Medroxyprogesterone acetate (MPA), commonly known as medroxyprogesterone, is a synthetic progestin hormone derived from progesterone that mimics its biological effects in the body. It is widely used in various medical contexts, including contraception, treatment of menstrual disorders, and hormone replacement therapy, and is available in oral tablet and injectable formulations.[1][2]Medroxyprogesterone is indicated for the treatment of secondary amenorrhea (absence of menstrual periods in women who previously menstruated) and abnormal uterine bleeding due to hormonal imbalances, typically administered orally at doses of 5 to 10 mg daily for 5 to 10 days. In postmenopausal women receiving estrogen therapy, it prevents endometrial hyperplasia—a precancerous condition—by opposing estrogen's effects on the uterine lining, with dosing of 5 to 10 mg daily for 12 to 14 days per month. The injectable form, such as Depo-Provera (150 mg/mL intramuscular) or depo-subQ provera 104 (104 mg/0.65 mL subcutaneous), provides long-acting contraception by preventing ovulation and is administered every 12 to 13 weeks. Additionally, it is used palliatively for advanced endometrial carcinoma and endometriosis-associated pain.[3][2][4][1]The mechanism of action involves binding to progesterone receptors, which inhibits the secretion of gonadotropins from the pituitary gland, suppresses follicular maturation and ovulation, thickens cervical mucus to impede sperm migration, and induces endometrial changes that support its therapeutic effects. While effective, medroxyprogesterone carries notable risks, including menstrual irregularities, weight gain, bone mineral density loss (particularly with prolonged injectable use, potentially exceeding 2 years), meningioma (particularly with prolonged injectable use), and increased chances of thromboembolic events, breast cancer, and cardiovascular disorders, necessitating careful patient monitoring and contraindication in cases of known hypersensitivity, active thromboembolic disease, or certain cancers.[1][4][3][5]
Medical uses
Contraception
Medroxyprogesterone acetate, commonly administered as depot medroxyprogesterone acetate (DMPA), is widely used as an injectable contraceptive to prevent pregnancy. The intramuscular formulation (DMPA-IM) is typically given in a 150 mg dose every 12 to 13 weeks, while the subcutaneous version (DMPA-SC) uses 104 mg at the same interval, providing effective contraception for up to three months per injection.[6][7] DMPA is classified as a long-acting reversible contraceptive (LARC) method due to its extended duration and high efficacy, with a Pearl Index of 0.2 to 0.3 pregnancies per 100 woman-years under perfect use conditions.[8] With typical use, accounting for occasional delays in reinjection, the failure rate is approximately 4%.[8][9] According to the 2024 U.S. Medical Eligibility Criteria for Contraceptive Use (MEC), DMPA use is now cautioned in individuals with increased thromboembolic risk and revised for those with chronic kidney disease.[10]The primary mechanism of action for DMPA in contraception involves suppression of ovulation through inhibition of gonadotropin secretion from the pituitary gland, preventing follicular maturation and the mid-cycle luteinizing hormone surge.[11] Additionally, it thickens cervical mucus to impede sperm penetration and thins the endometrium, reducing the likelihood of implantation if ovulation occurs.[12][13] These multifaceted effects contribute to its reliability as a progestin-only method, suitable for individuals unable to use estrogen-containing contraceptives. Recent studies (as of 2025) have associated long-term DMPA use (≥1 year) with an increased risk of meningioma (relative risk approximately 2.4), though absolute risk remains low.[14]Although less common for routine contraception, medroxyprogesterone acetate is incorporated into some oral contraceptive combinations with estrogens, where it serves primarily to regulate the menstrual cycle and provide progestogenic support for endometrial stability.[15]According to guidelines from the World Health Organization (WHO) and the Centers for Disease Control and Prevention (CDC), DMPA initiation can occur at any time in the menstrual cycle if pregnancy is ruled out, with immediate protection if started within the first 5 days of menses or 7 days postpartum for non-breastfeeding individuals.[16] For continuation, repeat injections should be administered no later than 13 weeks after the prior dose to maintain efficacy, with counseling on potential bleeding irregularities emphasized before starting.[6] Self-administration of DMPA-SC is endorsed by both organizations as a safe and effective option to improve access and continuation rates.[17]
Hormone therapy
