Fact-checked by Grok 2 weeks ago

Prasterone

Prasterone, also known as dehydroepiandrosterone (DHEA), is an endogenous C19 produced primarily by the zona reticularis of the , with smaller amounts synthesized in the gonads and , serving as a key precursor to androgens such as testosterone and estrogens such as . In its native form, prasterone exhibits minimal direct hormonal activity but is enzymatically converted in target tissues to active sex steroids, influencing physiological processes including sexual development, immune function, and metabolic regulation. Levels of prasterone peak in early adulthood and decline progressively with age, a pattern associated with conditions such as and age-related hormonal changes. Medically, prasterone is approved in intravaginal insert form (brand name Intrarosa) for the of moderate to severe —a symptom of vulvovaginal —arising from postmenopausal , where it undergoes local to estrogens and androgens, alleviating vaginal dryness, during intercourse, and tissue without inducing significant systemic hormonal effects. Clinical trials have demonstrated statistically significant improvements in scores, vaginal , and epithelial maturation indices compared to , positioning prasterone as a targeted alternative to systemic therapies that carry risks of or cardiovascular events. Over-the-counter oral prasterone supplements are marketed for purported benefits in energy, mood, and , though the U.S. has not approved these for any medical indication, and empirical evidence for broad anti-aging or performance-enhancing claims remains inconsistent and limited by methodological flaws in studies. Early investigations into systemic prasterone for conditions like systemic showed mixed results, with no sustained approval beyond investigational use due to insufficient efficacy data.

Chemical Properties

Structure and Synthesis

Prasterone, systematically known as dehydroepiandrosterone (DHEA), possesses the C₁₉H₂₈O₂ and a of 288.42 g/. It features an androst-5-ene backbone with a β-hydroxy group at carbon 3 and a at carbon 17, classifying it as 3β-hydroxy-5-androsten-17-one. This structure positions prasterone as a key precursor in the of androgens and estrogens. Biosynthetically, prasterone arises primarily in the zona reticularis of the from via enzymatic transformations. undergoes side-chain cleavage by cytochrome P450scc (CYP11A1) to yield , which is then converted to 17α-hydroxypregnenolone by 17α-hydroxylase activity of . Subsequent 17,20-lyase action of cleaves the side chain, producing prasterone. These reactions occur in the mitochondria and , with production peaking in early adulthood before declining with age. Pharmaceutical synthesis of prasterone typically employs semi-synthetic routes from microbial products like 4-androstene-3,17-dione. A chemoenzymatic approach involves stereoselective reduction of the Δ4 to the Δ5 position using and enzymes, followed by and deacetylation steps to isolate prasterone or its . Alternative multi-step chemical processes incorporate of the 17-keto group and selective dehydrogenation to establish the 5-ene unsaturation, minimizing side reactions.

Derivatives and Analogs

Prasterone enanthate, the 3β-enanthate of prasterone, serves as a for sustained release in pharmaceutical formulations, such as intramuscular injections combined with for menopausal . This derivative undergoes hydrolysis to release free prasterone and . Prasterone sulfate, the 3β- , constitutes the primary circulating form of prasterone, comprising over 99% of total prasterone in plasma due to sulfation in the adrenal glands and liver. Structural analogs include 7-ketodehydroepiandrosterone (7-oxo-prasterone), which bears an additional keto group at carbon 7 and functions as a that bypasses conversion to active sex steroids like testosterone or . This analog has been studied for metabolic effects, including potential support for , though clinical evidence remains limited to small trials. Fluorinated analogs, such as 16α-fluoro-5-androsten-17-one (fluasterone), exhibit amplified potency in preclinical models for inhibiting tumor growth and compared to native prasterone, attributed to modifications enhancing metabolic and receptor interactions. Similarly, 7α- and 7β-amino-prasterone derivatives have demonstrated apoptotic effects on Leydig and Sertoli cells , positioning them as candidates for investigating steroid-mediated pathways. Synthetic spiro-analogs like BNN27, a C-17 spiro , promote via non-genomic mechanisms independent of conversion, with preclinical data supporting roles in mitigating neurodegeneration. These modifications highlight efforts to decouple prasterone's precursor activity from androgenic or estrogenic side effects while preserving or enhancing therapeutic potential.

Pharmacology

Pharmacodynamics

Prasterone, known chemically as dehydroepiandrosterone (DHEA), functions primarily as an endogenous , undergoing enzymatic conversion in peripheral tissues to yield active . These transformations involve key steroidogenic enzymes, including to form , 17β-hydroxysteroid dehydrogenase to produce testosterone, to generate , and to synthesize estrogens such as estrone and . This biosynthetic pathway enables prasterone to support androgenic and estrogenic effects indirectly, with its own direct binding affinity to the and receptors being relatively weak. In vaginal tissues, following local administration as a 6.5 mg insert, prasterone is metabolized intracellularly via an mechanism, where target cells express the necessary enzymes to convert it into and that activate androgen receptors and estrogen receptors within the same cells. This local activation promotes physiological changes such as increased maturation of vaginal epithelial cells (e.g., rise in superficial cells from approximately 1% to 11% and decline in parabasal cells from 54% to 13% over 12 weeks), enhanced vascularization, reduced vaginal (from about 6.3 to 5.4), and restoration of lactobacilli , thereby alleviating symptoms of vulvovaginal without substantially elevating systemic levels beyond postmenopausal norms. Prasterone also interacts with non-steroid targets, acting as an at GABA_A receptors, an at NMDA and sigma-1 receptors, an activator of alpha (PPARα), and an of , which may underlie additional effects like , modulation of neuronal excitability, and potential metabolic influences observed in preclinical models. Its sulfate form, DHEA-S, further contributes as a precursor modulating and signaling. These multifaceted actions highlight prasterone's role beyond mere hormone precursor status, though clinical significance varies by context and dosage.

Pharmacokinetics

Prasterone, or dehydroepiandrosterone (DHEA), exhibits route-dependent pharmacokinetics, with leading to rapid but limited systemic due to extensive first-pass in the liver and gut, where much of it is converted to the sulfate conjugate DHEA-S. Following a single 50 mg oral dose in healthy elderly subjects, plasma DHEA levels peak within hours and restore concentrations comparable to those in younger adults, though absolute in humans remains unquantified and is estimated low based on animal data (3.1% in cynomolgus monkeys). Vaginal administration, as in the approved 6.5 mg daily insert, results in primarily local absorption with modest systemic exposure; mean serum trough DHEA concentrations increase by 47% (from 1.81 ng/mL baseline) after 12 weeks, accompanied by dose-dependent C_max of 5.97 ng/mL and AUC_{24} of 65.49 ng·h/mL on day 1. Distribution occurs widely, with prasterone taken up by peripheral tissues including the liver, kidneys, gonads, and , where it serves as a precursor for synthesis of active androgens and estrogens via enzymes such as hydroxysteroid dehydrogenases, 5α-reductases, and aromatases. The apparent terminal of DHEA exceeds 20 hours in oral studies, influenced by interconversion with DHEA-S, though unconjugated DHEA has a shorter intrinsic of approximately 12 hours; no significant sex differences in volume are noted, but women show greater DHEA-S to DHEA . Metabolism is primarily hepatic and peripheral, yielding major metabolites including DHEA-S, , and , with further transformation to testosterone, , , and estrone occurring locally in target tissues rather than systemically. Excretion occurs mainly via urine as inactive and conjugates of these metabolites, with no accumulation observed in long-term vaginal use where systemic and levels remain within postmenopausal norms (e.g., testosterone trough +21%, +19% after 12 weeks). Oral dosing at 25-50 mg daily sustains elevated baseline DHEA and metabolite levels without disproportionate increases in active steroids.

Clinical Dosing Considerations

The recommended dosage of prasterone for moderate to severe due to postmenopausal vulvovaginal is one 6.5 mg intravaginal insert administered once daily at bedtime. This regimen utilizes a disposable applicator for insertion, with individually wrapped inserts provided for nightly use. No dosage adjustments are required for elderly patients over 65 years of age, reflecting minimal systemic exposure and consistent pharmacokinetics in postmenopausal women. Similarly, no specific modifications are indicated for hepatic or renal impairment, as clinical data support the fixed 6.5 mg dose without evidence of accumulation or altered clearance necessitating changes. Clinical trials demonstrating efficacy, such as those evaluating 12 weeks of daily administration, employed this standard dose without titration, showing statistically significant improvements in vaginal symptoms and superficial cell counts. Prasterone lacks duration-of-use restrictions, unlike some estrogen therapies, due to its local conversion to estrogens and androgens with limited serum impact. Concurrent use with aromatase inhibitors warrants caution, as prasterone may interfere with their action, potentially requiring dose reevaluation.

