Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Horm Mol Biol Clin Investig ; 25(3): 181-90, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26725467

ABSTRACT

OBJECTIVE: Intravaginal DHEA (dehydroepiandrosterone, prasterone), the exclusive precursor of androgens and estrogens in postmenopausal women, has previously been shown to improve all the domains of sexual function by a strictly local action in the vagina. The well recognized female sexual function index (FSFI) questionnaire was used in the present study. DESIGN: The long-term effect of 52-week treatment with daily intravaginal 0.50% (6.5 mg) DHEA was evaluated on the various domains of female sexual function using the FSFI questionnaire at baseline, Week 26 and Week 52. SUBJECTS: One hundred and fifty-four postmenopausal women with at least one mild to severe symptom of vulvovaginal atrophy (VVA) and who have completed the FSFI questionnaire at baseline and at least one post-baseline timepoint were included in the analysis. RESULTS: The FSFI domains desire, arousal, lubrication, orgasm, satisfaction and pain were increased by 28%, 49%, 115%, 51%, 41% and 108%, respectively (p<0.0001 for all parameters) at 52 weeks vs. baseline, while the total score was increased from 13.4±0.62 at baseline to 21.5±0.82 (+60%, p<0.0001) at 52 weeks. CONCLUSION: As the serum levels of DHEA and all its metabolites, including estradiol and testosterone, show no meaningful change, the present clinical data indicate a stimulatory effect of intravaginal DHEA through a strictly local action in agreement with the preclinical data showing that the androgens made locally from DHEA in the vagina induce an increase in local nerve density.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Postmenopause/drug effects , Sexual Dysfunction, Physiological/drug therapy , Administration, Intravaginal , Adult , Aged , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/adverse effects , Double-Blind Method , Female , Humans , Middle Aged , Orgasm/drug effects , Placebo Effect , Surveys and Questionnaires
2.
Menopause ; 22(12): 1289-95, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25968836

ABSTRACT

OBJECTIVE: This study aims to evaluate the effects of intravaginal dehydroepiandrosterone (DHEA, prasterone) on the endometrium in postmenopausal women. METHODS: Intravaginal DHEA (6.5 mg) was administered daily for 52 weeks to 422 women who had endometrial biopsy at baseline and end of study, whereas 15 women were similarly treated for 26 to 52 weeks. Participants in three other studies received 3.25 mg (n = 126), 6.5 mg (n = 129), or 13 mg (n = 30) of DHEA for 12 weeks; women similarly had baseline and end-of-study biopsies. Endometrial biopsy samples were available for 668 women at baseline and end of study, with sufficient material for analysis. RESULTS: Endometrial atrophy or inactive endometrium (668 women) was found in all women treated with intravaginal DHEA. Similar atrophy was observed in 119 of 121 participants with sufficient material for analysis who received placebo. CONCLUSIONS: After cessation of estradiol secretion by the ovaries at menopause, the estrogens made by mechanisms of intracrinology are inactivated intracellularly at their site of formation and action, thus maintaining serum estradiol at biologically inactive concentrations to avoid stimulation of the endometrium. The absence of enzymes that are able to transform DHEA into estrogens in the endometrium explains the typical endometrial atrophy in all normal postmenopausal women in the presence of variable concentrations of circulating endogenous DHEA. According to these mechanisms, the inactive sex steroid precursor DHEA administered intravaginally acts exclusively in the vagina, whereas all serum sex steroids remain well within the biologically inactive postmenopausal reference range, thus avoiding any stimulation of the already atrophic endometrium.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Endometrium/drug effects , Postmenopause/physiology , Administration, Intravaginal , Adult , Aged , Atrophy , Biopsy , Dehydroepiandrosterone/adverse effects , Endometrium/pathology , Female , Humans , Middle Aged , Placebos , Vagina/pathology , Vulva/pathology
3.
Menopause ; 22(9): 950-63, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25734980

