Estrone is a naturally occurring steroid hormone and one of the three primary estrogens in humans, characterized by its aromatized C18 steroid structure featuring a 3-hydroxyl group and a 17-ketone functional group, with the molecular formula C₁₈H₂₂O₂.[1] It serves as a key female sex hormone, playing essential roles in reproductive physiology, particularly during menopause when it becomes the predominant circulating estrogen.[2]Estrone is biosynthesized primarily through the aromatization of androstenedione in peripheral tissues such as adipose tissue, ovaries, and the placenta during pregnancy.[1] In premenopausal women, it is produced in smaller amounts compared to estradiol, but its levels increase post-menopause due to enhanced extragonadal conversion from adrenal androgens.[2] The hormone can be reversibly converted to estradiol by the enzyme 17β-hydroxysteroid dehydrogenase, allowing interconversion between these estrogens in vivo.[1]Physiologically, estrone binds to estrogen receptors (ERα and ERβ) to regulate the development and maintenance of female secondary sexual characteristics, modulate gonadotropin secretion via negative feedback on the hypothalamic-pituitary axis, and support bone density conservation.[1] It also influences cardiovascular function, lipid metabolism, and neuroendocrine processes, though its weaker estrogenic potency compared to estradiol means it often acts as a reservoir for more active forms.[2] In men, trace amounts contribute to overall estrogen balance.Medically, estrone is utilized in hormone replacement therapy (HRT) to alleviate menopausal symptoms such as hot flashes and vaginal atrophy, and to prevent postmenopausal osteoporosis.[1] However, its use is associated with risks including increased chances of endometrial hyperplasia, breast cancer, stroke, and deep vein thrombosis, necessitating careful monitoring and risk-benefit assessment.[1] Elevated circulating levels of estrone have been linked to obesity and certain hormone-dependent cancers, highlighting its role as a biomarker in endocrine disorders.[3]
Chemistry
Structure and nomenclature
Estrone is an organic compound with the molecular formula C18H22O2 and a molecular weight of 270.366 g/mol.[1][4] Its systematic IUPAC name is 3-hydroxyestra-1,3,5(10)-trien-17-one, reflecting the specific positioning of functional groups on the estrane backbone.[1]The core structure of estrone consists of a steroidal skeleton, characterized by four fused rings labeled A, B, C, and D. Ring A is phenolic and aromatic, featuring a hydroxyl group at position 3 and conjugated double bonds that contribute to its estrogenic properties; ring B includes a double bond between C5 and C10; rings C and D form the saturated backbone with a methyl group at C13. At position 17 on ring D, estrone bears a keto (oxo) group, distinguishing its chemical identity within the estrogen family.[1][5]Estrone is classified as an estrane steroid, belonging to the subclass of estrogens and derivatives, which are C18 steroids with a 3-hydroxylated estrane nucleus. It is one of the three primary endogenous estrogens in mammals, alongside estradiol and estriol, playing a key role in hormonal physiology.[5][6][7]Regarding isomers and stereochemistry, estrone features a specific chiral configuration at key asymmetric centers, including (8R,9S,13S,14S), which is typical of naturally occurring steroid hormones. The 17-keto configuration sets estrone apart from its close analog estradiol, which instead has a 17β-hydroxyl group, resulting in distinct biological activities despite their structural similarity. While estrone lacks stereoisomerism at C17 due to the planar keto moiety, variations in ring fusions or substituents can yield synthetic isomers, though the natural form adheres to the standard gonane-derived stereochemistry.[1][8][9]
Physical and chemical properties