Approved Medical Uses

Treatment of Vulvovaginal Atrophy

Prasterone, administered as a 6.5 mg intravaginal insert (Intrarosa), is approved by the for the treatment of moderate to severe , a primary symptom of vulvovaginal atrophy (VVA), in postmenopausal women. Approval was granted on November 17, 2016, based on evidence of symptom relief without the systemic risks associated with therapies. The recommended regimen involves nightly insertion at bedtime, initially for 12 weeks, with potential maintenance dosing as clinically indicated. The therapeutic action relies on local intracrinology, where prasterone—a precursor—is taken up by vaginal epithelial and stromal cells and enzymatically converted into active (such as testosterone and ) and (such as and estrone). This site-specific transformation restores vaginal tissue integrity, reduces , increases superficial cell percentage in cytology, and alleviates dryness and pain without producing clinically significant elevations in serum or levels, minimizing risks like . Efficacy was demonstrated in two pivotal 12-week, multicenter, randomized, double-blind, -controlled phase III trials involving 406 healthy postmenopausal women aged 40 to 80 years with moderate to severe VVA symptoms. The primary , change in severity score (assessed via a 0-4 visual analog scale in patient-reported questionnaires and clinical evaluation), showed statistically significant improvements of 0.36 to 0.40 points greater than (p<0.001). Secondary outcomes included reductions in vaginal dryness severity and improvements in vaginal (decrease of approximately 1.3-1.5 units) and cytology parameters (e.g., increase in superficial cells by 10-15% and decrease in parabasal cells by 20-25%). These effects were observed as early as week 4 and sustained through week 12, with response rates for relief exceeding 60% in prasterone groups versus 40-45% with . Compared to low-dose conjugated equine cream (0.3 mg), prasterone demonstrated comparable efficacy in and dryness relief in a separate randomized , though direct head-to-head data remain limited. Long-term data beyond 52 weeks are sparse, but extensions of the phase III trials indicate sustained benefits with continued use and no new safety signals. Prasterone does not require endometrial , unlike systemic or certain local estrogens, due to negligible uterine exposure.

Specific Formulations and Administration

Prasterone is formulated as a vaginal insert under the brand name Intrarosa, containing 6.5 mg of prasterone (dehydroepiandrosterone) dispersed in 1.3 mL of off-white hard fat (Witepsol H15) as the vehicle. This ovule-like insert is designed for local intravaginal delivery to minimize systemic absorption while targeting vulvovaginal tissues. The U.S. approved this formulation on November 17, 2016, specifically for the treatment of moderate to severe , a symptom of vulvovaginal atrophy, in postmenopausal women. Administration involves inserting one 6.5 vaginal insert once daily at using a single-use disposable applicator provided with the product. The applicator is activated by pulling back the until it stops, loaded with the unwrapped insert, and then gently inserted into the while the patient lies on her back with knees drawn up, similar to standard insertion techniques. After insertion, the is pushed to release the insert, and the applicator is discarded; no additional lubrication or preparation is required beyond standard hygiene. Treatment is intended for continuous daily use, with efficacy observed in clinical trials after 12 weeks, though duration should be guided by symptom response and assessment. No other prasterone formulations are approved by the FDA for vulvovaginal atrophy; oral, injectable, or topical non-vaginal routes are not indicated for this use due to differing pharmacokinetic profiles and potential for greater systemic exposure. Patients should be advised to store inserts at controlled (20–25°C) and avoid use if allergic to prasterone or its excipients.

Safety Profile

Adverse Effects

In clinical trials evaluating intravaginal prasterone for vulvovaginal atrophy, the drug has demonstrated a generally favorable safety profile, with adverse effects primarily mild, local, and occurring at rates similar to or only modestly higher than . Across four 12-week randomized, -controlled studies involving over 1,800 postmenopausal women, treatment discontinuation due to adverse events was low at approximately 3.6% in the prasterone group versus 3.3% with . Serious adverse events were rare and balanced between groups, with no evidence of increased systemic risks such as cardiovascular events, , or malignancies directly attributable to prasterone; one case of was reported in a 52-week open-label extension trial among 318 participants, but long-term causality remains unestablished. The most common adverse reaction is , reported in 2.7% to 8.3% of prasterone users compared to 1.3% to 4% with , often described as leakage or increased wetness without . Abnormal Pap smear results represent another frequent finding, with incidences of 2% to 5% in treated groups versus 1.6% in controls, typically resolving without intervention and linked to local epithelial changes rather than precancerous lesions. Other local effects, such as vulvovaginal discomfort or pruritus, occur at rates below 2% and do not differ significantly from . Systemic absorption of prasterone and its metabolites (including and testosterone) is minimal with , resulting in negligible androgenic or estrogenic side effects compared to oral DHEA formulations, which can cause , , or voice deepening in 5-10% of users. No clinically meaningful changes in serum levels, profiles, or markers were observed in phase 3 trials up to 52 weeks, supporting the localized via intracrinology, where conversion to active steroids occurs primarily within vaginal tissues. reactions are contraindications, but post-marketing surveillance has not identified new safety signals beyond trial data as of 2024.

Contraindications and Precautions

Prasterone is contraindicated in women with undiagnosed abnormal genital bleeding, for which the underlying cause must be evaluated prior to considering treatment. In regulatory approvals outside the United States, such as in the European Union, contraindications additionally encompass hypersensitivity to prasterone or excipients, known or suspected breast cancer, estrogen-dependent malignancies (e.g., endometrial cancer), untreated endometrial hyperplasia, previous or active venous thromboembolism, thrombophilic disorders, active or recent arterial thromboembolic disease, acute or unresolved liver disease, and porphyria. Precautions are advised for women with a current or prior history of , as prasterone is metabolized to —a known for exogenous estrogen therapy—and clinical trials excluded such patients, leaving safety data absent in this population. Although prasterone exhibits primarily local action with minimal systemic absorption, any emergent or spotting during treatment warrants prompt investigation, potentially including endometrial , to exclude . Close monitoring is recommended in patients with conditions exacerbated by estrogens or androgens, such as , , , or a history of , with periodic pelvic and breast examinations advised before and during use. Prasterone is indicated solely for postmenopausal women and has not been studied in pregnant or lactating individuals, pediatric populations, or those with renal or hepatic impairment. Risks associated with systemic , including , , and , apply to a lesser degree but necessitate individualized risk-benefit assessment, particularly beyond one year of use where long-term endometrial safety remains unestablished.

Long-Term Safety Data

The primary source of long-term safety data for prasterone (intravaginal dehydroepiandrosterone, 6.5 mg daily) derives from the ERC-230 trial, a phase III, open-label, single-arm study involving 521 postmenopausal women with vulvovaginal treated for up to 52 weeks. In this trial, 80.2% of participants experienced treatment-emergent s, predominantly mild to moderate, with application site discharge reported in 14.0% and urinary tract infections in 10.2%; serious s occurred in 3.5%, and discontinuations due to s were 6.0%, with no deaths attributed to the . Pooled data from shorter placebo-controlled trials (ERC-231 and ERC-238, 12 weeks) showed comparable rates between prasterone and groups, suggesting many events reflect underlying postmenopausal conditions rather than , though the absence of a in ERC-230 limits definitive attribution. Serum concentrations of prasterone and its metabolites (including testosterone, , and estrone) increased following administration but remained within the normal postmenopausal range, with no evidence of supraphysiological elevations or clinically significant hormonal disruptions over 52 weeks. Endometrial assessments via in 94% of ERC-230 participants revealed atrophic , with no cases of or , and mean endometrial thickness of 2.2 mm by ; however, long-term endometrial safety beyond one year has not been systematically evaluated. Breast safety evaluations, including mammograms in 98% of participants, were predominantly normal (99%), though two cases of pathology—one and one infiltrating ductal —were reported, without established drug linkage given the population's baseline cancer risk. Cervical assessments via Pap smears showed 90% normal results, with 3% atypical findings mostly negative for high-risk HPV and not associated with prasterone exposure in comparative analyses.
Safety ParameterERC-230 Findings (52 Weeks)Notes
Application Site Discharge14.0%Attributed to insert melting; most common drug-related event.
10.2%Similar to shorter trials; likely age-related.
Serious Adverse Events3.5% (18/521)None deemed causally related; includes and infections.
Endometrial Biopsies94% atrophic (430/457)No ; limited to treated population excluding prior cancer history.
Pathology0.4% significant findingsRare; baseline postmenopausal risk not controlled.
Regulatory reviews by the FDA and concluded no major safety signals for up to one year of use, supporting approval for vulvovaginal atrophy, but emphasized monitoring for or breast changes due to local conversion to estrogens and androgens, with theoretical oncogenic risks unproven in human data. Data beyond 52 weeks are absent, and trials excluded women with cancer history, , or uncontrolled , potentially underrepresenting risks in broader populations; open-label design may inflate adverse event reporting via heightened awareness. Post-marketing surveillance, including drug utilization studies, continues to assess real-world long-term effects.