ABSTRACT

OBJECTIVE: This study aims to confirm the local effects of intravaginal prasterone on moderate to severe dyspareunia, a symptom of vulvovaginal atrophy (VVA) associated with menopause. METHODS: In a prospective, randomized, double-blind, placebo-controlled phase III clinical trial, we examined the effects of daily intravaginal prasterone (6.5 mg) on four co-primary objectives, namely, percentage of vaginal parabasal cells, percentage of vaginal superficial cells, vaginal pH, and moderate to severe dyspareunia identified by women as the most bothersome VVA symptom. RESULTS: After daily intravaginal prasterone administration for 12 weeks, the percentage of parabasal cells decreased by 45.8% compared with placebo (P < 0.0001), the percentage of superficial cells increased by 4.7% over placebo (P < 0.0001), and vaginal pH decreased by 0.83 pH units compared with placebo (P < 0.0001). The severity of most bothersome dyspareunia decreased by 46% over placebo (P = 0.013) at 12 weeks, whereas moderate to severe vaginal dryness decreased by 0.43 severity score units (or 42%) compared with placebo (P = 0.013). On gynecologic evaluation, a 14.4% to 21.1% improvement in vaginal secretions, epithelial integrity, epithelial surface thickness, and color over placebo (P = 0.0002 to P < 0.0001) was observed. Serum steroids, in agreement with the physiology of intracrinology and menopause, remained well within reference postmenopausal concentrations. All endometrial biopsies at 12 weeks have shown atrophy. CONCLUSIONS: Daily intravaginal prasterone (0.50%; 6.5 mg) treatment has clinically and statistically significant beneficial effects on the four co-primary objectives of VVA, according to US Food and Drug Administration guidelines. No significant drug-related adverse effect in line with the strictly local action of treatment has been reported, thus providing a high benefit-to-risk ratio for intravaginal prasterone.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Dyspareunia/drug therapy , Menopause , Administration, Intravaginal , Adult , Aged , Dehydroepiandrosterone/administration & dosage , Double-Blind Method , Dyspareunia/pathology , Female , Humans , Middle Aged , Pain Measurement , Prospective Studies , Treatment Outcome
4.
Maturitas ; 81(1): 46-56, 2015 May.
Article in English | MEDLINE | ID: mdl-25771041

ABSTRACT

OBJECTIVE: An objective was to analyze the time course of efficacy of daily intravaginal administration of 0.5% (6.5mg) DHEA (prasterone) for 52 weeks on the moderate to severe (MS) symptoms and signs of vulvovaginal atrophy (VVA). METHOD: Five hundred twenty-one postmenopausal women were enrolled and received daily intravaginal administration of 0.5% DHEA in an open-label phase III study. The severity of the VVA symptoms examined in detail in the different groups. RESULTS: A parallel improvement of pain at sexual activity was observed in women who had moderate to severe (MS) dyspareunia as their most bothersome symptom (MBS) (n=183) or not MBS (n=240) and MS without being MBS (n=57) with a 1.70 severity unit change in the MBS group for a decrease of 66.1% from baseline (p<0.0001 versus baseline) over 52 weeks. A further improvement of dyspareunia, namely 0.33 severity unit (19.4%), was observed with continuing treatment from 12 weeks to 52 weeks. Similar results were observed on vaginal dryness and irritation/itching. Highly significant beneficial effects (p<0.0001 versus baseline for all) were observed at gynecological examination on vaginal secretions, color, epithelial integrity and epithelial surface thickness. CONCLUSION: The present study shows, in addition to the parallel benefits on the three symptoms of VVA, that the choice of any of the MS symptoms as being or not being MBS by women has no influence on the observed therapeutic effect (NCT01256671).


Subject(s)
Dehydroepiandrosterone/administration & dosage , Hormones/administration & dosage , Vagina/pathology , Vaginal Diseases/drug therapy , Vulva/pathology , Vulvar Diseases/drug therapy , Administration, Intravaginal , Adult , Aged , Atrophy/complications , Atrophy/drug therapy , Double-Blind Method , Dyspareunia/drug therapy , Dyspareunia/etiology , Female , Humans , Middle Aged , Postmenopause , Pruritus/drug therapy , Severity of Illness Index , Sexual Behavior , Vaginal Diseases/complications , Vulvar Diseases/complications
5.
J Sex Med ; 11(7): 1766-85, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24774442

ABSTRACT

INTRODUCTION: We have previously observed that intravaginal prasterone (dehydroepiandrosterone, DHEA) improved all domains of female sexual dysfunction (FSD). AIM: Investigate the influence of moderate/severe pain at sexual activity (dyspareunia) (MSD) at baseline on FSD following prasterone administration. METHODS: The effect of daily administration of prasterone (0, 3.25 mg, 6.5 mg or 13 mg) for 12 weeks on FSD in 215 postmenopausal women with or without MSD at baseline was evaluated in a prospective, randomized, double-blind, and placebo-controlled phase III clinical trial. MAIN OUTCOME MEASURES: Differences were examined on desire, arousal and orgasm. RESULTS: Comparable benefits were observed in women not having MSD (n = 56) vs. those having MSD (n = 159). The benefits over placebo in prasterone-treated women for desire, avoiding intimacy and vaginal dryness as well as for the total sexual domain of the MENQOL (Menopause Specific Quality of Life) questionnaire, ranged between 18.0% and 38.2% with P values of <0.05 or <0.01 except in one out of 12 subgroups. For the arousal/sensation, arousal/lubrication and summary score of the ASF (Abbreviated Sexual Function) questionnaire, in the MSD+ group, improvements of 64.2% (P = 0.01), 118% (P = 0.001) and 31.1% (P = 0.03) were observed over placebo, respectively, while similar differences (58.0%, 67.6% and 32.1%) did not reach statistical significance in the MSD- group having up to only 44 prasterone-treated women compared with 119 in the MSD+ group. CONCLUSIONS: No MSD at baseline does not apparently affect the effects of intravaginal prasterone on sexual dysfunction. Knowing the absence of significant effects of estrogens on FSD, the present data suggest that vulvovaginal atrophy (VVA) and vulvovaginal sexual dysfunction (VVSD) are two different consequences of sex steroid deficiency at menopause which can respond independently. In addition, the present data seriously question the justification of pain being part of FSD as well as the separation of FSD into separate domains.