Medroxyprogesterone acetate (MPA) is commonly used in menopausal hormone therapy (MHT) for postmenopausal women with an intact uterus, where it is combined with estrogen to oppose estrogen-induced endometrial proliferation and prevent hyperplasia. In continuous combined regimens, typical oral doses range from 2.5 mg to 10 mg daily, providing adequate endometrial protection while minimizing breakthrough bleeding; for example, studies have shown no cases of hyperplasia across these doses when paired with 1.25 mg estrone sulfate over two years.[18] In sequential regimens, 5 mg or 10 mg is administered daily for 12 to 14 consecutive days per month alongside 0.625 mg conjugated estrogens.[19]MPA also treats secondary amenorrhea and abnormal uterine bleeding attributable to hormonal imbalances, such as ovulatory dysfunction, in premenopausal women without underlying organic pathology. For these indications, the standard oral dose is 5 mg to 10 mg daily for 5 to 10 days, often starting on the 16th or 21st day of the cycle if estrogen priming is needed to optimize endometrial response; withdrawal bleeding typically follows within 3 to 7 days of discontinuation.[1][20]As a synthetic progestin, MPA binds to progesterone receptors in the endometrium, inducing secretory transformation that counters estrogen-driven growth and thereby protects against hyperplasia in estrogen-exposed tissues.[1] This progestational activity allows uterine protection in MHT without promoting full proliferative changes seen with unopposed estrogen. The Women's Health Initiative trial, involving over 16,000 postmenopausal women, evaluated daily conjugated equine estrogens (0.625 mg) plus MPA (2.5 mg) versus placebo over 5.2 years and reported increased risks of coronary heart disease (hazard ratio 1.29), invasive breast cancer (1.26), stroke (1.41), and pulmonary embolism (2.13), offset by reductions in colorectal cancer (0.63) and hip fractures (0.66); overall, health risks exceeded benefits, leading to early trial cessation.[21]Off-label, MPA is utilized in transgender women's hormone therapy to suppress endogenous testosterone and mitigate masculinizing effects, often at doses of 5 to 10 mg daily alongside estrogens; retrospective data indicate significant testosterone reduction with minimal impact on estradiol levels and few adverse effects over long-term use.[22]
Cancer treatment
Medroxyprogesterone acetate (MPA) is employed in high doses, typically 400 to 1000 mg intramuscularly per week, for the palliative treatment of advanced or recurrent endometrial carcinoma, where it promotes tumor regression in hormone-responsive cases through agonism of the progesterone receptor.[23] This dosing regimen aims to reduce tumor burden and alleviate symptoms in patients unsuitable for surgery or more aggressive therapies, with initial administration followed by maintenance at 400 mg monthly if response is observed.[23] The U.S. Food and Drug Administration (FDA) approved MPA for advanced endometrial carcinoma in 1971, recognizing its role in hormone-sensitive malignancies.[24]In endometrial cancer, MPA serves as a second-line hormonal therapy option per National Comprehensive Cancer Network (NCCN) guidelines, particularly for progesterone receptor-positive tumors, with oral doses ranging from 200 to 600 mg daily.[25] Response rates to progestins like MPA in advanced endometrial cancer are approximately 15% to 30%, rising to 75% in progesterone receptor-positive cases, enabling disease stabilization and improved quality of life.[26]MPA also plays a palliative role in progesterone receptor-positive advanced breast cancer in postmenopausal women, where it is administered at 500 mg intramuscularly daily for 28 days followed by twice weekly, achieving response rates of 20% to 30% in selected cases.[23][27] The primary mechanism involves direct antiproliferative effects on tumor cells via progesterone receptor binding, alongside indirect actions such as inhibition of gonadotropin release (luteinizing hormone and follicle-stimulating hormone), which reduces estrogen production, and blockade of adrenocorticotropic hormone to lower glucocorticoid levels.[28] These effects counteract estrogen-driven proliferation in hormone-sensitive tumors.[26]In some regimens for endometrial cancer, MPA is combined with tamoxifen (e.g., 20 mg twice daily alternating with MPA), potentially enhancing response rates in resistant cases, though this approach requires vigilant monitoring for thromboembolic risks due to the additive pro-thrombotic effects of both agents.[29] High-dose MPA use in cancer therapy is associated with increased thromboembolic events, necessitating careful patient selection and risk assessment.[30]
Other indications