Investigational and Off-Label Applications

Patients with primary adrenal insufficiency exhibit a profound deficiency in dehydroepiandrosterone (DHEA) due to the absence of adrenal zona reticularis function, unlike the more commonly replaced glucocorticoids and mineralocorticoids. Oral DHEA replacement at doses of 25 to 50 mg daily has been studied primarily in women, with randomized controlled trials reporting improvements in subjective well-being, mood, fatigue, and sexual function in some cohorts. For instance, a 1999 double-blind trial involving 24 women found significant enhancements in overall well-being, depression scores, and sexual interest after 4 months of 50 mg daily DHEA compared to placebo. However, longer-term studies, such as a 9-month trial in 31 patients, detected no benefits for subjective health status or sexuality, highlighting inconsistent outcomes across trials. A 12-month study noted modest gains in femoral neck bone mineral density but no effects on fatigue, cognition, or sexual function, with supraphysiological DHEA sulfate levels in some older participants raising concerns about dosing precision. DHEA replacement remains investigational and non-standard, as evidence does not consistently support routine clinical adoption, and guidelines emphasize the need for further large-scale trials to clarify benefits versus risks like androgen excess. Serum DHEA concentrations peak in the third decade of life and decline by 70% to 80% by age 70, a pattern observed in both sexes and associated with reduced and precursors, potentially contributing to frailty, cognitive changes, and metabolic shifts. Supplementation with 50 mg daily oral DHEA in elderly populations (typically aged 60–80) restores levels toward youthful ranges, as demonstrated in a multicenter of 87 men and 57 women, which increased DHEA by over 10-fold but yielded no significant improvements in , strength, or metrics. Meta-analyses of randomized trials indicate modest elevations in testosterone (mean increase of 0.8 nmol/L) and but inconsistent effects on clinical endpoints like , insulin sensitivity, or depressive symptoms, with subgroup benefits occasionally noted in women with low baseline levels. For cognition, systematic reviews of supplementation trials report no reliable reversal of age-related decline, despite preclinical links between DHEA and ; human data from placebo-controlled studies show null or negligible impacts on or . Overall, while DHEA decline correlates with aging phenotypes, supplementation lacks robust empirical support for mitigating them, with potential risks including , , and unknown long-term oncogenic effects prompting caution and relegating its use to contexts.

Menopausal Symptoms Beyond VVA

Intravaginal prasterone, administered at a dose of 6.5 mg daily, has demonstrated efficacy in reducing (LUTS) in postmenopausal women, including urinary urgency, frequency, and urge incontinence, which extend beyond vulvovaginal atrophy as part of the genitourinary syndrome of menopause (). A of 11 randomized controlled trials involving postmenopausal women with found significant improvements in these symptoms, with standardized mean differences indicating reduced urgency (SMD -0.45, 95% -0.72 to -0.18) and frequency (SMD -0.38, 95% -0.65 to -0.11), alongside decreased episodes of urge . These effects are attributed to local conversion of prasterone to estrogens and androgens in urogenital tissues, enhancing epithelial integrity and reducing inflammation without substantial systemic hormone elevation. Clinical trials have also linked intravaginal prasterone to a lower incidence of recurrent urinary tract infections (UTIs) in women with . In a prospective of postmenopausal women, prasterone reduced UTI by approximately 50% over one year compared to baseline rates, potentially due to improved vaginal and urethral mucosa health that limits bacterial ascension. Similarly, validated questionnaires such as the Urogenital Distress Inventory-6 (UDI-6) and Questionnaire Short Form (OAB-q SF) showed statistically significant declines in symptom scores after 12 weeks of , with improvements persisting in subsets of women with concurrent . In contrast, prasterone exhibits negligible impact on systemic menopausal symptoms such as instability (e.g., hot flashes or ), owing to its pharmacokinetic profile of minimal serum absorption. Daily intravaginal dosing maintains circulating , testosterone, and DHEA levels within the lower of normal postmenopausal ranges, avoiding the hormonal fluctuations needed for vasomotor relief. Reviews of broader DHEA supplementation (primarily oral routes) similarly report insufficient evidence for vasomotor symptom reduction across multiple randomized trials, with no high-quality data supporting benefits for disturbances, , or overall psychological in . These findings underscore prasterone's targeted local action, limiting its utility for non-genitourinary manifestations while highlighting investigational promise for LUTS management pending larger confirmatory studies.

Other Potential Uses

Prasterone, also known as dehydroepiandrosterone (DHEA), has been investigated for its potential role in managing (SLE), with studies indicating reduced disease flares and improved patient well-being in women with mild-to-moderate disease activity. A Cochrane review of randomized trials found low-quality evidence suggesting DHEA may modestly improve SLE activity scores, though larger confirmatory studies are needed due to methodological limitations in existing data. DHEA levels are often inversely correlated with SLE activity, supporting a plausible mechanistic link via , but clinical benefits remain investigational and unapproved. In treatment, DHEA supplementation has shown promise as an adjunct or monotherapy, particularly in midlife-onset major or minor , with randomized trials demonstrating symptom reduction compared to . Systematic reviews and meta-analyses of controlled trials report a beneficial effect on depressive symptoms, though effect sizes are modest and results warrant caution due to heterogeneity and small sample sizes. Evidence from studies suggests a potential causal link between higher DHEA levels and better response, but this requires further validation in diverse populations. For bone health, DHEA has been explored in , with pooled analyses of clinical trials showing increases in lumbar spine density (BMD) of approximately 1.7-3.5% over 12-24 months in postmenopausal women, alongside potential enhancements in muscle mass. However, a of seven randomized controlled trials concluded no significant BMD improvement after adjusting for , highlighting mixed findings and the need for longer-term studies to assess risk reduction. These effects may stem from DHEA's conversion to estrogens and androgens, but applications remain off-label and unsupported by regulatory approvals. Preliminary research also suggests DHEA's utility in , where it may aid recovery in severely injured patients through and anabolic pathways, though evidence is limited to reviews rather than large-scale trials. Overall, while these uses leverage DHEA's precursor role in steroidogenesis, most lack robust, high-quality evidence for routine clinical adoption, and potential risks such as androgenic side effects necessitate cautious evaluation.

Research Evidence

Strong Empirical Support Areas

Intravaginal prasterone (6.5 mg daily) demonstrates strong empirical support for alleviating moderate to severe associated with vulvovaginal atrophy (VVA) in postmenopausal women, as evidenced by two pivotal phase III randomized, double-blind, -controlled trials involving 406 healthy participants aged 40 to 80 years. In these 12-week studies, prasterone significantly improved the dyspareunia score (primary endpoint), with reductions of approximately 0.36 to 0.44 points on a 4-point severity scale compared to , alongside histological improvements including decreased parabasal cells (from 53.8% to 15.6%), increased superficial cells (from 7.8% to 25.5%), and lowered vaginal (from 6.6 to 5.3). These changes correlated with clinical symptom relief, supporting local conversion of prasterone to estrogens and androgens without substantial systemic absorption. A of randomized controlled trials further corroborates these findings, concluding that prasterone therapy yields significant therapeutic effects on VVA symptoms, including , vaginal dryness, and overall sexual function, with effect sizes favoring prasterone over (standardized mean difference -0.89 for ; 95% CI -1.28 to -0.50). Long-term safety data from an open-label III extension (ERC-230; N=521) over 52 weeks confirmed sustained efficacy and tolerability, with no new safety signals beyond mild local irritation reported in less than 5% of users. This body of evidence underpinned FDA approval on November 17, 2016, as the first non-estrogen therapy specifically for postmenopausal due to VVA. No other indications exhibit comparable rigorous, replicated phase III ; systemic oral DHEA lacks similar validation for VVA or broader menopausal symptoms due to inconsistent and off-target effects.