Subject(s)
Androgens/administration & dosage , Dehydroepiandrosterone/administration & dosage , Dyspareunia/complications , Sexual Dysfunctions, Psychological/drug therapy , Administration, Intravaginal , Adult , Aged , Arousal/drug effects , Double-Blind Method , Estrogens/therapeutic use , Female , Humans , Menopause , Middle Aged , Nerve Fibers/drug effects , Orgasm/drug effects , Postmenopause/drug effects , Prospective Studies , Quality of Life , Sexual Behavior/drug effects , Suppositories , Surveys and Questionnaires , Vagina/innervation , Vaginal Diseases/drug therapy
6.
J Steroid Biochem Mol Biol ; 138: 359-67, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23954500

ABSTRACT

In order to avoid the risks of non-physiological systemic exposure, serum concentrations of estradiol (E2) and testosterone (as measured by mass spectrometry-based assays) should remain below the 95th centiles measured at 9.3pg/ml and 0.26ng/ml for these respective sex steroids in normal postmenopausal women. To document the possibility of achieving this therapeutic objective, we have measured individual 24h serum E2 and testosterone concentrations in women with vulvovaginal atrophy (VVA) receiving daily intravaginal administration of a clinically effective dose of 6.5mg prasterone (dehydroepiandrosterone, DHEA). Serum E2 and testosterone, as well as DHEA and nine of its other metabolites, were assayed at ten time intervals over 24h on the first and seventh days of daily vaginal administration of 6.5mg prasterone. No significant change from baseline of average 24h serum E2 or testosterone concentrations was observed. Moreover, average 24h serum DHEA remained within the normal postmenopausal range. Estrone sulfate and the androgen metabolites androsterone glucuronide and androstane-3α, 17ß-diol glucuronide did not change, thus confirming the absence of any biologically relevant systemic exposure to estrogens and androgens, respectively. Serum concentrations of metabolites of both estrogens and androgens remain within the normal postmenopausal range following daily intravaginal administration of 6.5mg prasterone. As other studies have shown, local formation of sex steroids in peripheral tissues without significant release of E2 or testosterone in the circulation can be achieved with intravaginal prasterone. Thus, prasterone is a promising physiological and attractive solution to treating VVA symptoms.


Subject(s)
Androgens/blood , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/pharmacology , Estrogens/blood , Administration, Intravaginal , Aged , Estradiol/blood , Female , Humans , Middle Aged , Testosterone/blood
7.
Gynecol Endocrinol ; 26(7): 524-32, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20459349

ABSTRACT

Following the compelling data obtained in a pivotal phase III clinical trial performed in 218 postmenopausal women suffering from vaginal atrophy who received daily intravaginal 0.25, 0.5 or 1.0% DHEA (dehydroepiandrosterone) ovules for 12 weeks, we have performed analysis of the four co-primary objectives at each site of that multicentre U.S. and Canadian trial. Comparison was made of the change in percentage of parabasal and superficial cells, vaginal pH and severity of the most bothersome symptom. The site-by-site (seven sites) analysis has shown that 10-13 women per group are generally sufficient to obtain a significant or highly statistically significant decrease in vaginal pH and percentage of parabasal cells and increased percentage of superficial cells at p values ranging from 0.02 to <0.0001. For vaginal pain as the most bothersome symptom, a statistically significant difference from baseline was found at six out of seven sites. The exceptionally high consistency between all sites in this phase III study and high potency of the compound permit to obtain a clinically and statistically significant to highly significant effect of treatment on all parameters of vaginal atrophy with the 0.5% DHEA daily intravaginal dose which does not significantly affect the serum levels of oestrogens, thus avoiding systemic risks.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Vagina/drug effects , Vagina/pathology , Vaginal Diseases/drug therapy , Administration, Intravaginal , Atrophy/drug therapy , Atrophy/pathology , Double-Blind Method , Female , Humans , Intention to Treat Analysis , Postmenopause/drug effects , Treatment Outcome , Vaginal Diseases/pathology
8.
Prostate ; 69(15): 1704-11, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19670238

ABSTRACT

BACKGROUND: Androgen receptor (AR) expression and its modulation through the carcinogenesis process have been investigated in several studies with conflicting results. MATERIALS AND METHODS: In situ hybridization and immunocytochemistry were used to examine AR expression in prostatic needle core biopsies of benign, high grade prostatic intraepithelial neoplasia (HGPIN) and prostatic adenocarcinoma. RESULTS: A significant increase in AR mRNA levels was found in the cancerous prostatic cells when compared with the benign tissue biopsies. AR abundance in HGPIN was found to be almost half-way between that observed in benign and in cancerous tissue. In the benign prostatic epithelium, the immunocytochemistry data show that AR is exclusively expressed in the nuclei of epithelial cells. However, in 72% of examined cancer biopsies, AR was expressed in both the cytoplasm and nuclei. After examination of medical records of 100 patients diagnosed with prostate cancer, it was found that the AR was expressed in both cellular compartments of cancer cells in 81% of cases when cancer was found to have metastasized outside the prostate. In contrast, when the cancer was organ-confined, AR was localized in both the nuclei and cytoplasm in only 66% of cases. Moreover, when the AR was expressed in the cytoplasm of cancerous cells, consecutive serial sections immunostained with the mitochondrial marker suggest that AR is localized in the mitochondria. CONCLUSIONS: AR mRNA expression is significantly higher in prostate cancer when compared to benign prostatic tissue.