Estrone is a white to off-white crystalline powder.[1] Its melting point ranges from 258 to 261 °C, reflecting its thermal stability as a solid under standard conditions.[1][5]Estrone demonstrates poor aqueous solubility, with a reported value of 1.30 mg/L in pure water at 25 °C, which limits its dissolution in biological fluids without solubilizing agents.[10] In contrast, it is readily soluble in organic solvents, including ethanol (approximately 4 mg/mL at 15 °C), acetone (20 mg/mL), and dioxane (50 mg/mL), facilitating its use in laboratory and pharmaceutical preparations.[1][11]Estrone is generally stable in air at room temperature but exhibits sensitivity to light, undergoing photodegradation with half-lives of 2–3 hours under UV irradiation in aqueous environments, and to aerial oxidation, necessitating storage under inert conditions to maintain integrity.[1][12]The phenolic hydroxyl group at the 3-position imparts acidic character to estrone, with a pKa of approximately 10.4, enabling ionization in basic media and influencing its polarity and solubility profile.[5] This group is reactive toward conjugation, forming sulfate or glucuronide esters at the 3-position through esterification, a process that enhances water solubility for excretion.[1]
Biochemistry
Biosynthesis
Estrone is primarily synthesized in the body through the peripheral aromatization of androstenedione, a key androgen precursor, catalyzed by the enzymearomatase (encoded by the CYP19A1 gene).[13] This rate-limiting step occurs predominantly in adipose tissue, where aromatase converts androstenedione directly into estrone, as well as in the ovaries and placenta.[14] In adipose tissue, particularly in postmenopausal women, this extraglandular pathway becomes the dominant source of estrone production due to the abundance of the enzyme and the availability of circulating androgens.[15]An alternative biosynthetic route for estrone involves the oxidation of estradiol, the more potent estrogen, mediated by 17β-hydroxysteroid dehydrogenase type 2 (17β-HSD2).[16] This reversible interconversion between estrone and estradiol allows for dynamic regulation of estrogen activity, with 17β-HSD2 favoring the inactivation of estradiol to estrone in certain tissues.[17] In premenopausal women, ovarian production of estrone is tightly regulated by gonadotropins: follicle-stimulating hormone (FSH) induces aromatase expression in granulosa cells, while luteinizing hormone (LH) promotes androgen synthesis in theca cells, providing substrates for aromatization.[18] Postmenopause, with ovarian function declining, estrone synthesis shifts to adipose tissue, where it accounts for the majority of residual estrogen production and becomes the principal circulating estrogen.[19]Quantitatively, estrone represents approximately 20–40% of total circulating estrogens (primarily estrone and estradiol) in premenopausal cycling women, where estradiol predominates, but its contribution rises dramatically to nearly 100% in postmenopausal women due to the reliance on peripheral aromatization.[20] During pregnancy, placental biosynthesis of estrone intensifies, drawing on dehydroepiandrosterone sulfate (DHEAS) precursors from both maternal and fetal adrenal sources to support elevated estrogen levels essential for gestation.[21] This fetal-placental unit ensures high estrone output, primarily through aromatase activity in the syncytiotrophoblast.[22]
Metabolism
Estrone undergoes extensive biotransformation in the body, primarily through phase I oxidation and phase II conjugation reactions, leading to its inactivation and preparation for elimination. The primary metabolic pathways involve conjugation to form more water-soluble derivatives and oxidative modifications in the liver. These processes occur mainly in the liver and target tissues, regulating estrone's bioavailability and activity.[23]The main primary metabolites of estrone are estrone sulfate and estrone glucuronide, formed via sulfation and glucuronidation, respectively. Sulfation is catalyzed by estrogen sulfotransferase SULT1E1, particularly at low physiological concentrations of estrone, producing estrone-3-sulfate, a major circulating form that serves as a reservoir for active estrogens. Glucuronidation occurs primarily through UDP-glucuronosyltransferase UGT1A8, yielding estrone-3-glucuronide, which enhances solubility for further processing. Additionally, estrone can be metabolized to estriol through 16α-hydroxylation followed by reduction at the 17-position.[24][24][23]In the liver, estrone is subject to cytochrome P450-mediated hydroxylation, generating key hydroxylated metabolites such as 2-hydroxyestrone and 16α-hydroxyestrone. 2-Hydroxyestrone formation is primarily catalyzed by CYP1A1, CYP1A2, and CYP1B1 enzymes, while 16α-hydroxyestrone is mainly formed by CYP3A4 and related enzymes; 2-hydroxyestrone is considered protective and 16α-hydroxyestrone potentially more genotoxic due to its reactivity. These metabolites often undergo subsequent conjugation to facilitate their removal.