979-32-8
Advanced Hormone Dependent Prostate Cancer, Heavy Menstrual Bleeding, Hypogonadism, Menopausal Symptoms, Menopause, Menstrual Irregularities, Postmenopausal Osteoporosis, Primary Amenorrhoea, Secondary Amenorrhea, Hypoestrogenism, Moderate Menopausal Vasomotor Symptoms, Moderate menopausal vulvovaginal atrophy, Severe Vasomotor Symptoms Associated With Menopause, Severe menopausal vulvovaginal atrophy
Estradiol valerate is a synthetic ester derivative of the primary and most potent endogenous human estrogen, 17β-estradiol. Its development as a pharmaceutical agent represents a key advancement in hormone therapy, engineered to overcome the inherent pharmacokinetic limitations of its parent compound. This section provides a definitive identification of estradiol valerate through standardized nomenclature and a comprehensive catalog of its identifiers across major scientific and regulatory databases. It further details the physicochemical properties that are fundamental to its function as a long-acting prodrug. The extensive documentation of this molecule across chemical, pharmaceutical, and metabolomic databases underscores its status as a well-established and extensively researched compound, implying a rich history of clinical use and scientific scrutiny.[1]
The precise identification of a pharmaceutical substance is paramount for research, clinical practice, and regulatory oversight. Estradiol valerate is cataloged under numerous synonyms and unique identifiers, reflecting its global use and study. The primary Chemical Abstracts Service (CAS) number for estradiol valerate is 979-32-8.[1] It is also known by its common abbreviation, E2V.[1]
Its systematic chemical name is (17β)-Estra-1,3,5(10)-triene-3,17-diol 17-pentanoate, which precisely describes the ester linkage of a pentanoyl (valerate) group at the 17-beta position of the estradiol steroid nucleus.[2] Other chemical synonyms include estradiol 17-pentanoate, estradiol valerianate, and 3-Hydroxy-17β-valeroyloxyestra-1,3,5(10)-triene.[2] The comprehensive list of its identifiers, consolidated in Table 1, provides a foundational reference for this agent.
Property | Value | Source(s) |
---|---|---|
Primary Name | Estradiol Valerate | 1 |
DrugBank ID | DB13956 | 1 |
CAS Number | 979-32-8 | 1 |
UNII (FDA) | OKG364O896 | 1 |
Chemical Formula | C23H32O3 | 2 |
Molecular Weight | 356.50 g/mol | 2 |
InChIKey | RSEPBGGWRJCQGY-RBRWEJTLSA-N | 2 |
SMILES | CCCCC(=O)O[C@H]1CC[C@@H]2[C@@]1(CC[C@H]3[C@H]2CCC4=C3C=CC(=C4)O)C | 3 |
Physical Appearance | White to almost-white crystalline powder | 2 |
Melting Point | Approx. 148 °C | 3 |
EC Number | 213-559-2 | 1 |
ChEBI ID | CHEBI:31561 | 1 |
ChEMBL ID | CHEMBL1511 | 1 |
KEGG ID | D01413 | 1 |
Estradiol valerate is a steroid ester with the chemical formula C23H32O3 and a molecular weight of 356.50 g/mol.[2] Its structure is derived from 17β-estradiol, featuring the characteristic four-ring steroid backbone composed of a phenolic A-ring and a cyclopentane D-ring.[2] The defining chemical modification is the attachment of a valerate (pentanoate) ester side chain to the hydroxyl group at the C17β position of the estradiol molecule.[1]
This esterification is not a trivial alteration; it is the central feature that dictates the drug's pharmacological profile. The addition of the five-carbon valerate chain significantly increases the lipophilicity (lipid solubility) of the molecule compared to the parent estradiol.[1] This enhanced lipophilicity is the direct cause of its altered pharmacokinetic properties, enabling its formulation as a long-acting depot injection when dissolved in oil and improving its absorption and stability when administered orally.[1] Physically, it presents as a white to nearly white crystalline powder and has a melting point of approximately 148 °C.[2] This direct link between a specific chemical modification and a profound change in clinical utility exemplifies a rational drug design strategy aimed at optimizing the therapeutic potential of a natural hormone.