Subject(s)
Adenocarcinoma/pathology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/biosynthesis , Adenocarcinoma/metabolism , Aged , Biopsy , Humans , Immunohistochemistry , In Situ Hybridization , Male , Middle Aged , Neoplasm Metastasis , Predictive Value of Tests , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Androgen/genetics , Retrospective Studies
9.
Menopause ; 16(5): 923-31, 2009.
Article in English | MEDLINE | ID: mdl-19424093

ABSTRACT

OBJECTIVE: The objective of this study was to provide evidence that the transformation of DHEA into both androgens and/or estrogens locally in cells of the three layers of the vagina (epithelium, lamina propria, and muscularis) would have effects of greater impact, including effects on sexual function, than only effects on superficial epithelial cells as achieved with estrogens. METHODS: This prospective, randomized, double-blind, and placebo-controlled phase III clinical trial has evaluated the effect of daily local intravaginal application of Prasterone (dehydroepiandrosterone; DHEA) for 12 weeks on the domains of sexual dysfunction, namely, desire/interest, arousal, orgasm, and pain at sexual activity, in 216 postmenopausal women with moderate to severe symptoms of vaginal atrophy. RESULTS: A time- and dose-dependent improvement of the four domains of sexual function was observed. At the 12-week time interval, the 1.0% DHEA dose led, compared with placebo, to 49% (P = 0.0061) and 23% (P = 0.0257) improvements of the desire domains in the Menopause Specific Quality of Life and Abbreviated Sex Function questionnaires, respectively. Compared with placebo, the Abbreviated Sex Function arousal/sensation domain was improved by 68% (P = 0.006), the arousal/lubrication domain by 39% (P = 0.0014), orgasm by 75% (P = 0.047), and dryness during intercourse by 57% (P = 0.0001). CONCLUSIONS: By a local action in the vagina, DHEA applied daily at doses at which serum steroids remain well within normal postmenopausal values exerts relatively potent beneficial effects on all four aspects of sexual dysfunction. Such data indicate that combined androgenic/estrogenic stimulation in the three layers of the vagina exerts important beneficial effects on sexual function in women without systemic action on the brain and other extravaginal tissues.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Libido/drug effects , Postmenopause/drug effects , Sexual Dysfunction, Physiological/drug therapy , Sexual Dysfunctions, Psychological/drug therapy , Vagina/drug effects , Administration, Intravaginal , Adult , Aged , Atrophy , Dehydroepiandrosterone/deficiency , Dehydroepiandrosterone/pharmacology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Libido/physiology , Middle Aged , Postmenopause/physiology , Postmenopause/psychology , Sexual Dysfunction, Physiological/etiology , Sexual Dysfunction, Physiological/psychology , Sexual Dysfunctions, Psychological/etiology , Sexual Dysfunctions, Psychological/psychology , Surveys and Questionnaires , Vagina/pathology
10.
Menopause ; 16(5): 897-906, 2009.
Article in English | MEDLINE | ID: mdl-19436226

ABSTRACT

OBJECTIVE: Because a previous 1-week study has shown no or minimal changes in the serum levels of dehydroepiandrosterone (DHEA) and its metabolites after up to daily 1.8% (23.4 mg) intravaginal DHEA, the objective of the present study was to investigate the serum steroid levels during a 12-week daily intravaginal administration of 0%, 0.25%, 0.5%, and 1.0% DHEA (Prasterone) 1.3 mL ovules. METHODS: In a double-blind, placebo-controlled phase III study, 218 postmenopausal women (age range, 42-74 y) were randomized to receive daily one of four DHEA concentrations intravaginally. Serum steroids were measured by a Good Laboratory Practice-validated mass spectrometry technology in samples obtained at time of visit. RESULTS: The serum levels of DHEA and 11 of its metabolites measured at screening, day 1, and weeks 2, 4, 8, and 12 in women showed no or minimal changes during the whole observation period, with all values remaining well within the limits of normal postmenopausal women. No accumulation of the steroid metabolites nor change in DHEA bioavailability was detected. CONCLUSIONS: The present data show that local daily intravaginal DHEA administration at DHEA doses of 3.25-13 mg was able to rapidly and efficiently achieve correction of all the signs and symptoms of vaginal atrophy and improve sexual function and caused no or minimal changes in serum sex steroid levels, which all remain within the normal postmenopausal range, thus avoiding the risks of all estrogen formulations.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Estradiol/blood , Hormone Replacement Therapy/methods , Postmenopause , Sexual Dysfunction, Physiological/drug therapy , Vagina/drug effects , Administration, Intravaginal , Aged , Atrophy , Biological Availability , Dehydroepiandrosterone/deficiency , Dehydroepiandrosterone/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Drug Monitoring/methods , Female , Humans , Mass Spectrometry , Middle Aged , Postmenopause/drug effects , Postmenopause/physiology , Prospective Studies , Sexual Dysfunction, Physiological/etiology , Time Factors , Vagina/pathology
11.
Menopause ; 16(5): 907-22, 2009.
Article in English | MEDLINE | ID: mdl-19436225