[23][25]Estrone is reversibly interconverted with the more potent estradiol in target tissues, such as the breast and endometrium, via reduction by 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1). This enzyme catalyzes the NADPH-dependent reduction of estrone to estradiol, amplifying local estrogenic effects, while the reverse oxidation is mediated by other 17β-HSD isoforms.[26]The plasma half-life of unconjugated estrone is short, ranging from 10 to 70 minutes, reflecting rapid metabolism and clearance. However, conjugation to sulfate or glucuronide significantly prolongs its half-life, allowing estrone sulfate to persist for hours as a stable reservoir.[27][28]Genetic variations in CYP1B1, such as the Val432Leu polymorphism (rs1056836), influence the ratio of 4-hydroxyestrone to 2-hydroxyestrone. The cited study found no significant association with breast cancer risk but noted modestly higher estradiol levels in Leu allele carriers. These polymorphisms affect enzymatic activity and metabolite profiles, highlighting interindividual differences in estrone metabolism.[29]
Distribution and levels
Estrone circulates in the blood primarily bound to proteins, with greater than 95% bound to plasma proteins, primarily albumin, and a smaller fraction to sex hormone-binding globulin (SHBG), leaving about 2% in the free, biologically active form.[1] In plasma, estrone concentrations vary by sex and physiological state; in premenopausal women, levels typically range from 17 to 200 pg/mL during the follicular phase and can rise to 37 to 200 pg/mL in the luteal phase.[30] In men, plasma levels are lower, generally ranging from 10 to 60 pg/mL.[30] Postmenopause, estrone becomes the predominant circulating estrogen, with levels ranging from 7 to 40 pg/mL, though some assays report broader ranges up to 125 pg/mL depending on individual factors.[30][31]Tissue distribution of estrone is notable for high concentrations in adipose tissue, where local aromatization of androstenedione contributes significantly to its production, particularly in postmenopausal women.[32] Levels are low in children, remaining undetectable to 29 pg/mL prepubertally, and increase during puberty to peak in the reproductive years before declining slightly postmenopause, when estrone dominates over estradiol.[30] In males, estrone is present at minimal levels systemically but is produced in the testes via aromatization.[30]Clinical assessment of estrone levels employs methods such as radioimmunoassay (RIA) or liquid chromatography-tandem mass spectrometry (LC-MS/MS), with the latter offering higher specificity and sensitivity for low concentrations.[30] Factors influencing estrone levels include obesity, which elevates concentrations through increased adipose aromatase activity, and liver disease, which can disrupt SHBG production and alter protein binding.[30][31]
Excretion
Estrone and its metabolites are primarily eliminated from the body through renal and biliary routes, with conjugated forms (glucuronides and sulfates) being the main excreted species.[33] The liver conjugates estrone via sulfation and glucuronidation to form water-soluble derivatives, which facilitates their elimination.[33]The major portion of these conjugates undergoes biliary secretion into the intestine, where they contribute to fecal elimination, representing approximately 5-10% of total estrogen output after accounting for reabsorption.[34] However, due to enterohepatic circulation, a significant proportion—up to 80%—of biliary conjugates is deconjugated by intestinal bacteria and reabsorbed into the portal circulation, thereby prolonging the half-life of estrone in the body.[35] In the gut, bacterial β-glucuronidase and sulfatase enzymes hydrolyze the conjugates, releasing unconjugated estrone for potential reuptake or fecal loss.[36]Renal excretion accounts for the bulk of net elimination, with about 80-90% of estrogen metabolites appearing in urine as conjugates.[33]Estrone glucuronide is the predominant urinary form, alongside sulfate conjugates and other hydroxylated metabolites.[33] Total urinary estrogen excretion in women typically ranges from 5 to 20 μg per day, varying with menstrual cycle phase and physiological state.[37]Fecal elimination primarily involves unconjugated estrone resulting from intestinal deconjugation of biliary conjugates, with minimal direct unconjugated excretion from other sources.[34]Impaired hepatic or renal function disrupts these pathways, reducing conjugation efficiency in the liver or clearance via the kidneys, which leads to elevated circulating levels of estrone and its metabolites.[33]