The clinical effects of estradiol valerate are entirely dependent on its in-vivo conversion to 17β-estradiol. Its pharmacology is therefore best understood as a two-stage process: the initial pharmacokinetic phase governed by the valerate ester, followed by the pharmacodynamic phase mediated by the released, biologically active estradiol. This section elucidates the complete pharmacological journey of estradiol valerate, from its mechanism of action at the molecular level to its systemic physiological effects and its absorption, distribution, metabolism, and excretion (ADME) profile.
Estradiol valerate itself is a pharmacologically inactive substance; it functions as a prodrug of 17β-estradiol.[1] Following administration by either intramuscular injection or the oral route, the ester bond at the C17β position is rapidly and completely cleaved by non-specific esterase enzymes that are ubiquitously present in the blood plasma, liver, and other tissues.[5] This enzymatic hydrolysis releases two distinct molecules: the active hormone 17β-estradiol and valeric acid, a naturally occurring short-chain fatty acid that is further metabolized through standard pathways.[5]
The 17β-estradiol released from this process is structurally and functionally identical to the endogenous estradiol that is produced primarily by the ovarian follicles in premenopausal women.[1] For this reason, estradiol valerate is considered a source of "bioidentical" estrogen, a key characteristic that distinguishes it from synthetic, non-bioidentical estrogens like ethinylestradiol.[1]
The biological activity is initiated when the released 17β-estradiol binds with high affinity to two principal subtypes of nuclear estrogen receptors: Estrogen Receptor Alpha (ERα) and Estrogen Receptor Beta (ERβ).[1] These receptors are widely distributed throughout the body in varying proportions, with target tissues including the uterus, ovaries, breasts, bone, brain, liver, skin, prostate, and adipose tissue.[1] Upon ligand binding, the receptor undergoes a conformational change, dimerizes, and translocates to the cell nucleus. There, the activated estrogen-receptor complex functions as a transcription factor, binding to specific DNA sequences known as estrogen response elements (EREs) in the promoter regions of target genes.[5] This binding modulates the rate of gene transcription, leading to the synthesis or suppression of specific proteins and initiating the cascade of cellular and physiological responses characteristic of estrogen.[5]
The pharmacodynamic effects of estradiol valerate are the downstream consequences of the receptor-mediated genomic actions of its active metabolite, 17β-estradiol. As the most potent of all mammalian estrogens, estradiol orchestrates a wide array of physiological processes.[1]
Endocrine and Reproductive Effects: Estradiol is the principal hormone responsible for the development and maintenance of the female reproductive system and secondary sexual characteristics.[4] A key pharmacodynamic effect is its modulation of the hypothalamic-pituitary-gonadal axis. Through a negative feedback mechanism on the pituitary gland, circulating estrogens suppress the secretion of gonadotropins—luteinizing hormone (LH) and follicle-stimulating hormone (FSH).[8] This suppression of the LH surge is the primary mechanism for ovulation inhibition, which forms the basis of its use in hormonal contraception.[7] In postmenopausal women, who have naturally elevated levels of LH and FSH due to ovarian failure, estrogen replacement therapy acts to reduce these levels, which is correlated with the alleviation of vasomotor symptoms.[8]
Bone Metabolism: Estrogen receptors are present in bone cells (osteoblasts, osteoclasts, and osteocytes), and estradiol plays a critical role in maintaining skeletal homeostasis. It promotes bone health primarily by inhibiting bone resorption through the suppression of osteoclast activity and by promoting bone formation.[5] The decline in estradiol levels at menopause is a direct cause of accelerated bone loss, leading to osteoporosis; estrogen replacement therapy mitigates this process.
Cardiovascular and Lipid Effects: The effects of estradiol on the cardiovascular system are complex and multifaceted. It generally has a favorable impact on lipid metabolism, characteristically increasing levels of high-density lipoprotein (HDL) cholesterol ("good cholesterol") and decreasing levels of low-density lipoprotein (LDL) cholesterol ("bad cholesterol").[5] It also possesses vasodilatory properties that can improve blood flow.[5] However, a critical and counterbalancing pharmacodynamic effect is its pro-coagulant activity. Estrogens increase the hepatic synthesis of several clotting factors, which increases overall blood coagulability.[1] This effect is central to the increased risk of thromboembolic events associated with estrogen therapy.
