54 isomerase gene and its expression in mammalian cells. J Biol Chem 265:20469-20475 

Rheaume E, Lachance Y, Zhao HF, Breton N, Dumont M, de Launoit Y, Trudel C, Luu-The V, Simard J, Labrie F1991 Structure and expression of a new cDNA encoding the almost exclusive 3β-hydroxysteroid dehydrogenase/Δ54 isomerase in human adrenals and gonads. Mol Endocrinol 5:1147-1157 

Lachance Y, Luu-The V, Verreault H, Dumont M, Rhe aume E, Leblanc G, Labrie F 1991 Structure of the human type II 3β-hydroxysteroid dehydrogenase/Δ54 isomerase (3β-HSD) gene: adrenal and gonadal specificity. DNA Cell Biol 10:701-711 

Luu-The V, Takahashi M, de Launoit Y, Dumont M, Lachance Y, Labrie F 1991 Evidence for distinct dehydrogenase and isomerase sites within a single 3β-hydroxysteroid dehydrogenase 5-ene-4ene-isomerase protein. Biochemistry 30:8861-8865 

Huang XF, Luu-The V 2001 Gene structure, chromosomal localization and analysis of 3-ketosteroid reductase activity of the human 3(a3β-hydroxysteroid epimerase. Biochim Biophys Acta 1520:124-130 

Huang X-F, Luu-The V 2000 Molecular characterization of a first human 3 (α-;β)-hydroxysteroid epimerase. J Biol Chem 275:29452-29457 

Milewich L, Shaw CE, Mason JI, Carr BR, Blomquist CH, Thomas JL 1993 3β-Hydroxysteroid dehydrogenase activity in tissues of human fetus determined with 5α-androstane-3β,17β-diol and dehydroepiandrosterone as substrates. J Steroid Biochem Mol Biol 45:525-537 

Luu-The V, Labrie C, Zhao HF, Couet J, Lachance Y, Simard J, Cote J, Leblanc J, Lagace L, Berube D, Gagne R, Labrie F 1990 Purification, cloning complementary DNA structure and predicted amino acid sequence of human estradiol 17β-dehydrogenase. Ann NY Acad Sci 595:40-52 

Morissette J, Rhe aume E, Leblanc JF, Luu-The V, Labrie F, Simard J 1995 Genetic linkage mapping of the HSD3B1 and HSD3B2 genes encoding human types I and II3β-hydroxysteroid dehydrogenase/ Δ54 isomerase close to D1S514 and the centromeric D1Z5 locus. Cytogenet Cell Genet 69:59-62 

Rhe aume E, Simard J, Morel Y, Mebarki F, Zachmann M, Forest M, New MI, Labrie F 1992 Congenital adrenal hyperplasia due to point mutations in the type II 3β-hydroxysteroid dehydrogenase gene. Nat Genet 1:239-245 

Simard J, Rheaume E, Sanchez R, Laflamme N, de Launoit Y, Luu-The V, Van Seters AP, Gordon RD, Bettendorf M, Heinrich U, Moshang T, New MI, Labrie F 1993 Molecular basis of congenital adrenal hyperplasia due to 3β-hydroxysteroid dehydrogenase deficiency. Mol Endocrinol 7:716-728 

Simard J, Rhe aume E, Mebarki F, Sanchez R, New MI, Morel Y, Labrie F 1995 Molecular basis of human 3β-hydroxysteroid dehydrogenase deficiency. J Steroid Biochem Mol Biol 53:127-138 

Dufort I, Rheault P, Huang XF, Soucy P, Luu-The V 1999 Characteristics of a highly labile human type 5 17β-hydroxysteroid dehydrogenase. Endocrinology 140:568-574 

Luu-The V, Dufort I, Pelletier G, Labrie F 2001 Type 5 17β-hydroxysteroid dehydrogenase: its role in the formation of androgens in women. Mol Cell Endocrinol 171:77-82 

Peltoketo H, Isomaa V, Maentausta O, Vihko R 1988 Complete amino acid sequence of human placental 17β-hydroxysteroid dehydrogenase deduced from cDNA. FEBS Lett 239:73-77 

Luu-The V, Labrie C, Zhao HF, Couet J, Lachance Y, Simard J, Leblanc G, Co te J, Be rube D, Gagne R, Labrie F 1989 Characterization of cDNAs for human estradiol 17β-dehydrogenase and assignment of the gene to chromosome 17: evidence of two mRNA species with distinct 5' termini in human placenta. Mol Endocrinol 3:1301-1309 

Luu-The V, Labrie C, Simard J, Lachance Y, Zhao HF, Couet J, Leblanc G, Labrie F 1990 Structure of two in tandem human 17β-hydroxysteroid dehydrogenase genes. Mol Endocrinol 4:268-275 

Peltoketo H, Isomaa V, Vihko R 1992 Genomic organization and DNA sequences of human 17β-hydroxysteroid dehydrogenase genes and flanking regions: localization of multiple Alu sequences and putative cis-acting elements. Eur J Biochem 209:459-466 

Dumont M, Luu-The V, de Launoit Y, Labrie F 1992 Expression of human 17β-hydroxysteroid dehydrogenase in mammalian cells. J Steroid Biochem Mol Biol 41:605-608 

Lin SX, Yang F, Jin JZ, Breton R, Zhu DW, Luu-The V, Labrie F 1992 Subunit identity of the dimeric 17β-hydroxysteroid dehydrogenase from human placenta. J Biol Chem 267:16182-16187 

Zhu DW, Lee X, Breton R, Ghosh D, Pangborn W, Duax W, Lin SW 1993 Crystallization and preliminary x-ray diffraction analysis of the complex of human placental 17β-hydroxysteroid dehydrogenase with NADP+. J Mol Biol 234:242-244 

Breton R, Yang F, Jin JZ, Li B, Labrie F, Lin SX 1994 Human 17β-hydroxysteroid dehydrogenase: overproduction using a baculovirus expression system and characterization. J Steroid Biochem Mol Biol 50:275-282 

Ghosh D, Pletnev VZ, Zhu DW, Wawrzak Z, Duax WL, Pangborn W, Labrie F, Lin SX 1995 Structure of human estrogenic 17β-hydroxysteroid dehydrogenase at 2.20A resolution. Structure 3:503-513 

Wu L, Einstein M, Geissler WM, Chan KH, Elliston KO, Andersson S 1993 Expression cloning and characterization of human 17β-hydroxysteroid dehydrogenase type 2, a microsomal enzyme possessing 20α-hydroxysteroid dehydrogenase activity. J Biol Chem 268:12964-12969 

Andersson S, Geissler WM, Patel S, Wu L 1995 The molecular biology of androgenic 17β-hydroxysteroid dehydrogenases. J Steroid Biochem Mol Biol 53:37-39 

Geissler WM, Davis DL, Wu L, Bradshaw KD, Patel S, Mendoca BB, Elliston KO, Wilson JD, Russell DW, Andersson S 1994 Male pseudohermaphroditism caused by mutations of testicular 17β-hydroxysteroid dehydrogenase 3. Nat Genet 7:34-39 