Estrone functions primarily as a ligand for the estrogen receptors ERα (encoded by ESR1) and ERβ (encoded by ESR2), acting as an agonist at both subtypes to initiate estrogenic signaling. Its bindingaffinity is substantially lower than that of 17β-estradiol, with relative ligand binding affinities of approximately 4.5% for ERα and 4.0% for ERβ, corresponding to EC50 values for coactivator recruitment in the range of 30–75 nM depending on the receptor and assay conditions. This reduced potency arises from the 17-keto group in estrone, which diminishes the stability of receptor-ligand interactions compared to the 17β-hydroxyl group in estradiol, leading to weaker overall estrogenic activity—typically 1/10th to 1/20th that of estradiol across various assays.[38]In the classical genomic pathway, estrone-bound estrogen receptors undergo conformational changes that promote dimerization, either as homodimers (ERα-ERα or ERβ-ERβ) or heterodimers (ERα-ERβ), followed by nuclear translocation. The dimer then binds to specific DNA sequences known as estrogen response elements (ERE) in the promoter regions of target genes, recruiting coactivators such as SRC-3 to modulate transcription. For instance, this mechanism upregulates the expression of genes like the progesterone receptor (PGR), which is critical for coordinated hormonal responses in target tissues. Additionally, estrone influences hepatic production of proteins such as sex hormone-binding globulin (SHBG) and thyroid-binding globulin (TBG), altering circulating hormone levels.[5][39]Estrone also elicits rapid non-genomic effects through membrane-associated forms of ERα and ERβ, as well as potentially G protein-coupled estrogen receptor 1 (GPER1), bypassing nuclear transcription. These actions occur within seconds to minutes and involve activation of intracellular signaling cascades, including the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway and the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, leading to outcomes such as calcium mobilization and cell proliferation modulation. In pituitary tumor cells expressing high levels of membrane ERα, estrone activates ERK oscillations with potency comparable to estradiol at concentrations as low as 10^{-12} M, though it exhibits a longer delay in calcium signaling compared to estradiol.[40][41]The tissue selectivity of estrone's actions is influenced by the differential distribution and affinity preferences of ER subtypes, with ERα predominating in reproductive tissues like the uterus and mammary gland, where estrone exerts effects despite its lower potency. Furthermore, estrone is metabolized to 2-methoxyestrone, which exhibits antioxidant properties by acting as an electron donor and inhibiting lipid peroxidation, potentially contributing to protective effects independent of classical ER signaling.[42][43]
Physiological roles
Estrone, an endogenous estrogen with lower potency than estradiol but higher than estriol, plays a supportive role in reproductive physiology, primarily by contributing to endometrial proliferation during the menstrual cycle's proliferative phase, though its effects are less pronounced than those of estradiol.[44][45] It also aids in cervicalmucus production, facilitating sperm transport, as part of the broader estrogen-mediated changes in cervical secretions.[46] Compared to estradiol, estrone serves as a minor contributor to pubertal development and menstrual cycle regulation, acting mainly as a precursor and reservoir for more potent estrogens.[47]In postmenopausal women, where estrone becomes the predominant circulating estrogen due to peripheral aromatization in adipose tissue, it helps maintain bone density by inhibiting osteoclast activity and promoting osteoblast function, thereby mitigating age-related bone loss.[48] Estrone also supports vascular health through effects on endothelial cells, including enhanced proliferation and tube formation, with minimal effects on migration, which contribute to cardiovascular protection.[49] Additionally, its involvement in thermoregulation may underlie disruptions leading to hot flashes, a common menopausal symptom.[2]Beyond reproduction, estrone exerts non-reproductive effects, including neuroprotection primarily via activation of estrogen receptor β (ERβ) in the brain, where it modulates neuronal survival and reduces oxidative stress.