ABSTRACT

OBJECTIVE: Because the secretion of dehydroepiandrosterone (DHEA), the exclusive source of sex steroids in postmenopausal women, is already decreased by 60% and continues to decline at the time of menopause, the objective of this study was to examine the effect of intravaginal DHEA on the symptoms and signs of vaginal atrophy. METHODS: This prospective, randomized, double-blind and placebo-controlled phase III clinical trial studied the effect of Prasterone (DHEA) applied locally in the vagina on the signs and symptoms of vaginal atrophy in 216 postmenopausal women. RESULTS: All three doses (0.25%, 0.5%, and 1.0%) of DHEA ovules applied daily intravaginally induced a highly significant beneficial change in the percentage of vaginal parabasal and superficial cells and pH as well as in the most bothersome symptom at 2 weeks. At the standard 12-week time interval, 0.5% DHEA caused a 45.9 +/- 5.31 (P < 0.0001 vs placebo) decrease in the percentage of parabasal cells, a 6.8 +/- 1.29% (P < 0.0001) increase in superficial cells, a 1.3 +/- 0.13 unit (P < 0.0001) decrease in vaginal pH, and a 1.5 +/- 0.14 score unit (P < 0.0001) decrease in the severity of the most bothersome symptom. Similar changes were seen on vaginal secretions, color, epithelial surface thickness, and epithelial integrity. Comparable effects were observed at the 0.25% and 1.0% DHEA doses. CONCLUSIONS: Local Prasterone, through local androgen and estrogen formation, causes a rapid and efficient reversal of all the symptoms and signs of vaginal atrophy with no or minimal changes in serum steroids, which remain well within the normal postmenopausal range. This approach avoids the fear of systemic effects common to all presently available estrogen formulations and adds a novel physiological androgenic component to therapy.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/deficiency , Hormone Replacement Therapy/methods , Postmenopause/drug effects , Vagina , Administration, Intravaginal , Adult , Analysis of Variance , Atrophy , Dehydroepiandrosterone/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Hydrogen-Ion Concentration , Middle Aged , Postmenopause/physiology , Prospective Studies , Severity of Illness Index , Treatment Outcome , Vagina/drug effects , Vagina/pathology , Vaginal Smears
12.
Menopause ; 16(1): 30-6, 2009.
Article in English | MEDLINE | ID: mdl-18820592

ABSTRACT

OBJECTIVE: Approximately 50% of postmenopausal women suffer from vaginal atrophy, and a large proportion of them choose intravaginal estrogen preparations administered for local action to avoid systemic exposure to estrogens and its associated risk of breast and uterine cancer. The primary objective of this study was the evaluation of the systematic bioavailability of estradiol and estrone and the pharmacokinetics of two of the most frequently used intravaginal estrogen preparations, namely Vagifem and Premarin cream. DESIGN: While immunobased assays could not previously provide accurate measurement of serum estrogen concentrations in postmenopausal women, we have used validated mass spectrometry assays to measure the pharmacokinetics of serum estradiol and estrone during the 24 hours following the seventh daily application of 25 microg estradiol (Vagifem) and 1 g (0.625 mg) conjugated estrogens (Premarin) cream in 10 postmenopausal women in each group. RESULTS: Serum estradiol was increased on average by 5.4-fold from 3 to 17 pg/mL during the 24-hour period after daily administration of 25 microg estradiol or 1 g (0.625 mg) conjugated estrogens cream. Serum estrone, conversely, increased 150% with Vagifem and 500% with Premarin cream. CONCLUSIONS: The present data using validated, accurate, and sensitive mass spectrometry assays of estrogens show that the Vagifem pill and Premarin cream, after 1 week of daily treatment, cause an approximately fivefold increase in serum estradiol in postmenopausal women, thus indicating that the effects are unlikely to be limited to the vagina and that systemic actions are expected after application of these intravaginal estrogen preparations.