Medroxyprogesterone acetate (MPA) has been investigated for appetite stimulation in patients experiencing cachexia associated with AIDS or cancer. In clinical trials, high-dose oral MPA at 800 mg/day has demonstrated improvements in appetite and body weight, with some patients achieving 10–20% weight gain over several weeks, primarily through increased caloric intake and fat mass, though lean body mass gains are limited. For instance, a double-blind, placebo-controlled trial in patients with advanced cancer cachexia reported significant appetite enhancement and modest weight increases with MPA compared to placebo, attributing effects to reduced cytokine production involved in anorexia. Similar benefits were observed in small studies of AIDS-related cachexia, where MPA supplementation alongside nutritional support improved weight stabilization and quality of life metrics.MPA is used in the management of paraphilias, particularly in cases of deviant sexual behaviors, by suppressing testosterone levels and reducing libido. Low-dose oral regimens, such as 100–200 mg/day, combined with psychotherapy, have shown effectiveness in controlling impulses in long-term case series of sex offenders, with testosterone reductions correlating to behavioral improvements. In men undergoing androgen deprivation therapy for prostate cancer, MPA effectively alleviates hot flashes, a common vasomotor symptom. Randomized trials indicate that MPA at 20 mg twice daily reduces hot flash frequency and severity by up to 80% in this population, outperforming alternatives like venlafaxine in some comparisons, with benefits attributed to its progestogenic modulation of thermoregulatory centers.Due to the expression of progesterone receptors in many meningiomas, MPA has been explored historically as a potential hormonotherapeutic agent, but recent studies (2024–2025) have shown that prolonged use increases the risk of meningioma development, limiting its application.[5]In veterinary medicine, MPA is employed off-label for canine and feline contraception, suppressing estrus cycles through progestational effects, typically via subcutaneous injections every 3–6 months at doses scaled to body weight (e.g., 50–100 mg for medium-sized dogs), though risks like pyometra limit routine use.Investigational applications have included symptom relief in Alzheimer's disease, such as managing inappropriate sexual behaviors in dementia patients via low-dose MPA (e.g., 150 mg intramuscularly), with ongoing case reports supporting off-label use alongside behavioral interventions, though without benefits to core disease progression.Dosing for these indications typically ranges from 10–400 mg/day orally, adjusted based on response and monitoring for side effects, with evidence derived from small randomized controlled trials emphasizing individualized titration to balance efficacy and risks like thromboembolism.
Adverse effects
Common adverse effects
Medroxyprogesterone, particularly in its depot form (DMPA), commonly causes menstrual irregularities such as spotting, breakthrough bleeding, or amenorrhea, affecting 20–50% of users during the first year of therapy, with rates decreasing over time and typically resolving upon discontinuation.[31][1] These changes occur due to the suppression of ovulation and endometrial effects of the progestin.[32]Weight gain is another frequent adverse effect, with users experiencing an average increase of 2–5 kg over the first year, attributed to increased appetite and fluid retention. Clinical trials, including those evaluating DMPA, have reported mean gains around 2.5 kg after one year, though individual variation exists.[33][11]Mood changes, such as depression and irritability, are reported in 5–10% of users, often linked to the hormonal alterations induced by medroxyprogesterone.[34] These effects are generally mild and reversible but can impact quality of life during treatment.Breast tenderness is a common complaint, occurring due to the progestogenic influence on breast tissue, while acne may arise from the mild androgenic activity of medroxyprogesterone, affecting 1–5% of users.[35][1]For the injectable depot formulation (DMPA), injection site reactions including pain and subcutaneous atrophy occur in approximately 10% of administrations, based on clinical trial data and post-marketing surveillance.[36][37] Overall frequency data for these common effects derive from large-scale clinical trials and ongoing pharmacovigilance reports, confirming their prevalence at standard therapeutic doses.[1][38]
Serious adverse effects