Central Nervous System Effects: The presence of ERα and ERβ throughout the brain indicates that estradiol functions as a significant neuromodulator. It is implicated in neuroprotection, the maintenance of cognitive function, and the regulation of mood.[5] Its effects are mediated, in part, by modulating key neurotransmitter systems, including serotonin and dopamine, which may contribute to its effects on mood disturbances associated with menopause.[5]
The pharmacokinetic profile of estradiol valerate is fundamentally determined by its route of administration, a direct consequence of its prodrug design. The esterification strategy was deliberately employed to create two distinct and clinically valuable pharmacokinetic profiles: a sustained-release depot for parenteral use and an orally active form with improved bioavailability compared to native estradiol.[1]
Absorption:
Distribution:
Once in the bloodstream (either via absorption from an IM depot or the GI tract), the released estradiol is widely distributed throughout the body, with higher concentrations found in sex hormone target organs.8 It is highly bound to plasma proteins, with approximately 98% of circulating estradiol being bound.6 The primary binding proteins are sex hormone-binding globulin (SHBG), which binds estradiol with high affinity (~38% of the total), and albumin, which binds with lower affinity but higher capacity (~60%).8 Only the small unbound fraction (~2%) is considered biologically active and able to diffuse into tissues to interact with receptors.11 Chronic oral administration of estrogens is known to induce the hepatic synthesis of SHBG, which can, in turn, increase the total circulating concentration of estradiol by increasing the bound fraction.12 This phenomenon contributes to the significant inter-subject variability and greater-than-predicted accumulation of estradiol observed during long-term oral therapy, suggesting that fixed-dose regimens may produce highly variable clinical effects in different individuals.12
Metabolism:
Exogenous estradiol is metabolized in a manner identical to its endogenous counterpart, with the liver being the principal site of transformation.8 The major metabolic pathway involves the reversible oxidation of estradiol to estrone, a less potent estrogen, by the enzyme 17β-hydroxysteroid dehydrogenase. Both estradiol and estrone can be further hydroxylated and subsequently converted to estriol, which is the major urinary metabolite.5
A portion of estrogen metabolism is mediated by the cytochrome P450 enzyme system, specifically the CYP3A4 isoenzyme.[8] This is a point of major clinical significance, as it forms the basis for numerous drug-drug interactions. Furthermore, estrogens undergo extensive enterohepatic recirculation. In the liver, they are conjugated to form water-soluble sulfates and glucuronides, which are secreted into the bile, hydrolyzed back to their active form by gut bacteria, and then reabsorbed into the circulation.[8] This process creates a large circulating reservoir of estrogen conjugates, particularly estrone sulfate, which can be converted back to the more potent estradiol in peripheral tissues.[8]
Excretion:
The metabolites of estradiol, primarily in their conjugated forms as glucuronides and sulfates, are eliminated from the body mainly through renal excretion into the urine.5 A smaller fraction is excreted via the feces.11
The versatile pharmacological profile of estradiol valerate has led to its application across a wide spectrum of clinical contexts, from managing the physiological changes of menopause to serving as a component of modern contraceptives and gender-affirming care. This section details the formally approved therapeutic indications, explores its prevalent and guideline-supported off-label uses, and synthesizes evidence from key clinical trials that substantiate its efficacy and evolving role in medicine.
Regulatory bodies, including the U.S. Food and Drug Administration (FDA), have approved estradiol valerate for several distinct indications, primarily related to hormone replacement and palliative cancer care.
Beyond its formally approved indications, estradiol valerate is widely used in several important clinical areas based on its established pharmacology, clinical experience, and endorsement by professional guidelines. This reflects a common scenario in medicine where clinical practice and expert consensus advance more rapidly than the formal regulatory labeling process.
The efficacy of estradiol valerate for its various indications is supported by a long history of clinical use and numerous clinical trials.
The clinical application of estradiol valerate is highly dependent on its specific formulation and route of administration. The choice between an oral tablet and an intramuscular injection, or between monotherapy and a combination product, represents a fundamental clinical decision dictated by the therapeutic objective. This section provides a practical overview of the available product forms, their brand names, and the standard dosing regimens for major indications.
Estradiol valerate is commercially available in several formulations globally, with notable differences in availability between regions like the United States and Europe.
Dosage of estradiol valerate must be individualized based on the indication, patient response, and risk factors. The principle of using the lowest effective dose for the shortest duration consistent with treatment goals is paramount, especially in the context of menopausal hormone therapy.[27]
Administration of Intramuscular Injection:
The standard method is a deep intramuscular injection into the upper, outer quadrant of the gluteal muscle.27 A dry needle and syringe are recommended to prevent the oil solution from becoming cloudy.27 While IM is the labeled route, subcutaneous (SubQ) injection is an increasingly common off-label alternative, as it can be more comfortable for self-administration and is considered to have equivalent bioavailability for oil-based depots.10
The selection of a specific formulation and route is a strategic decision. For long-term, stable hormone replacement as required in menopause or gender-affirming care, the depot IM injection is often preferred for its convenience (less frequent dosing) and its ability to provide steady hormone levels while avoiding the metabolic burden and daily fluctuations of oral administration.[5] Conversely, for contraception, which demands precise cyclical hormonal control to inhibit ovulation, only a daily oral combination product is appropriate.[1]
Indication (Status) | Formulation | Typical Dosage | Frequency | Key Clinical Notes |
---|---|---|---|---|
Menopausal Symptoms (Approved) | IM Injection | 10 mg to 20 mg | Every 4 weeks | Use lowest effective dose. Re-evaluate need for therapy every 3-6 months. Add a progestin if uterus is intact.23 |
Hypoestrogenism (Approved) | IM Injection | 10 mg to 20 mg | Every 4 weeks | Used for conditions like hypogonadism, castration, or primary ovarian failure.23 |
Prostate Cancer (Palliative) (Approved) | IM Injection | 30 mg or more | Every 1 to 2 weeks | High doses are used to achieve androgen suppression.6 |
Feminizing Hormone Therapy (GAHT) (Off-label) | IM Injection | 5 mg to 20 mg (or higher) | Every 1 to 2 weeks | Dose is titrated based on clinical response and to achieve target serum estradiol and testosterone levels.6 |
Contraception / Heavy Menstrual Bleeding (Approved) | Oral Tablets (with Dienogest) | Varies (multi-phasic) | Daily | Follows a specific 28-day regimen as per product labeling (e.g., Natazia).1 |
The safety profile of estradiol valerate, like all estrogen therapies, is complex and has been the subject of intense scrutiny and debate for decades. While effective, its use is associated with a range of adverse effects and serious risks that necessitate careful patient selection, ongoing monitoring, and a thorough understanding of its contraindications. The drug's safety label is dominated by a class-wide boxed warning that has profoundly influenced prescribing patterns and patient perception, yet the applicability of this warning to all formulations and populations remains a critical area of clinical discussion.
Adverse reactions to estradiol valerate therapy can range from common, manageable side effects to rare but life-threatening events.
All estrogen-containing products for systemic use in the U.S. carry a stringent boxed warning, the FDA's most serious safety alert. This warning has profoundly shaped the clinical use of hormone therapy, but its origins and applicability warrant a nuanced analysis.
Origin and Content of the Boxed Warning:
The boxed warning was implemented in the early 2000s following the publication of initial results from the Women's Health Initiative (WHI), a large-scale set of clinical trials.31 It is crucial to recognize that the WHI did not study estradiol valerate. The trials investigated oral conjugated equine estrogens (CEE, brand name Premarin) alone or in combination with medroxyprogesterone acetate (MPA, brand name Provera) in a population of postmenopausal women with a mean age of 63 years.15 Despite these specific parameters, the FDA applied the resulting warnings as a class-wide label to all systemic estrogen products.
The key points of the warning are [29]:
Clinical Nuance and Controversy:
The "ghost of the WHI" continues to dominate the safety discussion around estrogen therapy. A significant evidence-practice gap has emerged, as clinicians and researchers question the direct applicability of the WHI findings to all clinical scenarios. The risks observed in an older population using oral, non-bioidentical hormones (CEE/MPA) may not accurately reflect the risk profile of bioidentical hormones like estradiol valerate, particularly when administered via non-oral routes (like IM injection) or in different patient populations (e.g., younger, recently menopausal women or transgender individuals).32 Professional societies, such as the North American Menopause Society, have formally petitioned the FDA to revise the boxed warning, arguing that it overstates the risks for many women and creates an unnecessary barrier to safe and effective treatment, especially for low-dose vaginal therapies.32 This creates a clinical dilemma where prescribers must reconcile a stark regulatory warning with a more nuanced understanding of the evidence as it applies to their individual patient.
The use of estradiol valerate is strictly contraindicated in individuals with certain pre-existing conditions due to an unacceptably high risk of serious harm.
Absolute Contraindications:
Precautions:
Caution is warranted when prescribing for patients with conditions that may be exacerbated by estrogen therapy, including:
Drug interactions with estradiol valerate are primarily pharmacokinetic, arising from interference with its metabolism. The central mechanism involves the modulation of the cytochrome P450 3A4 (CYP3A4) enzyme, which is responsible for a significant portion of estradiol metabolism.[8] Understanding this pathway provides a practical framework for anticipating and managing the most critical interactions.
Interaction Type | Interacting Drug/Substance Examples | Clinical Consequence | Source(s) |
---|---|---|---|
CYP3A4 Inducers | Rifampin, Carbamazepine, Phenobarbital, Phenytoin, St. John's Wort | Increased metabolism of estradiol, leading to decreased plasma concentrations. This can result in a loss of therapeutic efficacy (e.g., return of menopausal symptoms) and/or changes in uterine bleeding patterns (breakthrough bleeding). | 8 |
CYP3A4 Inhibitors | Ketoconazole, Itraconazole, Clarithromycin, Erythromycin, Ritonavir, Grapefruit juice | Decreased metabolism of estradiol, leading to increased plasma concentrations. This can elevate the risk of estrogen-related adverse effects. | 8 |
In addition to CYP3A4-mediated interactions, estrogens may diminish the therapeutic effect of antidiabetic agents by altering glucose tolerance.[9] Therefore, closer monitoring of blood glucose may be necessary in diabetic patients initiating estrogen therapy.
The trajectory of estradiol valerate from its chemical synthesis to its current place in the global pharmacopeia reflects key trends in pharmaceutical development and regulatory science over the past 70 years. Its history is marked by its early establishment as a long-acting estrogen and a more recent divergence in its primary applications between major markets like the United States and Europe.
The development of estradiol valerate was part of a broader effort in the mid-20th century to create more clinically useful forms of natural steroid hormones.
The regulatory pathways for estradiol valerate have differed notably between the United States and Europe, leading to distinct primary markets for the drug.
This divergence in regulatory and market focus is significant. While the U.S. market has maintained a strong clinical niche for the long-acting injectable formulation, primarily for hormone replacement in menopause and gender-affirming care, the major European innovation has been to leverage estradiol valerate's bioidentical nature to create a new generation of oral contraceptives with a potentially improved safety profile.
Estradiol valerate stands as a durable and pharmacologically significant agent in the field of endocrinology. Its design as a prodrug of 17β-estradiol was a seminal example of rational chemical modification to overcome the therapeutic limitations of a natural hormone, successfully enabling both a long-acting depot injection and a viable oral formulation. This versatility has allowed it to be adapted for a wide range of clinical applications, from its traditional role in managing menopausal symptoms and hypoestrogenism to its modern use in oral contraception and as a cornerstone of gender-affirming hormone therapy.
The clinical profile of estradiol valerate is defined by its efficacy in restoring physiological estrogen levels. However, its use is inextricably linked to a complex safety profile dominated by the class-wide risks of thromboembolic events and hormone-sensitive cancers. The ongoing debate surrounding the applicability of the WHI-derived boxed warnings to this bioidentical estrogen—especially when used in different populations and via different routes than those studied—highlights a critical tension between broad regulatory mandates and the need for nuanced, individualized risk assessment in clinical practice.
Recent clinical evidence, particularly studies demonstrating a less pronounced pro-coagulant effect compared to ethinylestradiol, reinforces the therapeutic rationale for using bioidentical estrogens like estradiol valerate. This trend suggests a continued evolution in its application, particularly in areas like contraception, where minimizing risk is paramount. In conclusion, estradiol valerate is not merely a historical hormone therapy but a dynamic therapeutic tool whose full potential and precise risk-benefit profile continue to be refined by ongoing clinical research and evolving standards of care. Its enduring presence in the pharmacopeia is a testament to its fundamental utility and the successful pharmacological strategy behind its creation.
Published at: September 2, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.