Mixed or Inconclusive Findings

Studies on dehydroepiandrosterone (DHEA) supplementation for cognitive enhancement in non-demented older adults have yielded inconclusive results, with systematic s indicating no consistent beneficial effects on or other cognitive domains. A Cochrane of randomized controlled trials found insufficient to improvements in cognitive following DHEA administration, noting that available data from controlled trials do not demonstrate efficacy in middle-aged or elderly populations without . Similarly, analyses specific to postmenopausal women concluded that DHEA therapy does not enhance cognitive performance, based on evaluations of multiple randomized clinical trials assessing domains such as episodic and executive . Evidence regarding DHEA's impact on bone mineral density (BMD) is mixed, particularly across sexes and durations of supplementation. While some randomized trials report modest increases in and BMD among older women with low baseline DHEA levels after 12 months of at doses of 50 mg daily, effects in men are less pronounced or absent, and shorter-term studies often show null or inconsistent outcomes. Systematic evaluations highlight that, despite potential small benefits in bone metabolism markers, DHEA does not reliably prevent fractures or demonstrate superiority over in broader management, necessitating further long-term data. Findings on DHEA for depressive symptoms remain inconclusive, with meta-analyses of randomized trials showing small improvements in mild to moderate cases but tempered by methodological limitations, including small sample sizes and heterogeneity in dosing (typically 30-450 mg daily). One analysis of 8 trials reported a beneficial effect versus , yet cautioned against firm conclusions due to variable study quality and potential , while reviews of endogenous DHEA levels find no clear association with risk in postmenopausal women. In systemic applications for menopausal beyond vulvovaginal , a of 28 studies involving over 1,200 postmenopausal women found no significant improvements in overall sexual satisfaction or desire with DHEA supplementation, contrasting with localized intravaginal use and underscoring inconsistent systemic hormonal modulation.

Debunked or Unsupported Claims

Claims that oral dehydroepiandrosterone (DHEA) supplementation serves as an effective anti-aging therapy, such as reversing age-related declines in , muscle , or overall , lack support from randomized controlled trials. A review of 11 placebo-controlled studies found no meaningful benefits in these areas, with effects limited to minor increases in insulin-like growth factor-1 levels that did not translate to functional improvements. Similarly, preclinical suggestions of DHEA's role in preventing cancer or age-related diseases have not been substantiated in clinical trials, where outcomes showed no clear preventive efficacy. Assertions that systemic DHEA improves cognitive function in non-demented middle-aged or older adults are unsupported by evidence. A of randomized controlled trials concluded there is no convincing benefit for memory, executive function, or other cognitive domains, despite some early observational associations with DHEA levels. For postmenopausal women, claims of systemic DHEA alleviating symptoms like low or overall have been refuted by meta-analyses, which reported no significant improvements over in scores or quality-of-life measures. In fertility treatment, the notion that DHEA priming enhances outcomes for women with diminished or poor response to IVF has been debunked by multiple meta-analyses. These analyses, aggregating data from randomized trials, found no increases in clinical rates, live birth rates, or yield compared to controls, despite initial enthusiasm from small observational studies. Long-term therapeutic claims beyond vaginal prasterone for vulvovaginal remain unsubstantiated due to insufficient adequately powered trials demonstrating sustained efficacy or safety.

History

Discovery and Early Development

Dehydroepiandrosterone (DHEA), also known as prasterone, was first isolated from human urine in 1934 by German biochemist Adolf Butenandt and his colleague H. Dannenbaum at the Kaiser Wilhelm Institute for Biochemistry. This isolation occurred amid broader efforts to characterize steroid hormones, following Butenandt's earlier work on androsterone (1931) and contributions to understanding sex hormone structures, for which he shared the 1939 Nobel Prize in Chemistry. The compound was identified as a C19 steroid with a Δ5-3β-hydroxyl structure, distinguishing it from previously known androgens like androsterone, and its chemical formula was confirmed through crystallization and spectroscopic analysis. The sulfated form, dehydroepiandrosterone sulfate (DHEA-S), was identified approximately 10 years later in 1944, marking an early step in recognizing conjugated metabolites. By 1954, DHEA itself was isolated from peripheral , expanding knowledge of its systemic circulation beyond urinary excretion. In 1959, French biochemist Étienne-Émile Baulieu demonstrated that DHEA-S constitutes the predominant circulating form of the in humans, comprising up to 99% of total DHEA pool and highlighting its role as a stable reservoir for the free hormone. These findings established DHEA's biosynthesis primarily in the adrenal zona reticularis, with minor contributions from gonads and brain, via enzymes converting precursors. Early research in the and focused on DHEA's function as a weak and precursor to testosterone and estrogens, with in vitro conversions documented through enzymatic assays. Its decline with advancing age was first quantified in 1965, linking serum levels peaking in early adulthood (around 20-30 years) to a progressive drop of 80-90% by age 70, prompting initial hypotheses on adrenal dynamics. Synthetic routes, including partial from sterols like diosgenin, emerged in the mid-20th century to support biochemical studies, though therapeutic applications remained exploratory until later decades.

Key Milestones and Approvals

Dehydroepiandrosterone (DHEA), the active compound in prasterone, was first isolated from human urine in 1934 by and Hans Dannenbaum. Prior to regulatory approvals as a prescription , DHEA was marketed in the United States as a , particularly following the Dietary Supplement Health and Education Act of 1994, though it had been promoted as an unapproved drug product in various forms. The primary pharmaceutical milestone for prasterone occurred on November 17, 2016, when the U.S. (FDA) approved Intrarosa (prasterone vaginal inserts, 6.5 mg) for the treatment of moderate to severe —a symptom of vulvovaginal —due to in postmenopausal women. This approval represented the first FDA-authorized product containing prasterone as its , based on clinical trials demonstrating in improving vaginal tissue health without systemic hormonal effects. Subsequent international approvals expanded access: the () authorized Intrarosa in 2018 for the same indication, following positive committee recommendations on its benefit-risk profile. granted approval on November 6, 2019, aligning with the FDA and indications for postmenopausal vulvovaginal symptoms. The () in also approved prasterone (Intrarosa) for treating vulvar and vaginal in postmenopausal women, though specific launch timelines varied by market partnerships, such as licensing agreements with Theramex for , , and other regions in 2018.

Regulation and Societal Aspects

In the , prasterone (branded as Intrarosa in 6.5 mg intravaginal inserts) received approval from the (FDA) on November 17, 2016, specifically for treating moderate to severe pain during sexual intercourse (), a symptom of vulvovaginal , in postmenopausal women. This approval represented the first instance of the FDA authorizing a product with prasterone as its active ingredient for any indication. Oral or systemic formulations of dehydroepiandrosterone (DHEA), the chemical equivalent of prasterone, remain unapproved by the FDA as drugs and are instead sold over-the-counter as dietary supplements under an exemption from the , though the agency has issued warnings against unapproved therapeutic claims for such products. In the , the () authorized Intrarosa on January 8, 2018, for the same indication associated with vulvovaginal atrophy in postmenopausal women, with the marketing authorization valid across member states. similarly approved the 6.5 mg prasterone vaginal inserts for this use. Globally, DHEA faces stricter controls outside the ; in most countries, it is classified as a controlled , often requiring a prescription or facing outright bans for non-pharmaceutical use due to its potential conversion to testosterone and other androgens. Prasterone has not received regulatory approval for indications beyond vulvovaginal atrophy in jurisdictions where it is authorized, and systemic DHEA supplements are not recognized as medicines by major agencies like the FDA or for treating conditions such as menopausal symptoms outside localized vaginal application.