Medroxyprogesterone acetate (MPA), particularly when used in combined estrogen-progestin therapy for more than five years, has been associated with an increased risk of invasive breast cancer, with a relative risk (RR) of 1.24 observed in the Women's Health Initiative (WHI) study, corresponding to an absolute risk of 41 versus 33 cases per 10,000 women-years compared to placebo.[2] This elevated risk underscores the need for careful risk-benefit assessment in long-term users.Thromboembolic events, including deep vein thrombosis (DVT) and pulmonary embolism (PE), represent serious risks with medroxyprogesterone use, especially in high-dose formulations or combined regimens; injectable depot medroxyprogesterone acetate (DMPA) is linked to a 3.6-fold increased risk of venous thrombosis (95% CI, 1.8-7.1), while estrogen plus progestin therapy approximately doubles the risk.[39][40]Long-term use of DMPA can lead to significant bone mineral density (BMD) loss, with reductions of up to 5.7% at the lumbar spine and hip after two years of use; this loss is largely reversible following discontinuation, though recovery may take several years.[41] The U.S. Food and Drug Administration (FDA) has issued a black box warning for DMPA regarding the potential for substantial BMD loss, particularly in adolescents and young adults, where the impact on peak bone mass acquisition remains a concern.[42]In postmenopausal women, medroxyprogesterone as part of combined hormone therapy elevates cardiovascular risks, including stroke (hazard ratio [HR] 1.32; 95% CI, 1.12-1.56) and nonfatal myocardial infarction (HR 1.28).[43][44]Some observational studies have suggested an increased risk of HIV acquisition with DMPA use, with estimates up to 40% higher incidence compared to non-users, though a 2025 Cochrane systematic review found little to no difference (RR 1.02; 95% CI, 0.82-1.26 compared to copper intrauterine device). The World Health Organization recommends DMPA in HIV-endemic areas with appropriate counseling on potential risks.[45]Exposure to medroxyprogesterone during pregnancy carries a potential risk of fetal masculinization, primarily evidenced by animal studies showing virilization of female fetuses at high doses; human data are limited, but progestins with androgenic properties warrant caution.[46]For long-term DMPA users, particularly those with additional osteoporosis risk factors, monitoring with dual-energy X-ray absorptiometry (DEXA) scans may be considered on a case-by-case basis, though routine BMD screening is not recommended by major guidelines.[47]
Contraindications and interactions
Contraindications
Medroxyprogesterone is contraindicated in patients with known or suspected pregnancy, as it poses significant risks to the fetus, including potential teratogenic effects such as hypospadias, clitoral enlargement, and labial fusion.[2] It is also absolutely contraindicated in individuals with current or past breast cancer or other hormone-sensitive malignancies, due to the potential for hormonal stimulation of tumor growth.[48][10] Additional absolute contraindications include undiagnosed abnormal vaginal bleeding, which requires evaluation to rule out underlying pathology; significant liver disease, such as decompensated cirrhosis or active viral hepatitis; and active or history of thromboembolic disorders, including deep veinthrombosis, pulmonary embolism, or cerebrovascular disease, as per FDA labeling for both oral and injectable forms.[2][48][10] Medroxyprogesterone is contraindicated in patients with current or history of meningioma per international guidelines (e.g., EMA 2024); emerging evidence from studies as of 2025 associates long-term use, particularly injectable forms, with increased risk of meningioma, though this is not yet reflected in US FDA labeling.[49]Relative contraindications, where risks may outweigh benefits depending on individual assessment, encompass conditions such as migraine with aura, complicated diabetes (e.g., with nephropathy, retinopathy, or neuropathy), and current breast cancer in remission for less than 5 years.[10]Smoking is a risk factor; per 2024 U.S. Medical Eligibility Criteria, medroxyprogesterone is contraindicated (category 4) in women aged ≥35 years who smoke ≥15 cigarettes per day due to high cardiovascular risk, and a relative risk (category 3) for those smoking <15 cigarettes per day.[10]In pediatric patients, medroxyprogesterone use, particularly in adolescents, requires caution due to potential impacts on bone mineral density; per current guidelines, it is acceptable (category 1) for postmenarcheal adolescents, with counseling on risks and discussion of alternatives.[32] Updated guidelines from the American College of Obstetricians and Gynecologists, aligning with the 2024 U.S. Medical Eligibility Criteria, emphasize avoidance of medroxyprogesterone in patients with hormone-sensitive cancers to prevent recurrence risks.[10]
Drug interactions