Subject(s)
Estrogens/administration & dosage , Estrogens/blood , Postmenopause , Administration, Intravaginal , Adult , Aged , Estradiol/administration & dosage , Estradiol/blood , Estradiol/pharmacokinetics , Estrogens/pharmacokinetics , Estrogens, Conjugated (USP)/administration & dosage , Estrogens, Conjugated (USP)/blood , Estrogens, Conjugated (USP)/pharmacokinetics , Estrone/blood , Female , Gas Chromatography-Mass Spectrometry , Humans , Middle Aged , Prospective Studies , Sensitivity and Specificity
13.
J Steroid Biochem Mol Biol ; 113(1-2): 52-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19073258

ABSTRACT

The objective of this study was comparison of circulating androgens and their metabolites as well as estrogens measured for the first time by a validated mass spectrometry technology in 60-80-year-old men and women of comparable age. Castration in men (n=34) reduces the total androgen pool by only about 60% as indicated by the decrease in the serum levels of the glucuronide metabolites of androgens compared to intact men (n=1302). Such data are in agreement with the 50 to 75% decrease in intraprostatic dihydrotestosterone (DHT) concentration after castration. Most interestingly, the same amounts of androgens and estrogens are found in postmenopausal women (n=369) and castrated men of comparable age. The most significant therapeutic implication of these findings is the absolute need to add a pure (nonsteroidal) antiandrogen to castration in men with prostate cancer in order to block the action of the 25 to 50% DHT left in the prostate after castration. Not adding an antiandrogen to castration in men treated for prostate cancer is equivalent to not prescribing a blocker of estrogens in women suffering from breast cancer because they are postmenopausal and have low circulating estradiol.


Subject(s)
Breast Neoplasms/drug therapy , Gonadal Steroid Hormones/biosynthesis , Gonadal Steroid Hormones/therapeutic use , Gonads/metabolism , Hormone Replacement Therapy , Prostatic Neoplasms/drug therapy , Sex Characteristics , Aged , Aged, 80 and over , Breast Neoplasms/blood , Castration , Female , Gas Chromatography-Mass Spectrometry , Gonadal Steroid Hormones/blood , Humans , Male , Models, Biological , Postmenopause/blood , Prostatic Neoplasms/blood
14.
J Steroid Biochem Mol Biol ; 111(3-5): 178-94, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18598765

ABSTRACT

The primary objective of this study was measurement of the systemic bioavailability of DHEA and its metabolites following daily intravaginal application of the sex steroid precursor. Forty postmenopausal women were randomized to receive a daily dose of one ovule of the following DHEA concentrations: 0.0%, 0.5%, 1.0% or 1.8%. After only 7 days of treatment, the maturation value of the vaginal epithelial cells was significantly increased while the vaginal pH was significantly decreased at all DHEA doses. These important local effects were observed while the serum concentrations of estradiol and testosterone remained within the values found in normal postmenopausal women at all DHEA doses. Similar observations were made for serum androstenedione, estrone, estrone-sulfate and DHEA-sulfate. Even at the highest 1.8% DHEA dose, serum DHEA was increased at the levels found in normal premenopausal women. The present data show that the intravaginal administration of DHEA permits to rapidly achieve the local beneficial effects against vaginal atrophy without significant changes in serum estrogens, thus avoiding the increased risk of breast cancer associated with the current intravaginal or systemic estrogenic formulations. In addition, the recent observation that DHEA is transformed into both androgens and estrogens in the vagina permits to exert benefits on all the three layers of the vaginal wall.


Subject(s)
Dehydroepiandrosterone , Postmenopause/metabolism , Vagina/drug effects , Area Under Curve , Dehydroepiandrosterone/blood , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone/pharmacology , Double-Blind Method , Female , Humans , Middle Aged , Placebos , Vagina/cytology , Vagina/pathology
15.
J Steroid Biochem Mol Biol ; 110(1-2): 1-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18359622

ABSTRACT

Healthy postmenopausal women aged 60-65 years (n=150) were randomized to receive twice daily application on the skin of 3g of a 0.3% dehydroepiandrosterone (DHEA) or placebo emulsion for 12 months. Serum DHEA and eleven of its metabolites were measured at screening and on day 1, as well as at 1, 3, 6, 9 and 12 months to study long-term metabolism. While serum DHEA and androst-5-ene-3beta, 17beta-diol (5-diol) increased by 203% and 178%, respectively, on average, during the 12-month period, the sum of concentrations of the metabolites of androgens, namely androsterone glucuronide (ADT-G), androstane-3alpha,17beta-diol-3G and -17G increased by only 71% while usually non statistically significant changes of 30%, 17% and 20% were observed for estrone (E(1)), estradiol (E(2)) and E(1) sulfate (E(1)-S), respectively. Despite the return of serum DHEA to normal premenopausal values with the present DHEA treatment regimen, the 65% decrease in the androgen pool found in this group of postmenopausal women is in fact corrected by only 24%, thus remaining 41% below the values found in normal premenopausal women. In fact, the changes in serum DHEA observed after percutaneous DHEA administration are a 186% overestimate of the true changes in androgen formation while the overestimate of estrogen production is even much higher. On the other hand, the pharmacokinetics of the steroids are stable over the 12-month period with no significant induction or decrease of activity of the enzymatic systems transforming DHEA predominantly into androgens.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/blood , Administration, Cutaneous , Aged , Androgens/blood , Androgens/metabolism , Androstane-3,17-diol/blood , Androstane-3,17-diol/metabolism , Androsterone/analogs & derivatives , Androsterone/blood , Androsterone/metabolism , Dehydroepiandrosterone/metabolism , Estradiol/blood , Estradiol/metabolism , Estrogens/blood , Estrogens/metabolism , Female , Humans , Mass Spectrometry , Middle Aged , Skin/metabolism , Steroids/blood , Steroids/metabolism , Time Factors
16.
J Steroid Biochem Mol Biol ; 103(2): 178-88, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17084625