Use in Sports and Doping Controls

Prasterone, also known as dehydroepiandrosterone (DHEA), is classified as a prohibited substance under the (WADA) Prohibited List in the category of anabolic agents (S1.1a: anabolic androgenic steroids, exogenous). It is banned at all times, both in-competition and out-of-competition, due to its potential to serve as a precursor to endogenous androgens like testosterone, which could confer ergogenic advantages such as increased muscle mass and strength. This prohibition extends to major sporting bodies including the (USADA) and the (NCAA), where it is tested for in routine anti-doping controls via urine and blood analysis targeting the substance and its metabolites. Athletes have historically sought prasterone supplementation for purported performance-enhancing effects, including improved endurance, recovery, and , particularly in strength-based or contexts. Marketing of over-the-counter DHEA products often emphasizes these benefits, positioning it as a legal alternative to synthetic steroids in non-competitive settings, though such claims drive its illicit use in sports. However, for significant ergogenic benefits remains limited and context-dependent; short-term administration failed to enhance or anabolic markers in young recreational athletes, suggesting minimal impact in individuals with endogenous levels. In older populations or those with low baseline DHEA, supplementation combined with heavy resistance training has shown modest increases in muscle mass and strength, as demonstrated in a study where 50 mg daily doses amplified training-induced gains in elderly men and women. Conversely, reviews indicate that perceptions of DHEA's in or younger athletes outpace validation, with no consistent improvements in combined endurance-strength protocols or overall athletic output. Doping controls enforce zero-tolerance thresholds, with sanctions for positive tests including suspensions; detection relies on WADA-accredited labs quantifying prasterone above endogenous reference ranges, often adjusted for age and sex. Despite inconclusive performance data, its inclusion on prohibited lists reflects precautionary principles against any androgenic modulation in competitive sport.

Availability and Marketing

In the United States, prasterone (dehydroepiandrosterone, or DHEA) is widely available over-the-counter as an oral in forms such as capsules, tablets, and powders, with typical dosages ranging from 25 mg to 200 mg daily. This status stems from its classification under the Dietary Supplement Health and Education Act of 1994, which grandfathered ingredients marketed before October 15, 1994, allowing sales without FDA pre-approval for non-disease-related structure/function claims like supporting levels or . However, the FDA has explicitly stated that oral prasterone lacks approval for any therapeutic use and has issued warnings against its marketing as an unapproved new drug for conditions like aging or . A prescription-only vaginal insert formulation, Intrarosa (6.5 mg prasterone), was approved by the FDA on November 17, 2016, solely for treating moderate to severe associated with genitourinary syndrome of in postmenopausal women, via local conversion to active androgens and estrogens. In the , prasterone is not authorized as a and is restricted to prescription medicinal products due to regulatory prohibitions on its non-medicinal sale, reflecting concerns over inconsistent quality, potential hormonal risks, and insufficient evidence for broad claims. received marketing authorization from the on January 8, 2018, for vulvar and vaginal in postmenopausal women, with availability limited to this indication across member states. Certain ester forms, such as prasterone enanthate, are available by prescription in select European countries including , , and for in conditions like , typically via . In , Intrarosa is prescription-only for postmenopausal vulvovaginal , filed for review in 2016. Marketing of oral prasterone supplements in the focuses on purported benefits for , enhancement, muscle maintenance, and countering age-related declines in adrenal , often positioned as a "youth hormone" precursor despite limited clinical substantiation and FDA disclaimers against claims. Manufacturers must avoid implying prevention or to evade enforcement, though historical instances of unsubstantiated as an anti-aging or performance aid have prompted regulatory scrutiny. In contrast, Intrarosa's emphasizes evidence from phase III trials showing improvements in vaginal pH, epithelial integrity, and dyspareunia scores without systemic elevation, targeting gynecologists and postmenopausal patients via pharmaceutical channels. Global supplement marketing is curtailed outside the , where bans in countries like , the , and much of limit it to or veterinary contexts, underscoring divergent regulatory philosophies on endogenous precursors.