Leenders F, Adamski J, Husen B, Thole HH, Jungblut PW 1994 Molecular cloning and amino acid sequence of the porcine 17β-estradiol dehydrogenase. Eur J Biochem 222:221-227 

Adamski J, Normand T, Leenders F, Monte D, Begue A, Stehelin D, Jungblut PW, de Launoit Y 1995 Molecular cloning of a novel widely expressed human 80 kDa 17β-hydroxysteroid dehydrogenase IV. Biochem J 311:437-443 

de Launoit Y, Adamski J 1999 Unique multifunctional HSD17B4 gene product: 17β-hydroxysteroid dehydrogenase 4 and D-3Hydroxyacyl-coenzyme A dehydrogenase/hydratase involved in Zellweger syndrome. J Mol Endocrinol 22:227-240 

Russell P, Bannatyne P 1989 Surgical pathology of the ovaries. New York, Edinburgh: Churchill-Livingstone Sluijmer AV, Heineman MJ, Koudstaal J, Theunissen PH, de Jong FH, Evers JL 1998 Relationship between ovarian production of estrone, estradiol, testosterone, and androstenedione and the ovarian degree of stromal hyperplasia in postmenopausal women. Menopause 5:207-210 

Pelletier G, Luu-The V, Tetu B, Labrie F 1999 Immunocytochemical localization of type 5 17β-hydroxysteroid dehydrogenase in human reproductive tissues. J Histochem Cytochem 47:731-737 

Pelletier G, Luu-The V, El-Alfy M, Li S, Labrie F 2001 Immunoelectron microscopic localization of 3β-hydroxysteroid dehydrogenase and type 5 17β-hydroxysteroid dehydrogenase in the human prostate and mammary gland. J Mol Endocrinol 26:11-19 

Biswas MG, Russell DW 1997 Expression cloning and characterization of oxidative 17β-and 3α-hydroxysteroid dehydrogenase from rat and human prostate. J Biol Chem 272:15959-15966 

Duan WR, Linzer DIH, Gibori G 1996 Cloning and characterization of an ovarian-specific protein that associates with the short form of the prolactin receptor. J Biol Chem 271:15602-15607 

Nokelainen P, Peltoketo H, Vihko R, Vihko P 1998 Expression cloning of a novel estrogenic mouse 17β-hydroxysteroid dehydrogenase/17-ketosteroid reductase (m17HSD7), previously described as a prolactin receptor-associated protein (PRAP) in rat. Mol Endocrinol 12:1048-1059 

Krazeisen A, Breitling R, Imai K, Fritz S, Moller G, Adamski J 1999 Determination of cDNA, gene structure and chromosomal localization of the novel human 17β-hydroxysteroid dehydrogenase type 7(1). FEBS Lett 460:373-379 

Kikuti YY, Tamiya G, Ando A, Chen L, Kimura M, Ferreira E, Tsuji K, Trowsdale J, Inoko H 1997 Physical mapping 220 kb centromeric of the human MHC and DNA sequence analysis of the 43-kb segment including the RING1, HKE6, and HKE4 genes. Genomics 42:422-435 

Aziz N, Maxwell MM, St. Jacques B, Brenner BM 1993 Downregulation of Ke 6, a novel gene encoded within the major histocompatibility complex, in murine polycystic kidney disease [published erratum appears in Mol Cell Biol 13(10):6614]. Mol Cell Biol 13:1847-1853 

Fomitcheva J, Baker ME, Anderson E, Lee GY, Aziz N 1998 Characterization of Ke 6, a new 17β-hydroxysteroid dehydrogenase, and its expression in gonadal tissues. J Biol Chem 273:22664-22671 

Luu-The V 2001 Analysis and characteristics of multiple types of human 17β-hydroxysteroid dehydrogenase. J Steroid BiochemMol Biol 76:143-151 

Andersson S, Russell DW 1990 Structural and biochemical properties of cloned and expressed human and rat steroid 5α-reductases. Proc Natl Acad Sci USA 87:3640-3644 

Andersson S, Berman DM, Jenkins EP, Russell DW 1991 Deletion of steroid 5α-reductase 2 gene in male pseudohermaphroditism. Nature 354:159-161 

Jenkins EP, Hsieh CL, Milatovich A, Normington K, Berman DM, Francke U, Russell DW 1991 Characterization and chromosomal mapping of human steroid 5α-reductase gene and pseudogene and mapping of the mouse homologue. Genomics 11:1102-1112 

Wilson JD, Griffin JE, Russell DW 1993 Steroid 5α-reductase 2 deficiency. Endocr Rev 14: 577-593 

Luu-The V, Sugimoto Y, Puy L, Labrie Y, Lopez I, Singh M, Labrie F 1994 Characterization, expression and immunohistochemical localization of 5α-reductase in human skin. J Invest Dermatol 102: 221-226 

Barbier O, Girard C, Lapointe H, El-Alfy M, Hum DW, Belanger A 2000 Cellular localization of uridine diphosphoglucuronosyltransferase 2B enzymes in the human prostate by in situ hybridization and immunohistochemistry. J Clin Endocrinol Metab 85: 4819-4826 

Turgeon D, Carrier JS, Levesque E, Hum DW, Belanger A 2001 Relative enzymatic activity, protein stability, and tissue distribution of human steroid-metabolizing UGT2B subfamily members. Endocrinology 142:778-787 

Arlt W, Callies F, Koehler I, van Vlijmen JC, Fassnacht M, Strasburger CJ, Seibel J, Huebler D, Ernst M, Oettel M, Reincke M, Schulte HM, Allolio B 2001 Dehydroepiandrosterone (DHEA) supplementation in healthy men with an age-related decline of DHEA secretion. J Clin Endocrinol Metab 86:4686-4692 

Casson PR, Santoro N, Elkind-Hirsch K, Carson SA, Hornsby PJ, Abraham G, Buster JE 1998 Postmenopausal dehydroepiandrosterone administration increases free insulin-like growth factor-I and decreases high-density lipoprotein: a six-month trial. Fertil Steril 70:107-110 

Buster JE, Casson PR, Straughn AB, Dale D, Umstot ES, Chiamori N, Abraham GE 1992 Postmenopausal steroid replacement with micronized dehydroepiandrosterone: preliminary oral bioavailability and dose proportionality studies. Am J Obstet Gynecol 166:1163-1168; discussion, 1168-1170 

Adashi EY 1994 The climacteric ovary as a functional gonadotropin-driven androgen-producing gland. Fertil Steril 62:20-27 

Longcope C 1998 The relationship of ovarian production of hormones and degree of ovarian stromal hyperplasia in postmenopausal women. Menopause 5:205-206 

Judd HL, Judd GE, Lucas WE, Yen SS 1974 Endocrine function of the postmenopausal ovary: concentration of androgens and estrogens in ovarian and peripheral vein blood. J Clin Endocrinol Metab 39:1020-1024 

Longcope C, Hunter R, Franz C 1980 Steroid secretion by the postmenopausal ovary. Am J Obstet Gynecol 138:564-568 

Lucisano A, Russo N, Acampora MG, Fabiano A, Fattibene M, Parlati E, Maniccia E, Dell'Acqua S 1986 Ovarian and peripheral androgen and oestrogen levels in post-menopausal women: correlations with ovarian histology. Maturitas 8:57-65 

Dowsett M, Cantwell B, Lal A, Jeffcoate SL, Harris AL 1988 Suppression of postmenopausal ovarian steroidogenesis with the luteinizing hormone-releasing hormone agonist goserelin. J Clin Endocrinol Metab 66:672-677 

Sluijmer AV, Heineman MJ, De Jong FH, Evers JL1995 Endocrine activity of the postmenopausal ovary: the effects of pituitary downregulation and oophorectomy. J Clin Endocrinol Metab 80:2163-2167 

Couzinet B, Meduri G, Lecce MG, Young J, Brailly S, Loosfelt H, Milgrom E, Schaison G 2001 The postmenopausal ovary is not a major androgen-producing gland. J Clin Endocrinol Metab 86:5060-5066 

Rannevik G, Carlstrom K, Jeppsson S, Bjerre B, Svanberg L 1986 A prospective long-term study in women from pre-menopause to post-menopause: changing profiles of gonadotrophins, oestrogens and androgens. Maturitas 8:297-307 

Roger M, Nahoul K, Scholler R, Bagrel D 1980 Evolution with ageing of four plasma androgens in postmenopausal women. Maturitas 2:171-177 

Jiroutek MR, Chen MH, Johnston CC, Longcope C 1998 Changes in reproductive hormones and sex hormone-binding globulin in a group of postmenopausal women measured over 10 years. Menopause 2:90-94 

Meldrum DR, Davidson BJ, Tataryn IV, Judd HL 1981 Changes in circulating steroids with aging in postmenopausal women. Obstet Gynecol 57:624-628 

Adashi EY 1991 The climacteric ovary: a viable endocrine organ. Semin Reprod Endocrinol 9:200-205 

Judd HL, Lucas WE, Yen SS 1974 Effect of oophorectomy on circulating testosterone and androstenedione levels in patients with endometrial cancer. Am J Obstet Gynecol 118:793-798 

Maroulis GB, Abraham GE 1976 Ovarian and adrenal contributions to peripheral steroid levels in postmenopausal women. Obstet Gynecol 48:150-154 

Vermeulen A 1976 The hormonal activity of the postmenopausal ovary. J Clin Endocrinol Metab 42:247-253 

Giagulli VA, Giorgino R, Vermeulen A 1993 Origin and significance of plasma androsterone glucuronide levels: a parameter of adrenal androgen secretion and hepatic 5 a-reductase activity. J Clin Endocrinol Metab 76:918-923 

Ulrich P 1939 Testosterone (hormone male) et son role possible dans le traitement de certains cancers du sein. Acta Unio Internationalis Contra Cancrum 4:377-379 

Huggins C,Briziarelli G, Sutton HJ 1959 Rapid induction of mammary carcinoma in the rat and the influence of hormone on the tumors. J Exp Med 109:25-42 

Lippman ME 1985 Growth regulation of human breast cancer. Clin Res 34:375-382 

McGuire WL, Carbone PP, Sears ME, Escher GC 1975 Estrogen receptors in human breast cancer: an overview. In: McGuire WL, Carbone PP, Vollmer EP, eds. Estrogen receptors in human breast cancer. New York: Raven Press; 1-7 

Davidson NE, Lippman ME 1989 The role of estrogens in growth regulation of breast cancer. Crit Rev Oncog 1:89-111 

Fels E 1944 Treatment of breast cancer with testosterone propionate. A preliminary report. J Clin Endocrinol 4:121-125 

Segaloff A,GordonD,HorwittBN,SchlosserJV,MurisonPJ 1951 Hormonal therapy in cancer of the breast. 1. The effect of testosterone propionate therapy on clinical course and hormonal excretion. Cancer 4:319-323 

Cooperative Breast Cancer Group 1964 Testosterone propionate therapy of breast cancer. JAMA 188:1069-1072 

Kennedy BJ 1958 Fluxymesterone therapy in treatment of advanced breast cancer. N Engl J Med 259:673-675 

Tormey DC, Lippman ME, Edwards BK, Cassidy JG 1983 Evaluation of tamoxifen doses with and without fluoxymesterone in advanced breast cancer. Ann Intern Med 98:139-144 

Gordan GS, Halden A, Horn Y, Fuery JJ, Parsons RJ, Walter RM 1973 Calusterone (7β,17α-dimethyltestosterone) as primary and secondary therapy of advanced breast cancer. Oncology 28:138-146 

Gordan GS 1976 Anabolic-androgenic steroids. In: Handbook of experimental pharmacology. New York: Springer-Verlag; 499-513 

Segaloff A1977 The use of androgens in the treatment of neoplastic disease. Pharm Ther C2:33-37 

Ingle JN, Twito DI, Schaid DJ, Cullinan SA, Krook JE, Mailliard JA, Tschetter LK, Long HJ, Gerstner JG, Windschitl HE, Levitt R, Pfeifle DM 1991 Combination hormonal therapy with tamoxifen plus fluoxymesterone vs. tamoxifen alone in postmenopausal women with metastatic breast cancer. A phase II study. Cancer 67:886-891 

Luo S, Sourla A, Labrie C, Belanger A, Labrie F 1997 Combined effects of dehydroepiandrosterone and EM-800 on bone mass, serum lipids, and the development of dimethylbenz(a)anthracene (DMBA)-induced mammary carcinoma in the rat. Endocrinology 138:4435-4444 

Sourla A, Martel C, Labrie C, Labrie F 1998 Almost exclusive androgenic action of dehydroepiandrosterone in the rat mammary gland. Endocrinology 139:753-764 

Trams G, Maass H 1977 Specific binding of estradiol and dihydrotestosterone in human mammary cancers. Cancer Res 37:258-261 

Allegra JC, Lippman ME, Thompson EB, Simon R, Green L, Barlock A, Green L, Huff KK, Do HM, Aitken SC 1979 Distribution, frequency and quantitative analysis of estrogen, progesterone, androgen and glucocorticoid receptors in human breast cancer. Cancer Res 39:1447-1454 

Miller WR, Telford J, Dixon JM, Hawkins RA 1985 Androgen receptor activity in human breast cancer and its relationship with estrogen and progesterone receptor activity. Eur J Cancer Clin Oncol 21:539-542 

Bryan RM, Mercer RJ, Bennett RC, Rennie GC, Lie TH, Morgan FJ 1984 Androgen receptors in breast cancer. Cancer 54:2436-2440 

Würz H, Schulz KO, Citoler P, Kaiser R 1982 Verteilung von ostrogen gestagen androgen uno kkotikosteroidrezeptoren in mammakarzinomen. In: Jonat W, Maass H, eds. Steroiohormonrez eptoren im karzinomgewebe. Stuttgart, Germany: Enke; 23-30 

Horwitz KB, Costlow ME, McGuire WL 1975 MCF-7: a human breast cancer cell line with estrogen, androgen, progesterone, and glucocorticoid receptors. Steroids 26:785-795 

Zava DT, McGuire WL 1978 Androgen action through estrogen receptor in a human breast cancer cell line. Endocrinology 103: 624-631 

Horwitz HB 1987 The structure and function of progesterone receptors in breast cancer. J Steroid Biochem 27:447-457 

Wooster R, Mangion J, Eeles R, Smith S, Dowsett M, Averill D, Barrett-Lee P, Easton DF, Ponder BA, Stratton MR 1992 A germline mutation in the androgen receptor in two brothers with breast cancer and Reifenstein syndrome. Nat Genet 2:132-134 

Lobaccaro JM, Lumbroso S, Belon C, Galtier-Dereure F, Bringer J, Lesimple T, Namer M, Cutuli BF, Pujol H, Sultan C 1993 Androgen receptor gene mutation in male breast cancer. Hum Mol Genet 2:1799-1802 

Bulbrook RD, Herian M, Tong D, Hayward JL, Swain MC, Wang DY 1973 Effect of steroidal contraceptives on levels of plasma androgen sulphates and cortisol. Lancet 1:628-631 

Juret P, Hayem M 1968 New biological evidence on the hormone dependence of breast cancer (preliminary note). Rev Fr Etud Clin Biol 13:884-887 

Masnyk IJ, Silverman DT, Hankey BF1978 Prediction of response to adrenalectomy in the treatment of advanced breast cancer. J Natl Cancer Inst 60:271-278 

Bulbrook RD, Hayward JL, Spicer CC 1971 Relation between urinary androgen and corticoid excretion and subsequent breast cancer. Lancet 2:395-398 

Brennan MJ, Wang DY, Hayward JL, Bulbrook RD, Deshpande N 1973 Urinary and plasma androgens in benign breast disease. Possible relation to breast cancer. Lancet 1:1076-1079 

Wang DY, Bulbrook RD, Hayward JL 1975 Urinary and plasma androgens and their relation to familial risk of breast cancer. Eur J Cancer 11:873-877 

Dorgan JF, Longcope C, Stephenson Jr HE, Falk RT, Miller R, Franz C, Kahle L, Campbell WS, Tangrea JA, Schatzkin A 1996 Relation of prediagnostic serum estrogen and androgen levels to breast cancer risk. Cancer Epidemiol Biomarkers Prev 5:533-539 

Berrino F, Muti P, Micheli A, Bolelli G, Krogh V, Sciajno R, Pisani P, Panico S, Secreto G 1996 Serum sex hormone levels after menopause and subsequent breast cancer. J Natl Cancer Inst 88:291-296 

Secreto G, TonioloP, Berrino F,RecchioneC, Di Pietro S,Fariselli G, Decarli A 1984 Increased androgenic activity and breast cancer risk in premenopausal women. Cancer Res 44:5902-5905 

Secreto G, Toniolo P, Berrino F, Recchione C, Cavalleri A, Pisani P, Toris A, Fariselli G, Di Pietro S 1991 Serum and urinary androgens and risk of breast cancer in postmenopausal women. Cancer Res 51:2572-2576 

Staiman VR, Lowe FC 1997 Tamoxifen for flutamide/finasterideinduced gynecomastia. Urology 50:929-933 

Rebbeck TR, Kantoff PW, Krithivas K, Neuhausen S, Blackwood MA, Godwin AK, Daly MB, Narod SA, Garber JE, Lynch HT, Weber BL, Brown M 1999 Modification of BRCA1-associated breast cancer risk by the polymorphic androgen-receptor CAG repeat. Am J Hum Genet 64:1371-1377 

Zhou J, Ng S, Adesanya-Famuiya O, Anderson K, Bondy CA 2000 Testosterone inhibits estrogen-induced mammary epithelial proliferation and suppresses estrogen receptor expression. FASEB J 14:1725-1730 

Park JJ, Irvine RA, Buchanan G, Koh SS, Park JM, Tilley WD, Stallcup MR, Press MF, Coetzee GA 2000 Breast cancer susceptibility gene 1 (BRCAI) is a coactivator of the androgen receptor. Cancer Res 60:5946-5949 

Korkia P, Stimson GV1997 Indications of prevalence, practice and effects of anabolic steroid use in Great Britain. Int J Sports Med 18:557-562 

Burgess HE, Shousha S 1993 An immunohistochemical study of the long-term effects of androgen administration on female-to-male transsexual breast: a comparison with normal female breast and male breast showing gynaecomastia. J Pathol 170:37-43 

Lacassagne A 1936 Hormonal pathogenesis of adenocarcinoma of the breast. Am J Cancer 27:217-228 

Costlow ME, Buschow RA, McGuire WL 1976 Prolactin receptors and androgen-induced regression of 7,12-dimethylbenz(a)anthracene-induced mammary carcinoma. Cancer Res 36:3324-3329 

Quadri SK, Kledzik GS, Meites J 1974 Counteraction by prolactin of androgen-induced inhibition of mammary tumor growth in rats. J Natl Cancer Inst 52:875-878 

Teller MN, Budinger JM, Zvilichovsky G, Watson AA, McDonald JJ, Stohrer G, Brown GB 1978 Oncogenicity of purine 3-oxide and unsubstituted purine in rats. Cancer Res 38:2229 -2232 

Poulin R, Baker D, Labrie F 1988 Androgens inhibit basal and estrogen-induced cell proliferation in the ZR-75-1 human breast cancer cell line. Breast Cancer Res Treat 12:213-225 

Poulin R, Baker D, Poirier D, Labrie F 1989 Androgen and glucocorticoid receptor-mediated inhibition of cell proliferation by medroxyprogesterone acetate in ZR-75-1 human breast cancer cells. Breast Cancer Res Treat 13:161-172 

Poulin R, Simard J, Labrie C, Petitclerc L, Dumont M, Lagace L, Labrie F1989 Down-regulation of estrogen receptors by androgens in the ZR-75-1 human breast cancer cell line. Endocrinology 125: 392-399 

Simard J, Hatton AC, Labrie C, Dauvois S, Zhao HF, Haagensen DE, Labrie F 1989 Inhibitory effects of estrogens on GCDFP-15 mRNA levels and secretion in ZR-75-1 human breast cancer cells. Mol Endocrinol 3:694-702 

Dumont M, Dauvois S, Simard J, Garcia T, Schachter B, Labrie F 1989 Antagonism between estrogens and androgens on GCDFP-15 gene expression in ZR-75-1 cells and correlation between GCDFP-15 and estrogen as well as progesterone receptor expression in human breast cancer. J Steroid Biochem 34:397-402 

Simard J, Dauvois S, Haagensen DE, Le vesque C, Me rand Y, Labrie F 1990 Regulation of progesterone-binding breast cyst protein GCDFP-24 secretion by estrogens and androgens in human breast cancer cells: a new marker of steroid action in breast cancer. Endocrinology 126:3223-3231 

Theriault C, Labrie F 1990 Hormonal regulation of estradiol 17β-hydroxysteroid dehydrogenase activity in the ZR-75-1 human breast cancer cell line. Ann NY Acad Sci 595:419-421 

Rochefort H, Garcia M 1983 The estrogenic and antiestrogenic activities of androgens in female target tissues. Pharmacol Ther 23:193-216 

Abraham GE 1974 Ovarian and adrenal contribution to peripheral androgens during the menstrual cycle. J Clin Endocrinol Metab 39:340-346 

Vermeulen A, Verdonck L 1979 Factors affecting sex hormone levels in postmenopausal women. J Steroid Biochem 11:899-904 

Mistry P, Griffiths K, Maynard PV1986 Endogenous C19-steroids and estradiol levels in human primary breast tumor tissues and their correlation with androgen and estrogen receptors. J Steroid Biochem 24:1117-1125 

Horwitz KB, Zava DT, Thilager AK, Jensen ET, McGuire WL1978 Steroid receptor analyses of nonhuman breast cancer cell lines. Cancer Res 38:2434-2439 

MacIndoe JH, Woods GR, Lee FJ 1981 The specific binding of androgens and the subsequent distribution of androgen receptor complexes within MCF-7 human breast cancer cells. Steroids 38: 439-452 

Chalbos D, Haagensen DE, Parish T, Rochefort H 1987 Identification and androgen regulation of two proteins released by T47D human breast cancer cells. Cancer Res 47:2787-2792 

Murphy LC, Tsuyuki D, Myal Y, Shiu RPC 1987 Isolation and sequencing of a cDNA clone for a prolactin-inducible protein (PIP). J Biol Chem 262:15236-15241 

Perel E, Daniilescu D, Kharlip L, Blackstein ME, Killinger DW 1985 The relationship between growth and androstenedione metabolism in four cell lines of human breast carcinoma cells in culture. Mol Cell Endocrinol 41:197-203 

Griffiths K, Jones D, Cameron EHD, Gleave EN, Forrest APM 1972 Transformation of steroids by mammary cancer tissue. In: Dao TL, ed. Estrogen target tissues and neoplasia. Chicago: University of Chicago Press; 151-162 

Perel E, Killinger DW 1983 The metabolism of androstenedione and testosterone of C10-metabolites in breast carcinoma, andbenign prostatic hypertrophy tissue. J Steroid Biochem 19:1135-1139 

Neri R, Peets E, Watnick A 1979 Anti-androgenicity of flutamide and its metabolite Sch 16423. Biochem Soc Trans 7:565-569 

Simard J, Luthy I, Guay J, Belanger A, Labrie F 1986 Characteristics of interaction of the antiandrogen flutamide with the androgen receptor in various target tissues. Mol Cell Endocrinol 44:261-270 

Le vesque C, Me rand Y, Dufour JM, Labrie C, Labrie F 1991 Synthesis and biological activity of new halo-steroidal antiestrogens. J Med Chem 34:1624-1630 

de Launoit Y, Veilleux R, Dufour M, Simard J, Labrie F 1991 Characteristics of the biphasic action of androgens and of the potent antiproliferative effects of the new pure antiestrogen EM-139 on cell cycle kinetic parameters in LNCaP human prostatic cancer cells. Cancer Res 51:5165-5170 

Dauvois S, Geng CS, Le vesque C, Merand Y, Labrie F 1991 Additive inhibitory effects of an androgen and the antiestrogen EM170 on estradiol-stimulated growth of human ZR-75-1 breast tumors in athymic mice. Cancer Res 51:3131-3135 

Poulin R, Baker D, Poirier D, Labrie F 1990 Multiple actions of synthetic "progestins" on the growth of ZR-75-1 human breast cancer cells: an in vitro model for the simultaneous assay of androgen, progestin, estrogen and glucocorticoid agonistic and antagonistic activities of compounds. Breast Cancer Res Treat 17:197-210 

Cusan L, Dupont A, Cossette M, Labrie F 1993 Flutamide in the treatment of female androgenic alopecia. CanJ Dermatol 5:421-427 

Cheng Y, Prusoff WH 1973 Relationship between the inhibition constant (Ki) and the concentration of inhibitor which causes 50 per cent inhibition (I50) on an enzymatic reaction. Biochem Pharmacol 22:3099-3108 

Lippman ME, Bolan G, Huff K 1976 The effect of androgens and antiandrogens on hormone-responsive human breast cancer in long-term tissue culture. Cancer Res 36:4610-4618 

Zava DT, McGuire WL 1977 Estrogen receptors in androgen-induced breast tumor regression. Cancer Res 37:1608-1610 

Garcia M, Rochefort H 1978 Androgen effects mediated by estrogen receptor in 7,12-dimethylbenz(a)anthracene-induced rat mammary tumors. Cancer Res 38:3922-3929 

Zava DT, McGuire WL 1978 Human breast cancer: androgen action mediated by estrogen receptor. Science 199:787-788 

Kasid A, Strobl JS, Huff K, Greene GL, Lippman ME 1984 A novel nuclear form of estradiol receptor in MCF-7 human breast cancer cells. Science 225:1162-1165 

Engel LW, Young NA,TralkaTS, Lippman ME, O'Brien SJ, Joyce MJ 1978 Establishment and characterization of three new continuous cell lines derived from human breast carcinomas. Cancer Res 38:3352-3364 

MacIndoe JH, Etre LA 1981 An antiestrogenic action of androgens in human breast cancer cells. J Clin Endocrinol Metab 53:836-842 

Poulin R, Merand Y, Poirier D, Levesque C, Dufour JM, Labrie F 1989 Antiestrogenic properties of keoxifene trans-4-hydroxytamoxifen, and ICI 164384, a new steroidal antiestrogen, in ZR-75-1 human breast cancer cells. Breast Cancer Res Treat 14:65-76 

Dauvois S, Li S, Martel C, Labrie F 1989 Inhibitory effect of androgens on DMBA-induced mammary carcinoma in the rat. Breast Cancer Res Treat 14:299-306 

Li S, Lepage M, Me rand Y, Belanger A, Labrie F 1992 Growth inhibition of 7,12-dimethylbenz(a)anthracene-induced rat mammary tumors by controlled-release low-dose medroxyprogesterone acetate. Breast Cancer Res Treat 24:127-137 

Labrie F, Li S, Belanger A, Cote J, Merand Y, Lepage M 1993 Controlled release low dose medroxyprogesterone acetate (MPA) inhibits the development of mammary tumors induced by dimethylbenz(a)anthracene in the rat. Breast Cancer Res Treat 26: 253-265 

Bentel JM, Birrell SN, Pickering MA, Holds DJ, Horsfall DJ, Tilley WD 1999 Androgen receptor agonist activity of the synthetic progestin, medroxyprogesterone acetate, in human breast cancer cells. Mol Cell Endocrinol 154:11-20 

Birrell SN, Hall RE, Tilley WD 1998 Role of the androgen receptor in human breast cancer. J Mammary Gland Biol Neoplasia 3:95-103 

Birrell SN, Roder DM, Horsfall DJ, Bentel JM, Tilley WD 1995 Medroxyprogesterone acetate therapy in advanced breast cancer: the predictive value of androgen receptor expression. J Clin Oncol 13:1572-1577 

Jayo MJ, Register TC, Hughes CL, Blas-Machado U, Sulistiawati E, Borgerink H, Johnson CS 2000 Effects of an oral contraceptive combination with or without androgen on mammary tissues: a study in rats. J Soc Gynecol Investig 7:257-265 