Medroxyprogesterone acetate (MPA) is primarily metabolized by the cytochrome P450 3A4 (CYP3A4) enzyme in the liver, making it susceptible to interactions with drugs that induce or inhibit this pathway, which can alter its plasma concentrations and therapeutic efficacy.[2] Concomitant use with strong or moderate CYP3A4 inducers, such as rifampin, carbamazepine, phenytoin, phenobarbital, or St. John's wort, can significantly reduce MPA exposure by accelerating its metabolism and clearance.[50] Pharmacokinetic studies of depot MPA formulations have shown that efavirenz alone increases clearance by approximately 25%, while efavirenz combined with rifampin and isoniazid increases it by over 50%, leading to AUC reductions of 20–50% and potential subtherapeutic levels, particularly in contraception or hormone therapy.[51] In such cases, dose adjustments, alternative contraceptives, or additional non-hormonal methods are recommended to maintain efficacy.[32]Anticonvulsants like phenytoin, a potent CYP3A4 inducer, decrease MPA concentrations and can compromise its contraceptive effectiveness by enhancing hepatic metabolism, necessitating backup contraception or alternative therapies.[50] Similarly, aminoglutethimide, used in cancer treatment, significantly lowers serum MPA levels through induction of hepatic enzymes, potentially reducing its progestogenic effects and requiring an alternative birth control method or monitoring for diminished therapeutic response.[52]CYP3A4 inhibitors, such as ketoconazole, ritonavir, or clarithromycin, may increase MPA concentrations by slowing metabolism, heightening the risk of adverse effects, and should generally be avoided during coadministration.[50] There are no major food interactions with MPA, though oral administration with meals can modestly increase bioavailability, raising AUC by 18–33% without clinical significance.[53]Grapefruit juice, a mild CYP3A4 inhibitor, may slightly elevate MPA exposure in oral formulations, but this effect is generally not clinically relevant.[54]In high-dose MPA regimens for cancer therapy, concomitant use of interacting agents warrants close monitoring of clinical response and, where feasible, therapeutic drug monitoring of MPA plasma levels to ensure adequate exposure and adjust dosing as needed.[55]
Pharmacology
Pharmacodynamics
Medroxyprogesterone acetate (MPA) acts primarily as a high-affinity agonist at the progesterone receptors (PR-A and PR-B), mimicking the effects of endogenous progesterone to regulate gene transcription in target tissues. In vitro assays demonstrate that MPA binds to PR with a Ki of approximately 0.34 nM, exhibiting potent transcriptional activation with an EC50 of around 0.12 nM in reporter gene systems.[56] This agonism leads to downstream effects such as endometrial transformation and inhibition of gonadotropin secretion, though efficacy can vary by cell type due to differences in PR isoform ratios and co-regulators.[57]MPA also displays moderate affinity for the glucocorticoid receptor (GR), with a relative binding affinity of 62% compared to dexamethasone and an IC50 of 5.58 nM in whole-cell binding assays. This interaction results in partial to full GR agonism, promoting anti-inflammatory effects through transactivation (EC50 ≈ 42 nM for glucocorticoid response element-driven genes) and transrepression (EC50 ≈ 0.09 nM for AP-1 inhibition).[58] In contrast, MPA shows weak androgenic activity via binding to the androgen receptor (AR) with a Ki of approximately 19 nM, contributing to limited anti-androgenic or pro-androgenic outcomes depending on context.[59]MPA exhibits weak antiestrogenic activity through low-affinity binding to the estrogen receptor (ER), with negligible overall estrogenic potency and no significant antimineralocorticoid effects. By suppressing hypothalamic GnRH pulsatility and pituitary LH/FSH release, MPA induces a hypoestrogenic state, preventing ovulation without direct estrogenic interference.[1]Tissue-specific effects of MPA highlight differential PR signaling: in uterine tissue, it promotes secretory changes and opposes estrogen-induced proliferation, whereas in breast tissue, it can exhibit mitogenic activity at equivalent doses, underscoring context-dependent receptor interactions.[60]
Pharmacokinetics
Medroxyprogesterone acetate (MPA) is rapidly absorbed after oral administration, with peak plasma concentrations achieved within 1 to 3 hours post-dose.[53] The oral bioavailability of MPA is approximately 90%, allowing for effective systemic exposure following tablet ingestion.[13] For the intramuscular (IM) depot formulation, absorption is slow and sustained, providing release over approximately 3 months, with steady-state plasma concentrations typically ranging from 1 to 2 ng/mL.[61]MPA exhibits high protein binding of about 90%, primarily to albumin, with negligible binding to sex hormone-binding globulin.[53] The apparent volume of distribution (Vd/f) is approximately 40,000 to 80,000 L, indicating extensive tissue distribution.[53]Metabolism occurs primarily in the liver via cytochrome P450 3A4 (CYP3A4), producing inactive metabolites through hydroxylation and conjugation.[62] The elimination half-life is approximately 12 to 17 hours for oral MPA and about 50 days for the IM depot formulation.[53]Excretion is mainly renal (about 50% as metabolites, primarily glucuronides) and fecal (about 40% via biliary secretion), with no significant accumulation observed in patients with renal impairment.[63] Clearance is higher in obese patients, leading to reduced plasma exposure compared to normal-weight individuals, though no dosage adjustments are typically required.[64]
Chemistry
Chemical structure
Medroxyprogesterone has the molecular formula \ce{C22H32O3}.[65] Its acetate ester, medroxyprogesterone acetate (MPA), possesses the molecular formula \ce{C24H34O4}.[66]Medroxyprogesterone features a pregnane steroid skeleton derived from 17α-hydroxyprogesterone, with a characteristic 6α-methyl substitution on the pregn-4-ene-3,20-dione core.[65] This structure relates closely to progesterone (pregn-4-ene-3,20-dione), from which it differs by the addition of a methyl group at the 6α position and a hydroxy group at 17α; the 6α-methyl modification sterically hinders enzymatic metabolism, enhancing oral bioavailability and progestogenic potency compared to the parent hormone.[13]The IUPAC name is (6S,8R,9S,10R,13S,14S,17R)-17-acetyl-17-hydroxy-6,10,13-trimethyl-2,6,7,8,9,11,12,14,15,16-decahydro-1H-cyclopentaphenanthren-3-one, equivalently expressed in steroid nomenclature as 17α-hydroxy-6α-methylpregn-4-ene-3,20-dione.[65] The molecule exhibits defined stereochemistry at multiple chiral centers, including 6α for the methyl group and 17α for the hydroxy substituent, with the 17β orientation of the acetyl side chain essential for effective binding to the progesterone receptor.[67]Key structural elements include a tetracyclic cyclopentaphenanthrene ring system with a double bond between C4 and C5, a ketonefunctional group at C3, another ketone at C20 (as part of the acetyl moiety), and a tertiary α-hydroxy group at C17, all contributing to its progestin-like properties.[65]
Physical properties
Medroxyprogesterone appears as a white to off-white, odorless, crystalline powder, with a molecular weight of 344.5 g/mol. It has a melting point of 214.5 °C and a computed logP of 3.5, indicating lipophilicity. Medroxyprogesterone is insoluble in water but freely soluble in chloroform, slightly soluble in ether and methanol, and soluble in acetone and dioxane (1 in 50 and 1 in 60 at 25 °C, respectively).[65]Medroxyprogesterone acetate (MPA) appears as a white to off-white, odorless, crystalline powder, which facilitates its handling in pharmaceutical manufacturing. Its molecular formula is \ce{C24H34O4}, with a molecular weight of 386.5 g/mol.[66]MPA exhibits a melting point in the range of 205–209 °C, contributing to its thermal stability during processing. The compound's lipophilicity is reflected in a logP value of approximately 3.9, which supports its suitability for long-acting depot formulations by enabling sustained release in lipid environments.[66][13][68]Solubility characteristics are key to its formulation: MPA is poorly water-soluble at less than 0.01 mg/mL (approximately 0.002 mg/mL), but it is freely soluble in chloroform and sparingly soluble in ethanol (96%). These properties necessitate the use of organic solvents or suspension vehicles in preparations. According to United States Pharmacopeia (USP) standards, MPA is practically insoluble in water, freely soluble in chloroform, soluble in acetone and dioxane, and sparingly soluble in alcohol and methanol.[66][69][13]Stability considerations include sensitivity to light, requiring protection during storage, and susceptibility to hydrolysis in aqueous solutions, which can lead to degradation products. USP analytical standards specify assays for purity (not less than 97.0% and not more than 103.0% of labeled amount), limits on related substances, and identification tests via infrared spectroscopy and chromatography to ensure quality.[70][66]In injectable formulations, such as depot suspensions, MPA is micronized to particle sizes typically in the range of 2–50 μm to optimize bioavailability and control the release rate, with smaller particles promoting faster dissolution and larger ones extending duration. This process enhances uniformity and predictability in long-acting intramuscular preparations.[71]
History and society
Development and approval