References

  1. [1]
    Prasterone: Uses, Interactions, Mechanism of Action - DrugBank
    A medication used vaginally to manage pain during sexual intercourse associated with changes in and around the vagina that are caused by menopause. Structure.
  2. [2]
    Dehydroepiandrosterone | C19H28O2 | CID 5881 - PubChem - NIH
    Prasterone, also known as dehydroepiandrosterone (DHEA) is a major C19 steroid produced by the adrenal cortex. It is also produced in small quantities in the ...
  3. [3]
    Prasterone (Intrarosa) - NCBI Bookshelf - NIH
    Prasterone is a synthetic form of dehydroepiandrosterone (DHEA), which is a natural steroid compound with no estrogenic, androgenic, or other hormonal activity.
  4. [4]
    Prasterone (vaginal route) - Side effects & dosage - Mayo Clinic
    Jan 31, 2025 · Prasterone is used to treat women with moderate to severe dyspareunia (painful intercourse) that is due to menopause.
  5. [5]
    Treatment of moderate to severe dyspareunia with intravaginal ...
    Prasterone has been shown to decrease the pain associated with dyspareunia, and to improve vaginal pH, as well as superficial and parabasal cell counts, while ...
  6. [6]
    Prasterone (Intrarosa) for Dyspareunia - AAFP
    Jan 15, 2019 · Prasterone (Intrarosa) is an intravaginal product used to treat moderate to severe dyspareunia due to vulvar and vaginal atrophy caused by menopause.
  7. [7]
    Prasterone, DHEA Capsules or Tablets - Cleveland Clinic
    DHEA may support sexual function. It may also improve energy and boost mood. The FDA has not evaluated this supplement for any medical use.
  8. [8]
    Prasterone - PubMed
    Initial trials investigated prasterone as a treatment to improve disease activity and symptoms in women with mild to moderate SLE.
  9. [9]
    Dehydroepiandrosterone Biosynthesis, Role, and Mechanism of ...
    Dehydroepiandrosterone (DHEA) is synthesized from cholesterol by activity of P450scc and P450c17, enzymes that we previously characterized in the developing ...
  10. [10]
    Overview of dehydroepiandrosterone biosynthesis - PubMed
    1 Division of Endocrinology and Metabolism, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas 75390-8857, ...
  11. [11]
    Development of a Chemoenzymatic Process for ... - ACS Publications
    Jul 5, 2016 · In this manuscript we describe a novel, concise, and cost-efficient route toward DHEA (2) and DHEA acetate (3) from 4-androstene-3,17-dione (4-AD, 1).
  12. [12]
    Processes for the preparation of dehydroepiandrosterone and its ...
    The above described synthetic processes for the preparation of dehydroepiandrosterone includes multiple steps, and a sequence of protection/deprotection steps ...
  13. [13]
    Prasterone enantate: Uses, Interactions, Mechanism of ... - DrugBank
    Feb 24, 2021 · Prasterone enantate is an estrogen indicated for hormone replacement therapy in postmenopausal women.Missing: derivative | Show results with:derivative
  14. [14]
    Plasma levels of dehydroepiandrosterone and 17 beta-estradiol ...
    Prasterone enanthate is completely hydrolysed into the free steroid and the fatty acid.
  15. [15]
    Dehydroepiandrosterone Sulfate - an overview | ScienceDirect Topics
    Dehydroepiandrosterone (DHEA) and its sulfate ester, DHEA sulfate, are C19 steroids synthesized in and secreted mostly from the adrenal gland and thus are ...
  16. [16]
    7-Keto-dehydroepiandrosterone | C19H26O3 - PubChem - NIH
    7-ketodehydroepiandrosterone is a 3beta-hydroxy-Delta(5)-steroid that is dehydroepiandrosterone carrying an additional oxo group at position 7.
  17. [17]
    A systematic review of the impact of 7-keto-DHEA on body weight
    7-Keto-DHEA has been commercially advertised as a dietary supplement to support weight loss. The objective of the present systematic review it to summarize ...
  18. [18]
    Dehydroepiandrosterone and its derivatives: potentially novel anti ...
    In addition, DHEA analogs such as 3beta-methyl-5-androsten-17-one, 16alpha-fluoro-5-androsten-17-one and 16alpha-fluoro-5alpha-androstan-17-one have been ...Missing: pharmaceuticals | Show results with:pharmaceuticals
  19. [19]
    Dehydroepiandrosterone (DHEA) and Synthetic DHEA Analogs Are ...
    This article reports on the effects of DHEA and its novel synthetic analogs 16α-fluoro-5-androsten-17-one (8354) and 3β-hydroxy-16α-fluoro-5α-androstan-17-one ( ...<|control11|><|separator|>
  20. [20]
    First synthesis of 7α- and 7β-amino-DHEA, dehydroepiandrosterone ...
    Efficient syntheses of new DHEA analogues, and their apoptotic and necrotic effects on Leydig cells and TM4 Sertoli cells are described.
  21. [21]
    Dehydroepiandrosterone and Its Metabolite 5-Androstenediol - MDPI
    The neurosteroid BNN27 is a novel synthetic derivative of DHEA, a synthetic C-17-spiro-dehydroepiandrosterone analogue. The compound is being studied and has ...
  22. [22]
    [PDF] 208470Orig1s000 - accessdata.fda.gov
    Nov 16, 2016 · DHEA (prasterone) is an endogenous steroid precursor that is secreted in the adrenal gland and is converted into active androgens and/or ...
  23. [23]
    [PDF] Intrarosa, INN-prasterone - European Medicines Agency
    PHARMACOLOGICAL PROPERTIES. 5.1 Pharmacodynamic properties ... Exogenous prasterone is metabolized in the same manner as endogenous prasterone.
  24. [24]
    Pharmacology and therapeutic effects of dehydroepiandrosterone in ...
    Three mechanisms of action of DHEA(S) have been identified. DHEA and DHEAS are precursors of testosterone and estradiol, DHEAS is a neurosteroid which modulates ...
  25. [25]
    Pharmacokinetic and Pharmacodynamic Studies in Healthy Elderly ...
    After DHEA administration, in both sexes there was a rapid and dose proportional increase in DHEAS levels. In men, 25 or 50 mg DHEA administration restored ...
  26. [26]
    Prasterone (Topical) Monograph for Professionals - Drugs.com
    Nov 18, 2024 · Prasterone (Topical) Pharmacokinetics. Absorption. Plasma Concentrations. Prasterone: Mean serum trough concentrations increased by 47% from ...
  27. [27]
    Intrarosa Dosage Guide - Drugs.com
    Jan 1, 2025 · Detailed dosage guidelines and administration information for Intrarosa (prasterone). Includes dose adjustments, warnings and precautions.Missing: clinical | Show results with:clinical
  28. [28]
    Clinical Review - Prasterone (Intrarosa) - NCBI Bookshelf
    Prasterone is indicated for the treatment of post-menopausal VVA. The sponsor has requested the reimbursement of prasterone as per the Health Canada indication.
  29. [29]
    Dosing and Administration
    Insert one INTRAROSA vaginal insert using the disposable applicator, each night at bedtime1; Individually wrapped vaginal inserts1.
  30. [30]
    [PDF] INTRAROSA® Prasterone vaginal ovules
    Sep 8, 2023 · 4 DOSAGE AND ADMINISTRATION. 4.1 Dosing Considerations. No dose adjustment is required in these situations: • elderly women (> 65 years of age ...
  31. [31]
    [PDF] 208470Orig1s000 - accessdata.fda.gov
    Nov 16, 2016 · The clinical pharmacology review team evaluated pharmacokinetic and pharmacodynamic data for the prasterone vaginal insert. The clinical ...
  32. [32]
    Efficacy of intravaginal dehydroepiandrosterone (DHEA ... - PubMed
    The daily intravaginal administration of 0.50% (6.5 mg) DHEA (Prasterone) has shown clinically and highly statistically significant effects on the four ...
  33. [33]
    Treatment of moderate to severe dyspareunia with intravaginal ...
    Prasterone is administered as a vaginal insert once daily at bedtime, does not carry a boxed warning in its label, and has no restrictions on duration of use.
  34. [34]
    intrarosa - Drug Summary
    Used to treat moderate to severe dyspareunia, a symptom of vulvar and vaginal atrophy associated with menopause.<|control11|><|separator|>
  35. [35]
    FDA: Prasterone therapy approved for vulvar, vaginal atrophy - Healio
    Nov 17, 2016 · The FDA today approved prasterone for treatment of moderate to severe pain during sexual intercourse, a symptom of vulvar and vaginal ...
  36. [36]
    FDA Approves Intrarosa for the Treatment of Dyspareunia - eMPR.com
    Nov 17, 2016 · The efficacy of Intrarosa was established in two 12-week placebo-controlled trials (n=406) of healthy postmenopausal women aged 40–80 years.<|separator|>
  37. [37]
    Intravaginal prasterone (DHEA) provides local action without ...
    The present data using low dose (6.5 mg) intravaginal prasterone (which exerts highly beneficial effects on VVA [12]) show that the average 24 h serum DHEA and ...Abstract · Serum Steroid Concentrations · References (27)
  38. [38]
    Pharmaceutical Approval Update - PMC - NIH
    This is the first agent approved by the Food and Drug Administration (FDA) to treat women experiencing moderate-to-severe pain during sexual intercourse ( ...
  39. [39]
    [PDF] 208470Orig1s000 - accessdata.fda.gov
    Nov 8, 2016 · Therefore the clinical trial data support the effectiveness of INTRAROSA (6.5 mg prasterone) vaginal inserts in the treatment of moderate to ...
  40. [40]
    [PDF] A Closer Look at the Women's Health PRN - ACCP
    Two clinical trials evaluated prasterone 6.5 mg (equivalent to DHEA 0.5%) in postmenopausal women with moderate to severe GSM over 12 weeks – one with an ...<|separator|>
  41. [41]
  42. [42]
    [PDF] Reference ID: 4014943 This label may not be the latest approved by ...
    The structural formula is: Each INTRAROSA (prasterone) vaginal insert contains 6.5 mg of prasterone in 1.3 ml of off-white hard fat (Witepsol).
  43. [43]
    Intrarosa (prasterone) FDA Approval History - Drugs.com
    Nov 17, 2016 · FDA Approved: Yes (First approved November 17, 2016) ; Brand name: Intrarosa ; Generic name: prasterone ; Dosage form: Vaginal Inserts ; Company: ...Missing: formulations | Show results with:formulations