Labrie C, Simard J, Zhao HF, Belanger A, Pelletier G, Labrie F 1989 Stimulation of androgen-dependent gene expression by the adrenal precursors dehydroepiandrosterone and androstenedione in the rat ventral prostate. Endocrinology 124:2745-2754 

Nathanson IT 1947 Endocrine aspects of human cancer. Recent Prog Res 1:261-291 

Adair FE, Herrmann JB 1946 The use of testosterone propionate in the treatment of advanced carcinoma of the breast. Ann Surg 123: 1023-1035 

Adair FE, Mellors RC, Farrow JH, Voodard HQ, Escher GS, Urban JA1949 The use of estrogens and androgens in advanced mammary cancer. JAMA 15:1193-2000 

Poulin R, Labrie F 1986 Stimulation of cell proliferation and estrogenic response by adrenal C19-Δ5-steroids in the ZR-75-1 human breast cancer cell line. Cancer Res46:4933-4937 

Labrie C, Flamand M, Belanger A, Labrie F 1996 High bioavailability of DHEA administered percutaneously in the rat. J Endocrinol 150:S107-S118 

Labrie F, Be langer A, Simard J, Luu-The V, Labrie C 1995 DHEA and peripheral androgen and estrogen formation: intracrinology. Ann NY Acad Sci 774:16-28 

Asselin J, Labrie F1978 Effects of estradiol and prolactin on steroid receptor levels in 7,12-dimethylbenz(a)anthracene-induced mammary tumors and uterus in the rat. J Steroid Biochem 9:1079-1082 

Asselin J, Kelly PA, Caron MG, Labrie F 1977 Control of hormone receptor levels and growth of 7,12-dimethylbenz(a)anthraceneinduced mammary tumors by estrogens, progesterone and prolactin. Endocrinology 101:666-671 

Li S, Yan X, Belanger A, Labrie F 1993 Prevention by dehydroepiandrosterone of the development of mammary carcinoma induced by 7,12-dimethylbenz(a)anthracene (DMBA) in the rat. Breast Cancer Res Treat 29:203-217 

Liu CH, Laughlin GA, Fischer UG, Yen SS 1990 Marked attenuation of ultradian and circadian rhythms of dehydroepiandrosterone in postmenopausal women: evidence for a reduced 17,20desmolase enzymatic activity. J Clin Endocrinol Metab 71:900 -906 

Martel C, Rheaume E, Takahashi M, Trudel C, Couet J, Luu-The V, Simard J, Labrie F 1992 Distribution of 17β-hydroxysteroid dehydrogenase gene expression and activity in rat and human tissues. J Steroid Biochem Mol Biol 41:597-603 

Labrie F, Simard J, Luu-The V, Pelletier G, Belanger A, Lachance Y, Zhao HF, Labrie C, Breton N, de Launoit Y, Dumont M, Dupont E, Rheaume E, Martel C, Couet J, Trudel C 1992 Structure and tissue-specific expression of 3β-hydroxysteroid dehydrogenase/5ene-4-ene isomerase genes in human and rat classical and peripheral steroidogenic tissues. J Steroid Biochem Mol Biol 41:421-435 

Hackenberg R, Luttchens S, Hofmann J, Kunzmann R, Holzel F, Schulz KD 1991 Androgen sensitivity of the new human breast cancer cell line MFM-223. Cancer Res51:5722-5727 

de Launoit Y, Dauvois S, Dufour M, Simard J, Labrie F 1991 Inhibition of cell cycle kinetics and proliferation by the androgen 5α-dihydrotestosterone and antiestrogen N, n-butyl-N-methyl-11(16'a-chloro-3',17β-dihydroxy-estra-1',3',5'-(10')triene-7'a-yl)undecanamide in human breast cancer ZR-75-1 cells. Cancer Res 51:2797-2802 

Dauvois S, Spinola PG, Labrie F 1989 Additive inhibitory effects of bromocriptine (CB-154) and medroxyprogesterone acetate (MPA) on dimethylbenz(a)anthracene (DMBA)-induced mammary tumors in the rat. Eur J Cancer Clin Oncol 25:891-897 

Couillard S, Labrie C, Belanger A, Candas B, Pouliot F, Labrie F 1998 Effect of dehydroepiandrosterone and the antiestrogen EM800 on the growth of human ZR-75-1 breast cancer xenografts. J Natl Cancer Inst 90:772-778 

Nicholson RI, Davies P, Griffiths K 1978 Interaction of estrogens with estradiol-17β- receptor proteins in DMBA-induced mammary tumors. A possible oncolytic mechanism. Eur J Cancer 14:439-445 

Kreitmann B, Bayard F 1979 Androgen interaction with the oestrogen receptor in human tissues. J Steroid Biochem 11:1589-1595 

Adams JB, Archibald L, Seymour-Munn K 1980 Dehydroepiandrosterone and androst-5-ene-3β,17β-diol in human mammary cancer cytosolic and nuclear compartments and their relationship to estrogen receptor. Cancer Res 40:3815-3820 

Poortman J, Prenen JA, Schwarz F, Thijssen JH 1975 Interaction of A-5-androstene-3β, 17β-diol with estradiol and dihydrotestosterone receptors in human myometrial and mammary cancer tissue. J Clin Endocrinol Metab 40:373-379 

Boccuzzi G, Aragno M, Brignardello E, Tamagno E, Conti G, Di Monaco M, Racca S, Danni O, Di Carlo F 1992 Opposite effects of dehydroepiandrosterone on the growth of 7,12-dimethylbenz(a)anthracene-induced rat mammary carcinomas. Anticancer Res 12: 1479-1484 

Luo S, Martel C, Leblanc G, Candas B, Singh SM, Labrie C, Simard J, Belanger A, Labrie F1996 Relative potencies of flutamide and casodex: preclinical studies. Endocrine-Related Cancer 3:229-241 

Poortman J, Thijssen JH, von Landeghem AA, Wiegerinck MA, Alsbach GP 1983 Subcellular distribution of androgens and oestrogens in target tissue. J Steroid Biochem 19:939-945 

Najid A, Ratinaud MH 1991 Comparative studies of steroidogenesis inhibitors (econazole, ketoconazole) on human breast cancer MCF-7 cell proliferation by growth experiments, thymidine incorporation and flow cytometric DNA analysis. Tumori 77:385-390 

Boccuzzi G, Di Monaco M, Brignardello E, Leonardi L, Gatto V, Pizzini A, Gallo M 1993 Dehydroepiandrosterone antiestrogenic action through androgen receptor in MCF-7 human breast cancer cell line. Anticancer Res 13:2267-2272 

Jordan VC 1976 Effect of tamoxifen (ICI 46,474) on initiation and growth of DMBA-induced rat mammary carcinoma. Eur J Cancer 12:419-424 

Jordan VC 1978 Use of the DMBA-induced rat mammary carcinoma system for the evaluation of tamoxifen as a potential adjuvant therapy. Rev Endocr Relat Cancer(Suppl):49-55 

Kawamura I, Mizota T, Kondo N, Shimomura K, Kohsaka M 1991 Antitumor effects of droloxifene, a new antiestrogen drug, against 7,12-dimethylbenz(a)anthracene-induced mammary tumors in rats. Jpn J Pharmacol57:215-224 

Labrie F, Li S, Labrie C, Levesque C, Me rand Y 1995 Inhibitory effect of a steroidal antiestrogen (EM-170) on estrone-stimulated growth of 7,12 dimethylbenz(a)anthracene (DMBA)-induced mammary carcinoma in the rat. Breast Cancer Res Treat 33:237-244 

Luo S, Labrie C, Belanger A, Labrie F 1997 Effect of dehydroepiandrosterone on bone mass, serum lipids, and dimethylbenz(a)anthracene-induced mammary carcinoma in the rat. Endocrinology 138:3387-3394 

Labrie F, Diamond P, Cusan L, Gomez JL, Belanger A 1997 Effect of 12-month DHEA replacement therapy on bone, vaginum, and endometrium in postmenopausal women. J Clin Endocrinol Metab 82:3498-3505 

Zumoff B, Levin J, Rosenfeld RS, Markham M, Strain GW, Fukushima DK 1981 Abnormal 24-hr mean plasma concentrations of dehydroepiandrosterone and dehydroisoandrosterone sulfate in women with primary operable breast cancer. Cancer Res 41:3360 3363 

Segaloff A, Hayward BF, Carter AC, Bundy B, Masnyk IJ 1980 Identification of breast cancer patients with high risk of early recurrence after radical mastectomy. III. Steroid hormones measured in urine. Cancer 46:1087-1092 

Bulbrook RD, Hayward JL, Spicer CC, Thomas BS1962 Abnormal excretion of urinary steroids by women with early breast cancer. Lancet 2:1238-1240 

Cameron EHD, Griffiths K, Gleave EN, Stewart HJ, Forrest APM, Campbell H 1970 Benign and malignant breast disease in south Wales: a study of urinary steroids. Br Med J 4:768-771 

Schwartz AG 1979 Inhibition of spontaneous breast cancer formation in female C3H (Avy/a) mice by long-term treatment with dehydroepiandrosterone. Cancer Res 39:1129-1132 

Gordon GB, Shantz LM, Talalay P 1987 Modulation of growth, differentiation and carcinogenesis by dehydroepiandrosterone. Adv Enzyme Regul 26:355-382 

Schwartz AG, Pashko L, Whitcomb JM 1986 Inhibition of tumor development by dehydroepiandrosterone and related steroids. Toxicol Pathol 14:357-362 

Melvin WS, Boros LG, Muscarella P, Brandes JL, Johnson JA, Fisher WE, Schirmer WJ, Ellison EC 1997 Dehydroepiandrosterone-sulfate inhibits pancreatic carcinoma cell proliferation in vitro and in vivo. Surgery 121:392-397 

Schulz S, Klann RC, Schonfeld S, Nyce JW 1992 Mechanisms of cell growth inhibition and cell cycle arrest in human colonic adenocarcinoma cells by dehydroepiandrosterone: role of isoprenoid biosynthesis. Cancer Res 52:1372-1376 

Stahl F, Schnorr D, Pilz C, Dorner G 1992 Dehydroepiandrosterone (DHEA) levels in patients with prostatic cancer, heart diseases and under surgery stress. Exp Clin Endocrinol 99:68-70 

Heinonen PK, Koivula T, Pystynen P 1987 Decreased serum level of dehydroepiandrosterone sulfate in postmenopausal women with ovarian cancer. Gynecol Obstet Invest 23:271-274 

Luu-The V, Dufort I, Paquet N, Reimnitz G, Labrie F 1995 Structural characterization and expression of the human dehydroepiandrosterone sulfotransferase gene. DNA Cell Biol 14:511-518 

Deleted in proof Labrie F, Luu-The V, Lin S-X, Simard J, Labrie C 2000 Role of 17β-hydroxysteroid dehydrogenases in sex steroid formation in peripheral intracrine tissues. Trends Endocrinol Metab 11:421-427 

Gower BA, Nyman L 2000 Associations among oral estrogen use, free testosterone concentration, and lean body mass among postmenopausal women. J Clin Endocrinol Metab 85:4476-4480 

Martel C, Labrie F, Important androgenic component in the stimulatory effect of dehydroepiandrosterone (DHEA) on bone density in the rat. Proc 8th International Congress on the Menopause, Sydney, Australia, 1996 

Diamond P, Cusan L, Gomez JL, Belanger A, Labrie F 1996 Metabolic effects of 12-month percutaneous DHEA replacement therapy in postmenopausal women. J Endocrinol 150:S43-S50 

Morales AJ, Nolan JJ, Nelson JC, Yen SS 1994 Effects of replacement dose of dehydroepiandrosterone in men and women of advancing age. J Clin Endocrinol Metab 78:1360-1367 

Stomati M, Rubino S, Spinetti A, Parrini D, Luisi S, Casarosa E, Petraglia F, Genazzani AR 1999 Endocrine, neuroendocrine and behavioral effects of oral dehydroepiandrosterone sulfate supplementation in postmenopausal women. Gynecol Endocrinol 13:15-25 

Arlt W, Justl HG, Callies F, Reincke M, Hubler D, Oettel M, Ernst M, Schulte HM, Allolio B 1998 Oral dehydroepiandrosterone for adrenal androgen replacement: pharmacokinetics and peripheral conversion to androgens and estrogens in young healthy females after dexamethasone suppression. J Clin Endocrinol Metab 83:1928-1934 

Mortola JF, Yen SS 1990 The effects of oral dehydroepiandrosterone on endocrine-metabolic parameters in postmenopausal women. J Clin Endocrinol Metab 71:696-704 

Berr C, Lafont S, Debuire B, Dartigues JF, Baulieu EE 1996 Relationships of dehydroepiandrosterone sulfate in the elderly with functional, psychological, and mental status, and short-term mortality: a French community-based study. Proc Natl Acad Sci USA 93:13410-13415 

Ravaglia G, Forti P, Maioli F, Boschi F, Bernardi M, Pratelli L, Pizzoferrato A, Gasbarrini G 1996 The relationship of dehydroepiandrosterone sulfate (DHEAS) to endocrine-metabolic parameters and functional status in the oldest-old. Results from an Italian study on healthy free-living over-ninety-year-olds. J Clin Endocrinol Metab 81:1173-1178 

Wolf OT, Neumann O, Hellhammer DH, Geiben AC, Strasburger CJ, Dressendorfer RA, Pirke KM, Kirschbaum C 1997 Effects of a two-week physiological dehydroepiandrosterone substitution on cognitive performance and well-being in healthy elderly women and men. J Clin Endocrinol Metab 82:2363-2367 

Wolkowitz OM, Reus VI, Roberts E, Manfredi F, Chan T, Raum WJ, Ormiston S, Johnson R, Canick J, Brizendine L, Weingartner H 1997 Dehydroepiandrosterone (DHEA) treatment of depression. Biol Psychiatry 41:311-318 

Taelman P, Kaufman JM, Janssens X, Vermeulen A 1989 Persistence of increased bone resorption and possible role of dehydroepiandrosterone as a bone metabolism determinant in osteoporotic women in late post-menopause. Maturitas 11:65-73 

Schriock ED, Buffington CK, Hubert GD, Kurtz BR, Kitabchi AE, Buster JE, Givens JR 1988 Divergent correlations of circulating dehydroepiandrosterone sulfate and testosterone with insulin levels and insulin receptor binding. J Clin Endocrinol Metab 66:1329-1331 

Coleman DL, Leiter EH, Schwizer RW 1982 Therapeutic effects of dehydroepiandrosterone (DHEA) in diabetic mice. Diabetes 31: 830-833 

Nestler JE, Barlascini CO, Clore JN, Blackard WG 1988 Dehydroepiandrosterone reduces serum low density lipoprotein levels and body fat but does not alter insulin sensitivity in normal men. J Clin Endocrinol Metab 66:57-61 

MacEwen EG, Kurzman ID 1991 Obesity in the dog: role of the adrenal steroid dehydroepiandrosterone (DHEA). J Nutr 121:S51-S55 

Tchernof A, Despres JP, Belanger A, Dupont A, Prud'homme D, Moorjani S, Lupien PJ, Labrie F 1995 Reduced testosterone and adrenal C19 steroid levels in obese men. Metabolism 44:513-519 

Haffner SM, Valdez RA, Mykkanen L, Stern MP, Katz MS 1994 Decreased testosterone and dehydroepiandrosterone sulfate concentrations are associated with increased insulin and glucose concentrations in nondiabetic men. Metabolism 43:599-603 

Suzuki T, Suzuki N, Daynes RA, Engleman EG 1991 Dehydroepiandrosterone enhances IL2 production and cytotoxic effector function of human T cells. Clin Immunol Immunopathol 61:202-211 

Rasmussen KR, Arrowood MJ, Healey MC 1992 Effectiveness of dehydroepiandrosterone in reduction of cryptosporidial activity in immunosuppressed rats. Antimicrob Agents Chemother 36:220-222 

Henderson E, Yang JY, Schwartz A 1992 Dehydroepiandrosterone (DHEA) and synthetic DHEA analogs are modest inhibitors of HIV-1 IIIB replication. AIDS Res Hum Retroviruses 8:625-631 

Casson PR, Andersen RN, Herrod HG, Stentz FB, Straughn AB, Abraham GE, Buster JE 1993 Oral dehydroepiandrosterone in physiologic doses modulates immune function in postmenopausal women. Am J Obstet Gynecol 169:1536-1539 

Johnston Jr CC, Epstein S1981 Clinical, biochemical, radiographic, epidemiologic, and economic features of osteoporosis. Orthop Clin North Am 12:559-569 

Bardon S, Vignon F, Chalbos D, Rochefort H 1985 RU486, a progestin and glucocorticoid antagonist, inhibits the growth of breast cancer cells via the progesterone receptor. J Clin Endocrinol Metab 60:692-697 

Cromer BA, Blair JM, Mahan JD, Zibners L, Naumovski Z 1996 A prospective comparison of bone density in adolescent girls receiving depot medroxyprogesterone acetate (Depo-Provera), levonorgestrel (Norplant), or oral contraceptives. J Pediatr 129: 671-676 

Urban RJ, Bodenburg YH, Gilkinson C, Foxworth J, Coggan AR, Wolfe RR, Ferrando A 1995 Testosterone administration to elderly men increases skeletal muscle strength and protein synthesis. Am J Physiol 269:E820-E826 

Simon D, Preziosi P, Barrett-Connor E, Roger M, Saint-Paul M, Nahoul K, Papoz L 1992 The influence of aging on plasma sex hormones in men: the Telecom study. Am J Epidemiol 135:783-791 

Turcato E, Zamboni M, De Pergola G, Armellini F, Zivelonghi A, Bergamo-Andreis IA, Giorgino R, Bosello O 1997 Interrelationships between weight loss, body fat distribution and sex hormones in pre- and postmenopausal obese women. J Intern Med 241:363-372 

Poehlman ET, Goran MI, Gardner AW, Ades PA, Arciero PJ, Katzman-Rooks SM, Montgomery SM, Toth MJ, Sutherland PT 1993 Determinants of decline in resting metabolic rate in aging females. Am J Physiol 264:E450—E455 

Poehlman ET, Toth MJ, Gardner AW 1995 Changes in energy balance and body composition at menopause: a controlled longitudinal study. Ann Intern Med 123:673-675 

Guralnik JM, Ferrucci L, Simonsick EM, Salive ME, Wallace RB 1995 Lower-extremity function in persons over the age of 70 years as a predictor of subsequent disability. N Engl J Med 332:556-561 

Wolfson L, Judge J, Whipple R, King M 1995 Strength is a major factor in balance, gait, and the occurrence of falls. J Gerontol A Biol Sci Med Sci 50(Spec No):64-67 

van Vollenhoven RF, Engleman EG, McGuire JL 1995 Dehydroepiandrosterone in systemic lupus erythematosus. Results of a double-blind, placebo-controlled, randomized clinical trial. Arthritis Rheum 38:1826-1831 

van Vollenhoven RF, Park JL, Genovese MC, West JP, McGuire JL 1999 A double-blind, placebo-controlled, clinical trial of dehydroepiandrosterone in severe systemic lupus erythematosus. Lupus 8:181-187 

Sugino M, Ohsawa N, Ito T, Ishida S, Yamasaki H, Kimura F, Shinoda K 1998 A pilot study of dehydroepiandrosterone sulfate in myotonic dystrophy. Neurology 51:586-589 

Flynn MA, Weaver-Osterholtz D, Sharpe-Timms KL, Allen S, Krause G 1999 Dehydroepiandrosterone replacement in aging humans. J Clin Endocrinol Metab 84:1527-1533 

Wolkowitz OM, Reus VI, Keebler A, Nelson N, Friedland M, Brizendine L, Roberts E 1999 Double-blind treatment of major depression with dehydroepiandrosterone. Am J Psychiatry 156:646-649 

Bloch M, Schmidt PJ, Danaceau MA, Adams LF, Rubinow DR 1999 Dehydroepiandrosterone treatment of midlife dysthymia. Biol Psychiatry 45:1533-1541 

Barnhart KT, Freeman E, Grisso JA, Rader DJ, Sammel M, Kapoor S, Nestler JE 1999 The effect of dehydroepiandrosterone supplementation to symptomatic perimenopausal women on serum endocrine profiles, lipid parameters, and health-related quality of life. J Clin Endocrinol Metab 84:3896-3902 

Welle S, Jozefowicz R, Statt M 1990 Failure of dehydroepiandrosterone to influence energy and protein metabolism in humans. J Clin Endocrinol Metab 71:1259 -1264 

Young J, CouzinetB,Nahoul K,Brailly S, Chanson P, Baulieu EE, Schaison G 1997 Panhypopituitarism as a model to study the metabolism of dehydroepiandrosterone (DHEA) in humans. J Clin Endocrinol Metab 82: 2578-2585 

Labrie F, Luu-The V, Labrie C, Simard J 2001 DHEA and its transformation into androgens and estrogens in peripheral target tissues: intracrinology. Front Neuroendocrinol 22:185-212