Medroxyprogesterone acetate (MPA) was synthesized in 1958 by researchers at the Upjohn Company as a synthetic progestin analog designed to mimic the effects of progesterone with enhanced potency and oral bioavailability.[72] The compound was developed through modifications to the progesterone structure, focusing on adding a 6α-methyl group to improve metabolic stability and reduce androgenic side effects compared to earlier progestins.[13]Initial clinical trials for MPA began in the late 1950s and early 1960s, primarily evaluating its efficacy in treating gynecological conditions such as secondary amenorrhea and dysfunctional uterine bleeding, with exploratory studies on contraception emerging by the mid-1960s.[72] The oral formulation, marketed as Provera, received FDA approval on June 18, 1959, for the treatment of secondary amenorrhea and abnormal uterine bleeding due to hormonal imbalance.[13] Early trials demonstrated MPA's ability to induce endometrial withdrawal bleeding and suppress ovulation, establishing its role in hormone replacement therapy.[73]The injectable depot formulation, depot medroxyprogesterone acetate (DMPA or Depo-Provera), faced prolonged regulatory scrutiny before approval for contraception. Upjohn submitted an application for contraceptive use in 1967, but the FDA repeatedly delayed approval due to concerns over carcinogenicity observed in animal studies, particularly mammary tumors in beagle dogs exposed to high doses.[74] These studies, conducted in the 1970s, showed increased tumor incidence in beagles, leading to an FDA advisory committee recommendation against approval for contraception in 1978. In 1981, the U.S. restricted federal funding for its use in international family planning programs, despite endorsements from the World Health Organization (WHO) that year for its efficacy in family planning programs in developing countries.[75] After additional safety reviews and long-term human data accumulation, the FDA approved DMPA for contraception on October 29, 1992, at a dose of 150 mg every three months, concluding that the benefits outweighed risks when used as directed. In 2005, the FDA approved a subcutaneous formulation, depo-subQ Provera 104, offering an alternative injection method for contraception.[4]During the 1970s, key clinical trials further validated MPA's therapeutic applications, including randomized studies demonstrating its efficacy in reducing pain and lesion size in endometriosis through high-dose oral regimens that induced pseudopregnancy states.[76] These trials, involving hundreds of patients, reported symptom relief in up to 80% of cases with minimal short-term adverse effects, paving the way for its off-label use in this condition before formal indications were expanded.[77]In the 2000s, post-approval studies addressed emerging safety concerns, particularly regarding bone mineral density (BMD). Prospective cohort trials, such as a 2006 study of adolescent users, found significant BMD losses at the hip and spine after 24 months of DMPA use, averaging 5-7% reduction, prompting FDA updates to include a black box warning in 2004 about potential long-term bone effects and recommendations for calcium supplementation.[78] Follow-up research confirmed partial BMD recovery upon discontinuation, informing guidelines to limit use in adolescents and monitor at-risk populations.[47]MPA's original U.S. patents, including key composition claims filed in the late 1950s, expired in the mid-1970s, enabling the entry of generic manufacturers and reducing costs for oral formulations by the late 1970s.[79] More recent regulatory reviews include the European Medicines Agency's (EMA) 2024 Pharmacovigilance Risk Assessment Committee (PRAC) evaluation, which examined post-marketing data on cardiovascular risks such as venous thromboembolism associated with MPA-containing products, leading to updated prescribing information emphasizing risk factors like age and smoking.[80]
Generic and brand names
Medroxyprogesterone is the International Nonproprietary Name (INN) for the active substance, while medroxyprogesterone acetate is the ester form most commonly used in pharmaceutical preparations.[81][66]Common brand names include Provera for oral tablets, Depo-Provera for intramuscular injectable suspension, and Depo-subQ Provera 104 for subcutaneous injection.[82][83][84] Internationally, variations include Alti-MPA and Provera in Canada, and Farlutal in Italy.[3][85]Medroxyprogesterone is available exclusively by prescription in most countries worldwide and is not sold over-the-counter in any jurisdiction.[3][86]Generic versions have been available in the United States and European Union since the late 1970s, following the expiration of early patents on the original formulations, with costs typically ranging from $10 to $50 per dose depending on the form and location.[87][88]Depot medroxyprogesterone acetate has been included on the World Health Organization's Model List of Essential Medicines for contraceptive use since 1977.[89]