  44. [44]
    Intrarosa: Package Insert / Prescribing Information - Drugs.com
    Jan 17, 2025 · 12.3 Pharmacokinetics. In a study conducted in postmenopausal women, administration of the INTRAROSA vaginal insert once daily for 7 days ...
  45. [45]
    prasterone, intravaginal (Rx) - Medscape Reference
    Indicated for moderate-to-severe dyspareunia caused by postmenopausal vulvar/vaginal atrophy. Administer 1 vaginal insert qDay at bedtime using applicator ...Missing: dehydroepiandrosterone | Show results with:dehydroepiandrosterone
  46. [46]
    Summary Basis of Decision for Intrarosa
    The most commonly reported adverse reaction to Intrarosa, reported in 8.3% of patients, was application site discharge. The leakage from the vagina may be ...
  47. [47]
    Sustained moderate to severe dyspareunia relief for up to 1 year ...
    In one 52-week open-label clinical trial, the most common adverse reactions with an incidence ≥2 percent were vaginal discharge and abnormal Pap smear.
  48. [48]
    Intrarosa | European Medicines Agency (EMA)
    Intrarosa is a medicine used to treat postmenopausal women with moderate to severe symptoms of vulvar and vaginal atrophy.
  49. [49]
    Systemic and local effects of vaginal dehydroepiandrosterone (DHEA)
    This secondary analysis evaluated the impact of vaginal DHEA on hormone concentrations, bone turnover and vaginal cytology in women with a cancer history.Missing: peer- | Show results with:peer-
  50. [50]
    [PDF] 4221038 This label may not be the latest approved by FDA. For ...
    The most common side effects of INTRAROSA vaginal inserts are vaginal discharge and changes on Pap smear. These are not all of the possible side effects of ...<|separator|>
  51. [51]
    [PDF] Intrarosa - 4138 - European Medicines Agency
    Nov 9, 2017 · Intrarosa is presented as vaginal pessaries containing 6.5 mg of active substance prasterone (DHEA), to be administered in the vagina, with or ...
  52. [52]
    The role for long-term use of dehydroepiandrosterone in adrenal ...
    Jun 1, 2022 · Patients with adrenal insufficiency will have a deficiency of DHEA. Unlike glucocorticoid and mineralocorticoid replacement, DHEA ...Missing: clinical | Show results with:clinical
  53. [53]
    Dehydroepiandrosterone Replacement in Women with Adrenal ...
    Sep 30, 1999 · The physiologic role of dehydroepiandrosterone in humans is still unclear. Adrenal insufficiency leads to a deficiency of ...
  54. [54]
    DHEA replacement in adrenal insufficiency - ScienceDirect.com
    Adrenal insufficiency. 2021, Nature Reviews Disease Primers ...
  55. [55]
    Dehydroepiandrosterone replacement in women with ... - PubMed
    Conclusions: Dehydroepiandrosterone improves well-being and sexuality in women with adrenal insufficiency.Missing: outcomes | Show results with:outcomes
  56. [56]
    No Benefit for Subjective Health Status and Sexuality in a 9-Month ...
    If a precise definition of the clinical syndrome of DHEA(S) deficiency ... Trials of DHEA Treatment Effects on Quality of Life in Women with Adrenal Insufficiency.
  57. [57]
    Long-Term DHEA Replacement in Primary Adrenal Insufficiency
    There was no significant benefit of DHEA treatment on fatigue or cognitive or sexual function. Supraphysiological DHEAS levels were achieved in some older ...Missing: outcomes | Show results with:outcomes
  58. [58]
    DHEA - Mayo Clinic
    DHEA also might increase the risk of hormone-sensitive cancers, including prostate, breast and ovarian cancers.
  59. [59]
    A Review of Age-Related Dehydroepiandrosterone Decline and Its ...
    In women, only minimal adverse effects have been reported such as mild acne, seborrhea, facial hair growth, and ankle swelling. No adverse effects were observed ...
  60. [60]
    A dose-response and meta-analysis of dehydroepiandrosterone ...
    DHEA supplementation is effective for increasing testosterone levels, although the magnitude varies among different subgroups.
  61. [61]
    DHEA in Elderly Women and DHEA or Testosterone in Elderly Men
    Oct 19, 2006 · Panel A shows a significant increase in median plasma levels of sulfated DHEA in elderly men and women after treatment with DHEA, as compared ...
  62. [62]
    Impact of DHEA supplementation on testosterone and estradiol ...
    Jul 4, 2025 · DHEA levels decline significantly with age, decreasing by ... The meta-analysis revealed a significant increase in testosterone levels in the DHEA ...
  63. [63]
    A Meta-Analysis Study of Placebo-Controlled Trials - Oxford Academic
    In addition, in females, DHEAS levels gradually decline with aging, although no specific reduction is associated with menopause (3–5). In postmenopausal ...
  64. [64]
    A Systematic Review and Meta-Analysis - Frontiers
    Mar 28, 2019 · There is a growing body of evidence suggesting that concentrations of DHEA and DHEA-S might decrease with age. ... The decline in DHEA-S levels ...
  65. [65]
    Dehydroepiandrosterone and age-related cognitive decline - PMC
    In humans the circulating concentrations of dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEAS) decrease markedly during aging, and have been implicated ...
  66. [66]
    DHEA supplements: Are they safe? Or effective? - Harvard Health
    Jan 27, 2025 · DHEA is generally considered safe for use for up to two years, at doses up to 50 milligrams (mg) per day. Mild side effects may include acne, ...Missing: adverse | Show results with:adverse
  67. [67]
    [PDF] DHEA supplementation: The claims in perspective
    ABSTRACT. Deficiency of dehydroepiandrosterone (DHEA) is associated with lupus erythematosus, diabetes mellitus,. Alzheimer disease, and some cancers, ...Missing: excretion | Show results with:excretion
  68. [68]
    Prasterone and Overactive Bladder in Postmenopausal Women
    Jun 12, 2025 · A meta-analysis of 11 studies showed a significant reduction in urinary frequency, urgency, and urge urinary incontinence, all hallmark symptoms ...
  69. [69]
    Intravaginal 6.5 mg prasterone administration in postmenopausal ...
    Jun 12, 2021 · Prasterone is an inactive precursor converted into estrogens and androgens into vaginal tissue. It leads to positive effects on VVA through ...
  70. [70]
    Study Details | NCT03854396 - Clinical Trials
    Large, prospective studies have shown prasterone to safely decrease vaginal pH, decrease parabasal cells, increase superficial cells, and decrease symptoms ...
  71. [71]
    Intravaginal prasterone for urinary urgency in postmenopausal ...
    The present study assessed the effects of intravaginal prasterone on urgency symptoms in those patients receiving treatment for concurrent vulvovaginal atrophy.
  72. [72]
    Intravaginal prasterone for urinary urgency in postmenopausal ...
    Prasterone is a prohormone transformed into its active metabolite dehydroepiandrosterone sulfate (DHEA-S), and then into androgen and estrogen by ...
  73. [73]
    Dehydroepiandrosterone for women in the peri- or postmenopausal ...
    Jan 22, 2015 · Dehydroepiandrosterone (DHEA) is one of the main precursors of androgens, which in turn are converted to testosterone and estrogens. It is ...
  74. [74]
    Dehydroepiandrosterone treatment of women with mild‐to ...
    Nov 8, 2002 · The overall results confirm that DHEA treatment was well-tolerated, significantly reduced the number of SLE flares, and improved patient's ...
  75. [75]
    Dehydroepiandrosterone (DHEA) for lupus erythematosus - Cochrane
    Oct 17, 2007 · DHEA may improve disease activity in people with severe or active lupus, but this result is based on low quality evidence so there is not enough evidence to be ...
  76. [76]
    Dehydroepiandrosterone in systemic lupus erythematosus - PMC
    Serum levels of DHEA are decreased in patients with inflammatory disease including lupus, and these levels seem to inversely correlate with disease activity.
  77. [77]
    Dehydroepiandrosterone Monotherapy in Midlife-Onset Major and ...
    We find DHEA to be an effective treatment for midlife-onset major and minor depression. Midlife in both men and women is characterized by a decline in the ...Missing: prasterone disturbances
  78. [78]
    A systematic review and meta‐analysis of randomized controlled trials
    Sep 15, 2020 · In conclusion, DHEA was associated with a beneficial effect on depressive symptoms compared to placebo. However, these results should be viewed ...
  79. [79]
    A systematic review and meta-analysis of randomized controlled trials
    Sep 15, 2020 · DHEA was associated with a beneficial effect on depressive symptoms compared to placebo. However, these results should be viewed with caution.
  80. [80]
    DHEA and response to antidepressant treatment: A Mendelian ...
    The aim of this study is to evaluate whether higher levels of DHEA(S) are causally linked to response to antidepressants using mendelian randomization (MR).
  81. [81]
    Endogenous DHEAS Is Causally Linked With Lumbar Spine Bone ...
    A recent pooled analysis of four clinical trials demonstrated that treatment with dehydroepiandrosterone (DHEA) increases lumbar spine bone mineral density ...Abstract · Materials and Methods · Results · Discussion
  82. [82]
    DHEA therapy may preserve bone, muscle mass in older women
    Nov 27, 2018 · In that study, women prescribed DHEA experienced a mean increase in lumbar spine BMD of 1.7% at 12 months and a further increase of 1.8% at 24 ...
  83. [83]
    A systematic review and meta-analysis - PubMed - NIH
    Apr 22, 2022 · A systematic review of seven RCTs found that DHEA monotherapy does not improve bone mineral density (BMD) compared with placebo after adjusting for weight gain.
  84. [84]
    Dehydroepiandrosterone and Bone Health: Mechanisms and Insights
    Dec 6, 2024 · Evidence from prior studies suggests that DHEA supplementation may enhance bone mineral density (BMD) and confer beneficial effects on bone ...
  85. [85]
    Dehydroepiandrosterone: a potential therapeutic agent in the ...
    Aug 2, 2019 · We review here the evidence for this potential therapeutic agent in the treatment and rehabilitation of the severely injured patient.
  86. [86]
    Dehydroepiandrosterone (DHEA): Pharmacological Effects and ...
    Evidence from clinical studies supports the use of DHEA in hypoadrenal individuals and in treating depression and associated cognitive disorders.
  87. [87]
    FDA approves vaginal insert to treat dyspareunia in menopause
    Nov 18, 2016 · The approval is based on the results of two 12-week placebo-controlled trials of 406 healthy, postmenopausal women, ranging in age from 40 to 80 ...<|control11|><|separator|>
  88. [88]
    Vaginal dehydroepiandrosterone compared to other methods ... - NIH
    Jan 7, 2021 · Vaginal DHEA has been found to be an effective and safe treatment in menopausal women with symptoms of vaginal atrophy.
  89. [89]
    (PDF) The Effectiveness of Prasterone vs Placebo Therapy as the ...
    Aug 6, 2025 · This meta-analysis concludes that Prasterone therapy has a significant therapeutic effect for Vulvovaginal Atrophy in menopausal women.
  90. [90]
    Review Dehydroepiandrosterone intra vaginal administration for the ...
    The effects of intravaginal administration of dehydroepiandrosterone (DHEA) for the management of symptomatic vulvovaginal atrophy are reviewed.
  91. [91]
    a systematic review of randomized clinical trial data - PubMed
    Nov 1, 2023 · Overall, this systematic review does not support a beneficial effect of DHEA therapy on cognitive performance in postmenopausal women.Missing: inconclusive | Show results with:inconclusive
  92. [92]
    Effects of Dehydroepiandrosterone Replacement Therapy on Bone ...
    Conclusions: DHEA replacement therapy for 1 yr improved hip BMD in older adults and spine BMD in older women.
  93. [93]
    Endogenous dehydroepiandrosterone and depression in ... - PubMed
    Jan 17, 2023 · This systematic review does not support an association between endogenous DHEA/DHEAS and depression in postmenopausal women.
  94. [94]
    Dehydroepiandrosterone sulfate supplementation in health and ...
    Apr 18, 2023 · They observed that vaginal DHEA (Prasterone) significantly relieved dyspareunia with improvement in vaginal secretions and epithelial integrity.<|control11|><|separator|>
  95. [95]
    Dehydroepiandrosterone, Cancer, and Aging - PMC - PubMed Central
    DHEA has preventive and therapeutic efficacy in treating major age-associated diseases, including cancer, atherosclerosis, diabetes, obesity.
  96. [96]
    Testosterone and Dehydroepiandrosterone Treatment in Ageing Men
    Table 1 [10,13,14,15,16] shows selected adverse effects of low T and low DHEA levels and illustrates the versatility of negative consequences that arise from ...
  97. [97]
    Benefits and Harms of Systemic Dehydroepiandrosterone (DHEA) in ...
    Oct 1, 2014 · We conducted a systematic review and meta-analysis to summarize the evidence supporting the use of systemic DHEA in postmenopausal women with ...
  98. [98]
    Efficacy of dehydroepiandrosterone priming in women with poor ...
    Jun 9, 2023 · Our data did not show that DHEA supplementation significantly improved clinical pregnancy rate and live birth rate in patients with DOR/POR in a ...
  99. [99]
    The effect of dehydroepiandrosterone (DHEA) supplementation on ...
    The results from recently randomized controlled trials and meta-analyses showed that pre-treatment with DHEA for IVF in women with DOR did not improve the ...
  100. [100]
    Dehydroepiandrosterone Uses, Benefits & Dosage - Drugs.com
    Jan 22, 2025 · DHEA is recommended as third-line monotherapy or adjunctive therapy for treatment of major depressive disorder (MDD) by Canadian Network for Mood and Anxiety ...
  101. [101]
    The Sex Hormone Precursors Dehydroepiandrosterone (DHEA) and ...
    Dehydroepiandrosterone (DHEA) and its sulfate ester form DHEAS, are multifunctional steroid hormones primarily produced in the adrenal cortex, ...
  102. [102]
    Adolf Butenandt – Biographical - NobelPrize.org
    In 1931 he isolated androsterone in pure, crystalline form. From androsterone he as well as Ruzicka, independently of each other, obtained testosterone in ...Missing: early dehydroepiandrosterone
  103. [103]
    Dehydroepiandrosterone sulfate and longevity: New clues for an old ...
    Dehydroepiandrosterone (DHEA) was isolated in urine in 1934, and DHEA 3β-sulfate (DHEAS) was identified 10 years later (1, 2). It took another decade to ...
  104. [104]
    Dehydroepiandrosterone (DHEA) (Prasterone) - SpringerLink
    In 1954, DHEA was isolated from the blood, and in 1959 the chemist E.E. Baulieu discovered that DHEA-S was the most abundant form of the hormone found in human ...
  105. [105]
    Dehydroepiandrosterone Research: Past, Current, and Future
    Mar 16, 2018 · This chapter highlights the founding research on DHEA from a historical perspective, provides an overview of DHEA biosynthesis and metabolism.
  106. [106]
    Dehydroepiandrosterone Research: Past, Current, and Future
    This chapter highlights the founding research on DHEA from a historical perspective, provides an overview of DHEA biosynthesis and metabolism.
  107. [107]
    FDA Approves Intrarosa for Sexual Symptoms of Menopause
    Jan 6, 2017 · The FDA approved Intrarosa to treat pain during sexual intercourse, but it's not clear if the product is safe for women who've been diagnosed with breast ...Missing: formulations | Show results with:formulations
  108. [108]
    [PDF] CENTER FOR DRUG EVALUATION AND RESEARCH
    Nov 16, 2016 · prasterone. Sponsor: EndoCeutics Inc. Approval Date: November 16, 2016. Indication: For the treat of moderate to severe dyspareunia, a symptom ...
  109. [109]
    Endoceutics Receives Health Canada Approval for INTRAROSA
    Nov 6, 2019 · INTRAROSA was approved in the US and in Europe in 2016 and 2018 ... Prasterone, also known as dehydroepiandrosterone (DHEA), is a ...
  110. [110]
    Intrarosa | Therapeutic Goods Administration (TGA)
    TGA decision: Intrarosa (prasterone) is approved to treat vulvar and vaginal atrophy in postmenopausal women.
  111. [111]
    Theramex to Exclusively License Intrarosa® In Europe, Australia ...
    Jul 19, 2018 · Intrarosa was first approved by the US Food and Drug Administration in November 2016 for the treatment of moderate to severe dyspareunia, a ...
  112. [112]
    What Should Athletes Know about DHEA? - Usada
    In almost all countries except the U.S., DHEA is treated as a controlled anabolic steroid. In the U.S., DHEA is exempt from the Controlled Substances Act, ...Missing: worldwide | Show results with:worldwide
  113. [113]
    Dehydroepiandrosterone (DHEA) - Medscape Reference
    Sep 4, 2025 · It is produced in the adrenal glands during cortisol and aldosterone formation. The de novo synthesis of DHEA occurs in the testes or the ...<|separator|>
  114. [114]
    [PDF] international standard - prohibited list - WADA
    The Prohibited List is a mandatory International Standard as part of the World. Anti-Doping Program. ... Prasterone (dehydroepiandrosterone, DHEA,. 3ß- ...
  115. [115]
    [PDF] 2022-23 NCAA Banned Substances
    The athletics director or athletics director's designee shall disseminate the list of banned-drug classes ... THERE IS NO COMPLETE LIST OF BANNED SUBSTANCES.
  116. [116]
    Exogenous DHEA administration and performance - WADA
    In conclusion, these data indicate that our short-term DHEA administration did not improve performance or have an anabolic effect in young female recreationally ...Missing: Prasterone | Show results with:Prasterone
  117. [117]
    DHEA enhances effects of weight training on muscle mass and ...
    This study provides evidence that DHEA replacement has the beneficial effect of enhancing the increases in muscle mass and strength induced by heavy resistance ...
  118. [118]
    Dehydroepiandrosterone to enhance physical performance - PubMed
    The widely perceived performance-enhancing activity of DHEA is still more myth than reality. However, because studies in female athletes are still lacking,
  119. [119]
    Lack of Dehydroepiandrosterone Effect on a Combined Endurance ...
    DHEA supplementation has been reported to potentiate the effects of heavy resistance training on muscle mass and performance in the elderly (6). Here, we ...Subjects And Methods · Statistical Analyses · Results<|separator|>
  120. [120]
    The Prohibited List | World Anti Doping Agency - WADA
    Jun 1, 2019 · If you cannot find a Substance or Method listed, please verify its status with your Anti-Doping Organization for accurate and authoritative ...Athletes & Support Personnel · Liste des interdictions · Footer
  121. [121]
    Review DHEA, physical exercise and doping - ScienceDirect.com
    WADA legislation. DHEA figures on the World Anti-Doping Agency (WADA) list of prohibited substances in sport and its use is prohibited both in competition ...
  122. [122]
    Mundipharma and Endoceutics to Exclusively License Intrarosa ...
    May 20, 2018 · Intrarosa™ was approved by the FDA in the US and by EMA for all countries of Europe. As the unique intracellular precursor of sex steroids in ...<|control11|><|separator|>
  123. [123]
    Regulatory Decision Summary for Intrarosa
    Prescription status: Intrarosa is available by prescription only. Date filed: 2016-10-05. Contact: Bureau of Metabolism, Oncology and Reproductive Science ...