ABSTRACT

The marked decline in serum dehydroepiandrosterone (DHEA) with age is believed to play a role in health problems associated with aging, these health issues being potentially preventable or reversible by the exogenous administration of DHEA. In the present study, liquid chromatography/mass spectrometry/mass spectrometry (LC/MS/MS) and gas chromatrography/mass spectrometry (GC/MS) were used to measure the serum levels of DHEA and 11 of its metabolites in seventy-five 60-65-year-old Caucasian women who received 3g of 0.1%, 0.3%, 1.0% or 2.0% DHEA cream or placebo applied twice daily on the face, upper chest, arms and legs. The serum levels of DHEA increased 574% over control at the 2.0% DHEA dose while the sum of the androgen metabolites androsterone glucuronide (ADT-G), 3alpha-androstenediol-3G (3alpha-diol-3G) and 3alpha-diol-17G increased by only 231%. On the other hand, serum testosterone and dihydrosterone were increased by 192% and 275%, respectively, above basal levels compared to 139% and 158% for estrone and estradiol. Such data show that the transformation of exogenous DHEA in postmenopausal women is preferentially into androgens rather than into estrogens. On the other hand, the present data indicate that serum DHEA measurements following DHEA supplementation in postmenopausal women are an overestimate of the formation of active androgens and estrogens and suggest a decreased efficiency of transformation of DHEA into androgens and estrogens with aging.


Subject(s)
Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/metabolism , Postmenopause/metabolism , Administration, Cutaneous , Aged , Dehydroepiandrosterone/blood , Dose-Response Relationship, Drug , Female , Humans , Middle Aged , Models, Biological , Placebos , Postmenopause/blood , Time Factors
17.
J Steroid Biochem Mol Biol ; 99(4-5): 182-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16621522

ABSTRACT

Despite the long series of cohort studies performed during the last 20 years, the correlation between serum testosterone and any clinical situation believed to be under androgen control in women has remained elusive. This is likely related to the recent finding that the androgens made locally in large amounts in peripheral tissues from the precursor dehydroepiandrosterone (DHEA) act in the same cells where synthesis takes place and are not released in significant amounts in the circulation, thus making unreliable the measurement of serum testosterone as marker of total androgenic activity. The objective is to determine if serum androgen glucuronides can be replaced by testosterone or another steroid as measure of androgenic activity. Since the glucuronide derivatives of androgens are the obligatory route of elimination of all androgens, these metabolites were measured by liquid chromatography tandem mass spectrometry under basal conditions in 377 healthy postmenopausal women aged 55-65 years as well as in 47 premenopausal women aged 30-35 years while testosterone was assayed by gas chromatography mass spectrometry. No correlation was found between the serum concentration of testosterone and that of androsterone glucuronide (ADT-G) or androstenediol glucuronide (3alpha-diol-G), the androgen metabolites which account for the total pool of androgens. The present data show that measurement of the total pool of androgens reflected by the serum levels of ADT-G and 3alpha-diol-G cannot be replaced by serum testosterone or any other steroid, including DHEA or DHEA sulphate. These findings may have implications for women with androgen deficiency involving osteoporosis, obesity, type 2 diabetes, sexual dysfunction, loss of muscular strength and a series of other clinical situations affecting women's health. Measuring ADT-G and 3alpha-diol-G might identify cases of true androgen deficiency and provide an opportunity to offer appropriate androgen therapy.


Subject(s)
Androgens/analysis , Biomarkers/analysis , Glucuronides/analysis , Adult , Aged , Dehydroepiandrosterone/analysis , Female , Humans , Middle Aged , Postmenopause , Premenopause , Testosterone/analysis , Testosterone/blood
18.
J Urol ; 175(2): 510-6; discussion 516-7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16406983