[50] In the cardiovascular system, estrone, as an estrogen, contributes to vasodilation and improved endothelial function, potentially reducing the risk of atherosclerosis, though with lower potency than estradiol.[51] Endogenous estrone levels have been associated with lower high-density lipoprotein (HDL) cholesterol and higher triglyceride levels in postmenopausal women.[52]In males, where estrone levels are low but present through aromatization of androgens, it supports spermatogenesis by regulating germ cell development and fluid reabsorption in the efferent ducts of the testis.[53] It also contributes to prostate health by balancing epithelial and stromal cell proliferation via estrogen receptors, preventing excessive growth.[54]Pathophysiologically, elevated estrone levels are associated with endometriosis, where local increases in endometriotic tissues promote lesion growth and inflammation through enhanced estrogen signaling.[55] Conversely, estrone deficiency, often occurring in hypogonadism, exacerbates symptoms such as reduced bone density and metabolic disturbances due to impaired estrogen-mediated homeostasis.[56]
Medical use
Hormone replacement therapy
Estrone has historically been employed in hormone replacement therapy (HRT) to address estrogen deficiency in menopausal and hypogonadal women, primarily through intramuscular injections of aqueous suspensions. Under the brand name Theelin, it was administered in doses ranging from 0.1 to 5 mg to alleviate symptoms of menopause.[1] These injectable formulations, introduced in the early 20th century, are now rarely utilized due to their inconvenience and have been supplanted by more patient-friendly oral and transdermalestradiol options that provide steadier estrogen delivery.[57]In HRT, estrone is indicated for the management of vasomotor symptoms such as hot flashes, treatment of vulvovaginal atrophy, and prevention of postmenopausal osteoporosis in women at high risk for fractures.[1] Historical dosing regimens typically involved 0.1 to 2 mg administered intramuscularly every 1 to 3 weeks.[5] When used in women with an intact uterus, estrone is combined with progestins to counteract the risk of endometrial hyperplasia.[57]Estrone effectively relieves vasomotor and genitourinary symptoms of menopause. Recent 2024-2025 clinical guidelines and studies highlight the role of low-dose conjugated equine estrogens—predominantly estrone sulfate—in supporting long-term bone health, with evidence showing sustained increases in bone mineral density and reduced fracture incidence in postmenopausal women.[58]
Other indications
In the mid-20th century, estrone and its sulfate conjugate were utilized in estrogen replacement therapies to address conditions associated with estrogen deficiency, including amenorrhea and dysmenorrhea. For instance, soluble estrone sulfate was administered to alleviate symptoms of estrogen deficiency, helping to restore menstrual regularity in affected women during the 1950s.[59]Investigational uses of estrone include its potential role in transgenderhormone therapy, where it serves as a weaker estrogenagonist compared to estradiol, with relative binding affinities of 4-10% to estrogen receptor α and 2-3.5% to β, making it less commonly prescribed due to reduced potency in feminizing effects.[60] Recent studies as of 2025 have examined estrone conjugates, such as GLP-1-estrogen hybrids, for Alzheimer's disease symptom management, demonstrating neuroprotective benefits through enhanced mitochondrial function and reduced neuroinflammation in preclinical models.[61]Diagnostically, measurement of urinary estrone-3-glucuronide (E1G) levels provides a non-invasive marker for assessing ovarian function, reflecting follicular development and estrogen production throughout the menstrual cycle. Elevated E1G concentrations indicate active ovarian steroidogenesis, aiding in the evaluation of fertility and ovulatory status in clinical settings.[62]
Safety profile
Contraindications
Estrone therapy is absolutely contraindicated in individuals with a known or suspected history of estrogen-sensitive cancers, including breast, endometrial, and ovarian cancers, due to the potential for hormone stimulation of tumor growth via estrogen receptors.[57] It is also prohibited in cases of undiagnosed abnormal vaginal bleeding, as this may indicate underlying endometrial pathology that could be exacerbated by estrogen exposure.[57] Active or recent thromboembolic disorders, such as deep veinthrombosis or pulmonary embolism, represent another absolute contraindication, given the prothrombotic effects of estrogens.[57]Relative contraindications include liver dysfunction, such as active liver disease or cholestasis, where estrogen metabolism may be impaired, leading to elevated levels and heightened toxicity risk.[63]Migraine with aura is considered a relative contraindication owing to the increased stroke risk associated with estrogen use in this population.[64] Uncontrolled hypertension similarly warrants caution, as it amplifies cardiovascular risks during therapy.[57]Breastfeeding is relatively contraindicated, as estrone can transfer into breast milk and potentially suppress lactation or affect the infant.In patients with a history of breast cancer, estrone therapy increases the risk of recurrence through estrogen receptor stimulation, making it unsuitable for those with prior estrogen-sensitive disease.[57] Venous thromboembolism risk is elevated 2- to 3-fold with oral estrogentherapy, particularly in the first year of use, due to changes in coagulation factors.[57] Estrone is classified as pregnancy category X by the FDA, indicating it is contraindicated during pregnancy because of potential teratogenic effects on fetal development.As of November 2025, the FDA has removed black box warnings for cardiovascular risks associated with estrogen therapies, recommending individualized risk-benefit assessment rather than broad avoidance in women over 60 for primary prevention, though caution is advised based on patient-specific factors.[65]
Side effects
Estrone administration, like other estrogen therapies, is associated with a range of adverse reactions that vary in frequency and severity. Common side effects, occurring in more than 10% of users, include nausea, breast tenderness, headache, weight gain due to fluid retention, and mood changes such as depression or irritability.[63][66]Serious adverse effects, reported in less than 1% of cases, encompass endometrial hyperplasia when estrone is used without opposing progestin, gallbladder disease requiring surgical intervention, and hypertriglyceridemia that may precipitate pancreatitis.[63][66][67]Long-term use of estrone in combined hormone replacement therapy elevates breast cancer risk, with a relative risk of 1.2 to 1.5 after five or more years of exposure.[68] Recent evidence as of 2025, including studies showing potential protective effects when initiated early post-menopause, has led the FDA to remove the boxed warning for probable dementia; however, risks may persist with late initiation in women aged 65 and older.[69]On November 10, 2025, the FDA initiated removal of black box warnings from menopausal hormone replacement therapy products, including those containing estrone, regarding risks of cardiovascular disease, breast cancer, and probable dementia, based on an updated scientific review emphasizing benefits for symptom relief when used appropriately.[65]Dose-dependent effects include injection site reactions such as pain and, rarely, abscess formation following intramuscular administration of estrone aqueous suspension, as well as withdrawal bleeding upon discontinuation.[5]To mitigate risks, regular monitoring with annual mammograms for breast cancer screening and lipid profile assessments for cardiovascular evaluation is recommended during estrone therapy.[70] Mechanisms underlying venous thromboembolism risks with estrone overlap with those in contraindications for high-risk patients.[63]
History
Discovery and isolation
Estrone, the first estrogen to be isolated in pure form, was discovered through collaborative efforts in reproductive endocrinology during the early 20th century. In 1923, American physiologist Edgar Allen and biochemist Edward Adelbert Doisy identified estrogenic activity in extracts from sow ovaries by demonstrating their ability to induce vaginal cornification—characterized by the appearance of cornified epithelial cells in vaginal smears—in ovariectomized rats; this observation established the foundational Allen-Doisy bioassay for detecting estrogenic substances.[71][72]Building on this, Doisy's team achieved a major breakthrough in 1929 by extracting, purifying, and crystallizing estrone from the follicular fluid of sow ovaries, marking the first isolation of a pure estrogenhormone; they initially named the compound "theelin" due to its role in inducing estrus.[73] Independently in the same year, German biochemist Adolf Butenandt isolated and crystallized estrone from human pregnancy urine, confirming its identity through bioassays and elemental analysis that yielded the molecular formula C₁₈H₂₂O₂.[74][75]The structural elucidation of estrone occurred throughout the 1930s, primarily through Butenandt's degradation studies that reduced the ketone group and identified phenolic characteristics, corroborated by UV spectroscopy revealing absorption bands indicative of an aromatic A-ring in the steroid framework.[76] These efforts established estrone as a phenolicsteroid derivative, distinguishing it from other known hormones. Butenandt's pioneering contributions to the isolation and characterization of sex hormones, including estrone, earned him the 1939Nobel Prize in Chemistry, shared with Leopold Ruzicka for related work on polymethylenes and higher terpenes.[74][77]
Therapeutic development
In the 1930s, estrone was rapidly commercialized as a therapeutic agent, with Parke-Davis introducing it under the brand name Theelin as an injectable preparation derived from pregnant mare urine for alleviating menopausal symptoms.[78] This marked one of the earliest widespread clinical applications of a purified estrogen, building on its isolation from human sources and enabling scalable production for hormone replacement. By the late 1930s, multiple pharmaceutical companies had followed suit, offering estrone injections as a standard treatment for estrogen deficiency states, which spurred further research into its pharmacokinetics and dosing regimens.[79]Estrone became commercially available in the late 1930s following the enactment of the Federal Food, Drug, and Cosmetic Act, which regulated drug approvals.[79]Advancements in synthesis significantly enhanced estrone's availability and purity for therapeutic use. In 1948, Swiss chemists Georg Anner and Karl Miescher achieved the first total synthesis of estrone, providing a non-biological route that confirmed its structure and facilitated production independent of animal sources. Complementing this, semi-synthetic methods emerged in the mid-20th century, converting plant sterols such as diosgenin from yams via microbial fermentation and chemical degradation; these processes, building on steroid synthesis techniques pioneered in the industry, lowered costs and supported industrial-scale manufacturing for clinical formulations.[80]Estrone reached peak clinical utilization from the 1950s through the 1970s, when it dominated estrogen replacement regimens due to its oral and injectable bioavailability, with sales of estrogen products broadly tripling during this period amid growing awareness of menopausal health needs.[81] However, its prominence waned as estradiol-based formulations, such as estradiol valerate and micronized estradiol introduced in the 1960s and 1970s, gained favor for their closer mimicry of endogenous estrogen profiles and improved tolerability.[81]Post-1975, estrone's use declined sharply following reports linking unopposed estrogentherapy to elevated endometrial cancer risk, prompting a 50% drop in overall estrogen prescriptions and a shift toward combined estrogen-progestin therapies as safer alternatives.[81] By the 2000s, pure estrone formulations were largely discontinued in many markets, including the U.S. and Europe, due to these safety concerns and the availability of more potent estradiol options; nonetheless, estrone conjugates like estropipate (piperazine estrone sulfate) have persisted in niche applications for vasomotor symptoms and osteoporosis prevention, remaining FDA-approved and commercially available.[82]As of 2025, estrone has seen a limited revival within bioidentical hormone therapy protocols, where it is semi-synthesized from plant sterols to match human estrone structure, appealing to patients seeking "natural" alternatives for menopausal symptom relief despite lacking superior efficacy evidence over conventional estrogens. In November 2025, the FDA removed the boxed warning on cardiovascular risks for certain menopausal hormone therapies, which may further support the use of estrone formulations.[65] Concurrently, preclinical research explores estrone-conjugated nanoparticles for targeted drug delivery, such as estrone-targeted PEGylated liposomes to enhance cisplatin uptake in estrogen receptor-positive cervical cancers, aiming to improve specificity and reduce systemic toxicity in oncology applications.[83]