ABSTRACT

PURPOSE: We evaluated the probability of positive serum PSA (3 ng/ml or greater) and CaP detection at annual followup visits in men with negative initial PSA (less than 3 ng/ml) to optimize the re-screening schedule. MATERIALS AND METHODS: Data on 5,387 men 45 to 80 years old with negative PSA and no CaP diagnosis at the first screening visit were obtained from the Laval University Prostate Cancer Screening Program database. Accelerated failure time regressions were fitted to time from baseline to positive PSA and to time from positive PSA to CaP detection. The models were combined to estimate the cumulative probability of positive PSA followed by CaP detection at re-screening. RESULTS: The 5-year cumulative probability of detecting CaP at annual visits in men with baseline PSA up to 1.5 ng/ml remained below 0.8%, while it was 1.3%, 4.8% and 8.3% in men with PSA 1.5 to less than 2, 2 to less than 2.5 and 2.5 to less than 3 ng/ml, respectively. Time to positive PSA significantly decreased with increasing baseline PSA and age, while the time between positive PSA and CaP detection depended only on age. Men with PSA below 1.0 ng/ml could wait for 4 to 5 years before being re-tested, while men with PSA between 1.0 and 1.5 ng/ml should be screened every second year and men with PSA 1.5 ng/ml or greater should be screened every year. CONCLUSIONS: The proposed retesting schedule using current PSA and age decreases the number of visits by 38.1%, while delaying the detection of only 2.4% of CaPs that would have been detected using annual PSA testing.


Subject(s)
Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/diagnosis , Aged , Aged, 80 and over , Disease Progression , Humans , Male , Middle Aged , Time Factors
19.
Endocr Rev ; 26(3): 361-79, 2005 May.
Article in English | MEDLINE | ID: mdl-15867098

ABSTRACT

In 1979, the first prostate cancer patient was treated with a GnRH agonist at the Laval University Medical Center in Quebec City, Canada, thus rapidly leading to the worldwide replacement of surgical castration and high doses of estrogens. The discovery of medical castration with GnRH agonists was soon followed by fundamental changes in the endocrine therapy of prostate cancer. Most importantly, the excellent tolerance accompanying the treatment with GnRH agonists has been a key factor that permitted a series of studies demonstrating a major reduction in the death rate from prostate cancer ranging from 31 to 87% at 5 yr of follow-up in patients with localized or locally advanced prostate cancer. In fact, a one third reduction in prostate cancer deaths has been calculated in the metaanalysis of all available studies. The general acceptance of this discovery by patients and physicians is illustrated by world sales above 3.0 billion U.S. dollars in 2003. Although extremely efficient in achieving complete medical castration and well tolerated, with no other side effects than the expected hypoandrogenicity, GnRH agonists should not be administered alone. In fact, shortly after discovery of the castration effects of GnRH agonists, we observed that approximately 50% of androgens remain in the prostate after castration, thus leading to the recognition of the role of adrenal dehydroepiandrosterone as an important source of the androgens synthesized locally in the prostate and in many peripheral target tissues. We therefore developed combined androgen blockade (CAB), whereby the androgens of both testicular and adrenal origins are blocked simultaneously at start of treatment with the combination of a GnRH agonist to block the testis and a pure antiandrogen to block the action of the androgens produced locally. CAB, first used in advanced metastatic disease, has been the first treatment shown to prolong life in prostate cancer. Most interestingly, in 2002, we made the observation that CAB alone given continuously for 6.5 yr or more leads to cure of the disease in at least 90% of cases, thus suggesting that androgen blockade combining a GnRH agonist and a pure antiandrogen could well be the most efficient treatment of localized prostate cancer, and thus offering the possibility of practically eliminating death from prostate cancer.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Gonadotropin-Releasing Hormone/agonists , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Gonadotropin-Releasing Hormone/therapeutic use , Humans , Male
20.
J Steroid Biochem Mol Biol ; 92(5): 327-44, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15698538

ABSTRACT

For about 50 years, androgen blockade in prostate cancer has been limited to monotherapy (surgical castration) or high doses of estrogens in patients with advanced disease and bone metastases. The discovery of medical castration with LHRH agonists has led to fundamental changes in the endocrine therapy of prostate cancer. In 1979, the first prostate cancer patient treated with an LHRH agonist received such treatment at the Laval University Medical Center. A long series of studies have clearly demonstrated that medical castration with an LHRH agonist has inhibitory effects on prostate cancer equivalent to those of surgical castration. The much higher acceptability of LHRH agonists has been essential to permit a series of studies in localized disease. Based upon the finding that the testicles and adrenals contribute approximately equal amounts of androgens in the human prostate, the combination of medical (LHRH agonist) or surgical castration associated with a pure antiandrogen (flutamide, nilutamide or bicalutamide) has led to the first demonstration of a prolongation of life in prostate cancer, namely a 10-20% decreased risk of death according to the various metaanalyses of all the studies performed in advanced disease. In analogy with the other types of advanced cancers, the success of combined androgen blockade in metastatic disease is limited by the development of resistance to treatment. To avoid the problem of resistance to treatment while taking advantage of the relative ease of diagnosis of prostate cancer at an "early" stage, the much higher acceptability of LHRH agonists has permitted a series of studies which have demonstrated a major reduction in deaths from prostate cancer ranging from 31% to 87% at 5 years of follow-up in patients with localized or locally advanced prostate cancer. Most importantly, recent data show that the addition of a pure antiandrogen to an LHRH agonist in order to block the androgens made locally in the prostate leads to a 90% long-term control or probable cure of prostate cancer.


Subject(s)
Hormones/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/mortality , Animals , Humans , Male , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL