Hydroxyprogesterone caproate
Hydroxyprogesterone caproate, also known as 17α-hydroxyprogesterone caproate (17-OHPC), is a synthetic progestogen ester derived from progesterone, featuring a caproate chain that enables prolonged release following intramuscular injection.[1][2] Its chemical structure is pregn-4-ene-3,20-dione with a 17-(1-oxohexyl)oxy substituent, conferring a molecular formula of C27H40O4 and extended half-life suitable for weekly dosing.[3] Marketed under brand names such as Makena, it was FDA-approved in 2011 under accelerated approval to reduce the risk of recurrent preterm birth before 37 weeks in women with singleton pregnancies and a history of spontaneous preterm delivery, based on a single pivotal trial showing a relative risk reduction of 34%.[4][5] However, the confirmatory PROLONG trial involving over 3,000 participants failed to demonstrate efficacy over placebo, prompting the FDA to withdraw approval in April 2023, as benefits did not outweigh potential risks and no clinical advantage was substantiated.[6][7] This decision highlighted controversies surrounding accelerated approvals reliant on surrogate endpoints without robust confirmatory data, though earlier observational and smaller studies had suggested benefits in select high-risk populations.[8][9] Beyond preterm birth prevention, hydroxyprogesterone caproate has been employed for managing endometrial hyperplasia, uterine cancers, and amenorrhea due to its progestational effects mimicking endogenous progesterone.[10][11]Chemical and Pharmacological Profile
Structure and Synthesis
Hydroxyprogesterone caproate (OHPC), also known as 17α-hydroxyprogesterone hexanoate, is a synthetic progestogen derived from the steroid hormone progesterone. It features a pregnane skeleton with a characteristic Δ⁴-3-keto configuration, a 17α-acetyl group, and an ester linkage at the 17α-position to hexanoic acid, conferring prolonged duration of action due to slow hydrolysis. The molecular formula is C₂₇H₄₀O₄, with a molecular weight of 428.60 g/mol. [12] The IUPAC name is (8R,9S,10R,13S,14S,17R)-17-(1-oxohexyl)oxy-10,13-dimethyl-3-oxo-2,6,7,8,9,11,12,14,15,16-decahydro-1H-cyclopentaphenanthrene-17-yl acetate, though it is commonly referred to by its trivial name reflecting the caproate (hexanoate) ester of 17α-hydroxyprogesterone. This structure distinguishes OHPC from natural progesterone by the addition of the 17α-hydroxy group and the lipophilic caproate chain, which enhances its solubility in oils for intramuscular administration and extends its pharmacokinetic profile.[13] [2] OHPC is synthesized through esterification of 17α-hydroxyprogesterone with hexanoic acid or its activated derivatives, such as caproic anhydride or caproyl chloride, typically in the presence of a base like pyridine to facilitate the reaction at the 17α-hydroxyl position. 17α-Hydroxyprogesterone, the key starting material, is obtained via semi-synthesis from plant sterols like diosgenin or through microbial transformation of progesterone. Industrial processes optimize yields through stepwise acylation, purification by crystallization, and control of by-products to achieve high purity for pharmaceutical use, with reported total yields up to 95% in refined methods.[14] [15]Pharmacodynamics
Hydroxyprogesterone caproate (OHPC), also known as 17α-hydroxyprogesterone caproate, is a synthetic progestin that functions as an agonist at the progesterone receptor (PR), exhibiting approximately 26% affinity for PR-A and 30% affinity for PR-B relative to progesterone itself.[16] This binding mediates progestogenic effects, including the promotion of endometrial secretory transformation and maintenance of pregnancy through uterine quiescence.[2] Unlike some synthetic progestins, OHPC demonstrates minimal affinity for androgen, estrogen, or glucocorticoid receptors, though it retains weak glucocorticoid receptor-mediated activity comparable to progesterone.[17] The primary physiological actions of OHPC mimic those of endogenous progesterone, such as inhibiting gonadotropin-releasing hormone (GnRH) pulsatility, which suppresses ovulation, and reducing myometrial contractility to sustain pregnancy.[18] In experimental models of preterm labor, OHPC has been observed to counteract inflammatory stimuli, including tumor necrosis factor-alpha (TNF-α) and thrombin, by inhibiting fetal membrane weakening and reducing production of matrix-degrading enzymes.[19] It also correlates with elevated anti-inflammatory interleukin-10 (IL-10) levels in response to lipopolysaccharide (LPS) challenge in at-risk pregnancies.[20] Despite these observed effects, the exact mechanism by which OHPC reduces recurrent preterm birth risk—its primary clinical indication—remains undetermined, with progestational receptor agonism presumed central but not definitively proven as the sole pathway.[4] OHPC lacks significant mineralocorticoid or other steroid receptor interactions that could confound its profile, distinguishing it from earlier progestins with broader hormonal cross-reactivity.[2]Pharmacokinetics
Hydroxyprogesterone caproate (17-OHPC) is administered via intramuscular injection as a depot formulation in castor oil, resulting in slow absorption and prolonged release. [21] Peak plasma concentrations (Cmax) occur between 1 and 7 days post-injection, with times varying by population: approximately 1-2 days in pregnant women with multifetal gestations and 3-7 days in non-pregnant women. [22] [21] For a single 1,000 mg dose in non-pregnant women, Cmax averages 27.8 ng/mL. [21] In pregnant women receiving weekly 250 mg doses, steady-state trough levels range from 7.5 to 10 ng/mL across gestation, with Cmax around 12-13 ng/mL. [23] The drug distributes widely, binding extensively to plasma proteins including albumin and corticosteroid-binding globulin. [21] Volume of distribution is influenced by maternal body weight. [24] Metabolism occurs primarily via cytochrome P450 enzymes CYP3A4 and CYP3A5, involving phase I reactions (hydroxylation, reduction) and phase II conjugation (sulfation, glucuronidation). [21] The caproate ester is retained during initial metabolism, yielding mono- and di-hydroxylated metabolites with lower plasma levels than the parent compound. [23] Elimination follows first-order kinetics with an apparent terminal half-life of 7.8 to 16 days, varying by study and population; for instance, 10 days in multifetal pregnancies and 16.3 days in singleton gestations. [21] [22] [23] Clearance increases with lean body mass and is affected by body mass index. [25] Approximately 30% is excreted in urine and 50% in feces, primarily as conjugated metabolites and free steroids. [21] Pharmacokinetic parameters exhibit high inter-individual variability, influenced by gestational age, body composition, and route of administration. [26]Clinical Applications
Prevention of Recurrent Preterm Birth
Hydroxyprogesterone caproate (OHPC), administered as weekly intramuscular injections of 250 mg starting between 16 and 20 weeks of gestation and continuing until 36 weeks 6 days or delivery, was indicated for reducing the risk of recurrent preterm birth prior to 35 weeks in women with a singleton gestation and a history of at least one prior spontaneous preterm singleton birth. This regimen derived from the 2003 Meis et al. randomized controlled trial, which enrolled 463 women and reported a relative risk reduction of 34% for preterm birth before 37 weeks (36.3% in OHPC group vs. 54.9% in placebo; risk ratio 0.66, 95% CI 0.52-0.83), though no significant differences were observed in perinatal mortality or composite neonatal morbidity.[27] The U.S. Food and Drug Administration (FDA) granted accelerated approval for Makena (OHPC) in February 2011 based primarily on this trial and earlier small studies suggesting efficacy. Subsequent large-scale confirmatory trials, however, failed to replicate these findings. The OPPTIMUM trial (2016), involving 1,229 women, found no reduction in preterm birth before 34 weeks (relative risk 0.93, 95% CI 0.72-1.20) or in a composite of adverse neonatal outcomes (relative risk 0.97, 95% CI 0.83-1.14). Similarly, the STOPPRETERM trial (2019) with 1,310 participants showed no benefit for preterm birth less than 34 weeks (11.0% vs. 11.3%; hazard ratio 0.93, 95% CI 0.72-1.20) or neonatal outcomes. The PROLONG trial (2020), a double-blind study of 1,707 women, also reported no significant difference in preterm birth before 35 weeks (27.6% vs. 28.6%; hazard ratio 0.97, 95% CI 0.84-1.11) or composite neonatal morbidity. Meta-analyses incorporating these trials, such as the 2021 EPPPIC review of 70 studies, indicated that intramuscular OHPC did not consistently reduce preterm birth rates or improve perinatal outcomes, unlike vaginal progesterone which showed benefits in high-risk groups.00217-8/fulltext)[28] In response to the lack of confirmatory evidence from these trials, the FDA withdrew approval for OHPC for preterm birth prevention effective April 5, 2023, concluding that it did not demonstrate effectiveness in reducing recurrent preterm birth or associated neonatal morbidity in any subgroup.00243-0/fulltext) The American College of Obstetricians and Gynecologists (ACOG) updated its guidance in April 2023 to no longer recommend OHPC for this indication, citing insufficient efficacy data, and instead endorses vaginal progesterone (90-200 mg daily from 16-20 weeks to 36 weeks) for women with a prior spontaneous preterm birth.[29] The Society for Maternal-Fetal Medicine (SMFM) similarly stated that OHPC should not be used routinely, emphasizing alternatives like vaginal progesterone or cervical cerclage based on cervical length screening.[30] Despite the withdrawal, compounded versions of OHPC remain available in some settings, though their use lacks FDA oversight and supporting large-trial data.[31] No reductions in long-term outcomes like infant mortality or neurodevelopmental issues have been causally linked to OHPC in randomized data.[32]Gynecological and Endocrine Disorders
Hydroxyprogesterone caproate has been indicated for the management of primary and secondary amenorrhea, where it is administered intramuscularly to induce menstrual bleeding in cases attributable to hormonal deficiency without underlying organic pathology.[33][34] Early clinical reports from the 1950s documented its use in secondary amenorrhea therapy, with doses typically ranging from 250 to 500 mg every 1 to 2 weeks, leading to resumption of cyclic bleeding in responsive patients.[35] In dysfunctional uterine bleeding due to hormonal imbalance, hydroxyprogesterone caproate serves to stabilize the endometrium by mimicking progesterone's effects, thereby reducing excessive bleeding; it is contraindicated in the presence of organic causes such as submucous fibroids or uterine cancer.[36][37] Dosing often involves 250 to 500 mg intramuscularly weekly or biweekly until bleeding control is achieved, with withdrawal bleeding expected 7 to 10 days post-administration in approximately 92% of cases in hormone replacement contexts.[38] For advanced endometrial carcinoma, hydroxyprogesterone caproate has been used as hormonal therapy, particularly in estrogen receptor-positive tumors, with intramuscular doses up to 7 g per week demonstrating objective tumor responses in 20 to 30% of patients in mid-20th-century studies, though without curative potential and with infrequent adverse effects.[39][40][41] Its progestogenic action opposes estrogen-driven proliferation, but modern progestins like medroxyprogesterone acetate have largely supplanted it due to superior bioavailability and evidence from randomized trials.[42] In endocrine contexts, such as corpus luteum insufficiency contributing to anovulatory cycles, it provides supplemental progestational support, though contemporary guidelines favor oral or vaginal progesterone for luteal phase defects.[1]Contraceptive Uses
Hydroxyprogesterone caproate (OHPC) was the first progestogen employed as a long-acting injectable contraceptive, with initial clinical evaluation occurring in 1963. In a preliminary study involving 25 women, monthly intramuscular injections of 500 mg OHPC effectively induced reversible sterility by suppressing ovulation and altering menstrual patterns, with no pregnancies reported during the treatment period.35893-9/pdf)[43] This progestin-only approach demonstrated contraceptive potential through sustained progestogenic activity, but it often resulted in amenorrhea or irregular bleeding, prompting further development of combination formulations for improved acceptability.[44] Subsequently, OHPC has been incorporated into combined injectable contraceptives, typically paired with an estradiol ester to enhance endometrial stabilization and cycle regularity while maintaining high contraceptive efficacy. One such formulation, estradiol valerate/hydroxyprogesterone caproate (EV/OHPC), contains 5 mg estradiol valerate and 250 mg OHPC and is administered monthly via intramuscular injection. Marketed as Injectable No. 1 in China since the 1970s, this regimen inhibits ovulation through combined estrogen-progestin action and has been associated with low pregnancy rates in large-scale use, though specific Pearl Indices vary by population and adherence, generally aligning with other monthly injectables at under 1 failure per 100 woman-years under typical use.[45] Similar combinations, such as estradiol cypionate/OHPC, were tested in the early 1970s for once-monthly contraception, providing durations of protection exceeding 30 days due to the slow release of OHPC. Despite early promise, progestin-only OHPC injectables saw limited long-term adoption as standalone contraceptives due to suboptimal bleeding control and the emergence of more potent alternatives like medroxyprogesterone acetate. Combined formulations persist in select regions, but global use remains niche, with efficacy dependent on precise timing of injections to overlap steroid levels for ovulation suppression. No major contemporary trials have reaffirmed OHPC's standalone contraceptive role, reflecting a shift toward higher-potency progestins in modern injectables.[46]Other Medical Indications
Hydroxyprogesterone caproate has been utilized in the palliative management of advanced endometrial carcinoma, particularly prior to the widespread adoption of modern chemotherapeutic regimens. Clinical evaluations from the 1970s documented its efficacy in inducing objective tumor responses, including regression or stabilization, in subsets of patients with disseminated disease. For instance, in a cohort treated with intramuscular doses escalating to 7.0 g weekly, substantial evidence supported its role as an adjunctive antineoplastic agent, with responses observed in approximately 20-30% of cases depending on tumor differentiation and prior therapies.27:3<485::AID-CNCR2820270302>3.0.CO;2-1)[39] Dosing regimens typically involved high cumulative amounts, such as 25-83 g over a treatment course, administered via intramuscular injection to leverage the drug's prolonged progestogenic activity. Well-differentiated tumors (G1) exhibited higher response rates, with complete or partial remissions reported in up to 40% of such cases, whereas moderately differentiated (G2) lesions showed more variable outcomes. The therapy was noted for its favorable safety profile at these doses, with minimal severe toxicities beyond occasional injection-site reactions or mild endocrine effects.[47]27:3<485::AID-CNCR2820270302>3.0.CO;2-1) Under the brand Delalutin, hydroxyprogesterone caproate received indications for treating advanced endometrial adenocarcinoma in non-pregnant women, reflecting its historical application in gynecologic oncology before progestins like medroxyprogesterone acetate became predominant. Early trials, including one involving 27 patients, reported objective improvements in 8 cases, underscoring its potential to extend survival in hormone-responsive malignancies. However, contemporary guidelines prioritize more potent progestogens or combination therapies, limiting its current off-label or legacy use.[48][41][42]Safety Profile
Contraindications and Precautions
Hydroxyprogesterone caproate is contraindicated in individuals with a current or prior history of thrombosis or thromboembolic disorders, as progestins may exacerbate such conditions.[49] It is also contraindicated in cases of known or suspected breast cancer, other hormone-sensitive malignancies, or a history thereof, due to the potential for hormone-dependent tumor stimulation.[4] Additional contraindications include undiagnosed abnormal vaginal bleeding, which requires evaluation to exclude underlying pathology; cholestatic jaundice of pregnancy or prior jaundice associated with progestin therapy; active hepatic disease or dysfunction; and hypersensitivity to hydroxyprogesterone caproate, castor oil, or any formulation excipients.[49][4] Precautions are advised in patients with risk factors for thromboembolism, including those with personal or family history of venous thromboembolism, obesity, smoking, or immobility, necessitating close monitoring for signs such as leg pain, swelling, or shortness of breath, with immediate discontinuation if suspected.[49] Hepatic function should be assessed prior to initiation and monitored periodically, particularly in those with mild impairment, as progestins can worsen liver conditions.[50] Patients experiencing depression or with a history thereof warrant vigilance, as progestin therapy may aggravate mood disorders.[4] Fluid retention may occur, potentially exacerbating conditions like epilepsy, migraine, asthma, or cardiac/renal dysfunction, requiring symptom surveillance.[50] Allergic reactions, including anaphylaxis, have been reported, mandating discontinuation upon onset of rash, urticaria, or respiratory distress.[4] Use during breastfeeding is not recommended due to potential transfer into milk and unknown effects on infants.[49] Injection site reactions, such as pain, swelling, or erythema, are common and should be managed with rotation of sites to minimize local complications.[51] Safety and efficacy have not been established in individuals under 16 years of age.[49]Adverse Effects and Risks
Common adverse effects of hydroxyprogesterone caproate (OHPC) administration primarily involve injection site reactions, occurring in a significant proportion of treated women. In the pivotal Meis trial, injection site pain was reported in 34.8% of OHPC recipients compared to 32.7% in the placebo group, with swelling affecting 17.1% versus 7.8%, and urticaria in 12.3% versus 11.1%.[49] Additional reactions included pruritus (7.7% versus 5.9%) and bruising or soreness in smaller subsets, with overall rates of local reactions higher due to the intramuscular route.[52] Serious maternal adverse events are infrequent but documented in clinical data. A safety review of trials identified four serious adverse events in the OHPC arm of one study, including pulmonary embolism, injection site cellulitis, and a maternal death, though causality was not definitively established.[53] Progestin exposure, including OHPC, carries theoretical risks of thromboembolism, consistent with class effects of synthetic progestogens, though randomized trial data showed no significant elevation over placebo for such events.[49] Observational data have also linked OHPC to increased gestational diabetes risk, with a meta-analysis reporting elevated odds in exposed pregnancies.[54] Fetal and neonatal risks include potential congenital anomalies and short-term toxicities. Clinical trials reported rare instances of infant congenital anomalies in OHPC-exposed groups, but rates did not differ significantly from placebo.[53] Animal studies and limited human data suggest endocrine-disrupting potential, with possible impacts on fetal development, though confirmatory human evidence remains limited.[55] Long-term offspring outcomes raise concerns from observational cohorts. A Danish registry study found in utero OHPC exposure associated with doubled risk of any cancer (odds ratio 2.08, 95% CI 1.27-3.40), particularly colorectal cancer (OR 4.78), with first-trimester exposure linked to adjusted hazard ratio 2.57 (95% CI 1.59-4.15), escalating with injection number.[55] These findings, derived from linkage of medical birth and cancer registries spanning 1998-2015, suggest possible carcinogenic effects but are prone to confounding by indication (e.g., maternal preterm history) and unmeasured variables; no randomized data confirm causality.[56] Multigenerational reproductive toxicology studies in rodents demonstrated no clear adverse fertility or teratogenic effects at clinical doses, supporting relative safety in preclinical models.[57]| Adverse Effect Category | Incidence in OHPC Group | Incidence in Placebo Group | Source |
|---|---|---|---|
| Injection Site Pain | 34.8% | 32.7% | FDA Label[49] |
| Injection Site Swelling | 17.1% | 7.8% | FDA Label[49] |
| Urticaria | 12.3% | 11.1% | FDA Label[49] |
| Pruritus | 7.7% | 5.9% | FDA Label[49] |
Overdose Management
No cases of overdose leading to adverse events have been documented in clinical trials of hydroxyprogesterone caproate injection.[3][58] In the event of overdosage, no specific antidote is available, and management consists of symptomatic and supportive care.[3][4] Patients should be monitored for vital signs, potential hypersensitivity reactions, or exacerbation of progestogenic effects, with interventions directed at alleviating presenting symptoms such as nausea or local injection site issues if they occur.[59] Immediate consultation with a poison control center or emergency medical services is advised to guide individualized care.[60] Given the intramuscular depot formulation, elimination is gradual, precluding rapid detoxification methods like dialysis.[58]Drug Interactions
No in vivo drug-drug interaction studies have been conducted with hydroxyprogesterone caproate.[49] In vitro assessments demonstrate that hydroxyprogesterone caproate has minimal potential to induce or inhibit CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, or CYP3A4 at clinically relevant concentrations.[49][4] Hydroxyprogesterone caproate is primarily metabolized via oxidation by CYP3A4 and, to a lesser extent, CYP3A5, with the caproate moiety retained during biotransformation.[2][61] Consequently, coadministration with strong CYP3A4 inducers (e.g., rifampin, carbamazepine) could theoretically accelerate metabolism and lower systemic exposure, potentially diminishing therapeutic effects, while strong inhibitors (e.g., ketoconazole, ritonavir) might increase exposure and risk of adverse effects; however, the clinical relevance of such interactions remains unestablished due to the absence of confirmatory human data.[62][2] Progestogens like hydroxyprogesterone caproate may antagonize the prolactin-suppressing effects of bromocriptine by inducing amenorrhea without addressing underlying hyperprolactinemia, and concurrent use is generally not recommended.[61] Additionally, as progestogens can influence carbohydrate metabolism, monitoring may be warranted with antidiabetic agents such as canagliflozin, though specific data for hydroxyprogesterone caproate are limited.[61] No significant interactions with alcohol or common foods have been identified.[49]Regulatory and Efficacy Controversies
FDA Approval Process and Initial Evidence
Hydroxyprogesterone caproate, a synthetic progestogen, received initial U.S. Food and Drug Administration (FDA) approval in 1956 under New Drug Application (NDA) 10-347 for various obstetric indications, including the treatment of threatened and habitual abortion in pregnant women, though evidence for efficacy in these uses was limited and based on observational data rather than randomized controlled trials.[63] By the early 2000s, the drug was commonly used in compounded form for preventing recurrent preterm birth, prompted by preliminary studies suggesting progestogen therapy might mitigate this risk in high-risk pregnancies.[63] The pivotal evidence supporting expanded approval for preterm birth prevention came from the Meis trial, a multicenter, randomized, double-blind, placebo-controlled study published in 2003, which enrolled 463 women with a singleton pregnancy and a prior spontaneous preterm birth before 37 weeks' gestation.[27] Participants received weekly intramuscular injections of 250 mg 17 alpha-hydroxyprogesterone caproate (17-OHPC) or placebo from 16 to 20 weeks' gestation until 36 weeks or delivery. The primary outcome was preterm birth before 37 weeks, occurring in 36.3% of the 17-OHPC group versus 54.9% in the placebo group, yielding a relative risk of 0.66 (95% CI, 0.52 to 0.83; P<0.001); secondary outcomes showed similar reductions before 35 weeks (20.9% vs. 30.7%) and 32 weeks (11.7% vs. 25.3%).[27] This trial built on smaller earlier studies, including a 1960s report of 17-OHPC reducing preterm birth in women with prior losses, but the Meis results provided the first robust randomized evidence, despite limitations such as reliance on historical rather than concurrent controls in some analyses and potential baseline imbalances.[27] In 2011, the FDA granted accelerated approval to Makena (17-OHPC injection, 250 mg/mL) under NDA 021945 specifically to reduce the risk of preterm birth prior to 37 weeks in women with a singleton pregnancy and a history of singleton spontaneous preterm birth, based primarily on the Meis trial as the single adequate and well-controlled study demonstrating clinical benefit.[63] This approval pathway was invoked due to the unmet medical need for interventions in recurrent preterm birth, a leading cause of neonatal morbidity, with the condition that the sponsor complete confirmatory post-approval trials to verify clinical benefit.[63] Prior to this, 17-OHPC had been available only as a custom-compounded product, which the approval aimed to standardize and ensure quality control for.[63] The decision prioritized access to the drug amid supportive observational data from registries, though critics noted the Meis trial's surrogate-like endpoint of preterm birth incidence without direct long-term neonatal outcome improvements and questioned generalizability given exclusion of multifetal gestations.[27]Confirmatory Trials and Efficacy Debates
The PROLONG trial (NCT01004029), mandated by the FDA as a confirmatory study following the 2011 accelerated approval of 17α-hydroxyprogesterone caproate (17-OHPC) for reducing recurrent preterm birth risk, enrolled 1,708 women with singleton pregnancies and a prior spontaneous preterm singleton delivery before 37 weeks gestation from 2010 to 2015 across 35 sites, primarily international. Participants, randomized 1:1 to weekly 250 mg intramuscular 17-OHPC or placebo from 160/7 to 206/7 weeks gestation until 366/7 weeks or delivery, showed no significant difference in the primary endpoint of preterm birth before 35 weeks: 11.0% (93/845) in the 17-OHPC group versus 11.6% (98/843) in placebo (adjusted hazard ratio 0.90, 95% CI 0.68-1.19). Preterm birth before 37 weeks occurred in 27.6% versus 28.8% (adjusted hazard ratio 0.95, 95% CI 0.81-1.12), with no improvements in composite neonatal outcomes like respiratory distress or intraventricular hemorrhage. Fetal or early infant death rates were similar (2.3% vs 2.4%).[64][65] Meta-analyses integrating PROLONG with prior trials have reinforced the lack of efficacy. A 2021 review of six randomized controlled trials (n=2,573 singleton gestations with prior preterm birth) reported no reduction in preterm birth before 37 weeks (relative risk 0.95, 95% CI 0.90-1.00), before 34 weeks (RR 0.96, 95% CI 0.82-1.12), perinatal death (RR 0.87, 95% CI 0.50-1.52), or respiratory distress syndrome (RR 1.00, 95% CI 0.85-1.18). Earlier meta-analyses, such as a 2019 pooled analysis excluding PROLONG, suggested modest benefits for delivery before 37 weeks (RR 0.67, 95% CI 0.48-0.94), but inclusion of confirmatory data shifted estimates toward null effects, highlighting reliance on smaller, potentially underpowered studies.[28][66] Debates over 17-OHPC efficacy stem from the original Meis trial's (2003) reported 34% reduction in preterm birth before 37 weeks (RR 0.66, 95% CI 0.52-0.83; n=463), contrasted by PROLONG's failure to replicate amid larger sample size and stricter controls. Reanalyses of Meis data indicate possible false positives from baseline imbalances (e.g., higher prior preterm severity in placebo) and unadjusted subgroups, with sensitivity analyses yielding nonsignificant results (RR 0.79, 95% CI 0.54-1.15 after exclusions). Proponents cited observational registries or post-hoc PROLONG subgroups (e.g., prior birth <28 weeks, HR 0.58, 95% CI 0.35-0.97), but these were exploratory, not prespecified, and lacked replication; FDA reviews deemed them insufficient for efficacy claims. The 2020 FDA advisory committee voted 13-3 against substantial evidence of effectiveness, prioritizing randomized trial data over mechanistic hypotheses or access concerns.[67][68][5] Empirical discrepancies underscore causal challenges: while progestogens may stabilize cervical function or reduce inflammation in select cases, population-level RCTs like PROLONG demonstrate no net benefit, aligning with null findings in related trials (e.g., OPPTIMUM for broader progestin use). Regulatory actions, culminating in FDA's 2023 withdrawal of Makena approval, reflect this evidence hierarchy, though compounded formulations persist amid debates over off-label utility despite absent verification.[68][69]Withdrawal of Approval and Post-Marketing Developments
In 2023, the U.S. Food and Drug Administration (FDA) withdrew approval for Makena (hydroxyprogesterone caproate injection, or 17-OHPC) and its generic equivalents, which had been granted accelerated approval in 2011 for reducing the risk of recurrent preterm birth before 35 weeks in women with a singleton pregnancy and prior spontaneous preterm birth.[6] The decision followed review of the confirmatory PROLONG trial (NCT01004029), a multicenter, randomized, double-blind, placebo-controlled study involving 1,130 women, which failed to demonstrate efficacy, showing no significant reduction in preterm birth rates (11.0% vs. 11.6% placebo for delivery <35 weeks; hazard ratio 0.93, 95% CI 0.67-1.30). An FDA advisory committee had voted 9-7 in October 2022 to recommend withdrawal, citing insufficient evidence of clinical benefit under accelerated approval regulations requiring confirmatory data.[32] The withdrawal, announced on April 6, 2023, and effective immediately, applied to all intramuscular and subcutaneous formulations of 17-OHPC referencing Makena, rendering them unapproved for the preterm birth indication.[70] The FDA emphasized that the action was based solely on lack of demonstrated effectiveness, not safety concerns, as post-marketing surveillance had not identified new risks beyond known injection-site reactions and potential progesterone-related effects.[71] Manufacturer AMAG Pharmaceuticals (later Viatris) discontinued marketing Makena by March 2023 but advocated for restricted use in high-risk subgroups, though the FDA rejected this narrower approval due to inadequate subgroup data from PROLONG.[72] Post-withdrawal, 17-OHPC remains unavailable as an FDA-approved product for preterm birth prevention in the U.S., but compounded versions—prepared by state-licensed pharmacies without FDA oversight—continue to be prescribed off-label, raising concerns about variable quality, sterility, and dosing accuracy.[71] Professional societies diverged in response: the Society for Maternal-Fetal Medicine endorsed the withdrawal and recommended against 17-OHPC use, favoring alternatives like intramuscular 17-OHPC discontinuation and vaginal progesterone for cervical shortening; the American College of Obstetricians and Gynecologists expressed regret but deferred to FDA evidence while awaiting further data.[73][74] Usage declined sharply pre-withdrawal, with fills dropping 65% from 2011-2020 amid efficacy doubts, and post-2023 shifts emphasize lifestyle modifications, cervical cerclage, and 17-alpha-hydroxyprogesterone alternatives without caproate ester.[75] Internationally, the European Medicines Agency reviewed 17-OHPC-containing products in 2024, restricting authorizations to non-pregnancy indications like endometriosis or habitual abortion where evidence supports use, while contraindicating for preterm birth prevention due to inconsistent trial outcomes.[76] Ongoing pharmacovigilance highlights no emergent long-term risks from prior exposure, but real-world studies post-withdrawal underscore the need for robust alternatives, as preterm birth rates persist at 10-12% in high-risk U.S. cohorts without effective pharmacotherapy.[65]Historical Context
Discovery and Early Development
17α-Hydroxyprogesterone caproate (OHPC) was synthesized in 1953 by Karl Junkmann at Schering AG through esterification of the naturally occurring steroid 17α-hydroxyprogesterone with caproic (hexanoic) acid.[77] This chemical modification produced a lipophilic prodrug designed for intramuscular administration, leveraging the ester's slow hydrolysis to achieve prolonged progesterone-like activity and extended duration of action relative to unmodified hydroxyprogesterone.[77] The synthesis addressed limitations of earlier short-acting progestins by enabling depot formulations that maintained therapeutic levels over days to weeks, as evidenced by pharmacokinetic properties observed in subsequent studies.[78] Preclinical evaluation began shortly after synthesis, with early animal models focusing on its potential to support pregnancy maintenance. In 1958, Junkmann and colleague G. Suchowsky reported that OHPC effectively sustained gestation in ovariectomized pregnant rabbits, mimicking endogenous progesterone's role in preventing luteolysis and uterine contractility.[79] These findings highlighted OHPC's superior potency and longevity compared to other esters, attributing efficacy to its resistance to rapid hepatic metabolism and sustained release from injection sites.[79] Such studies laid the groundwork for clinical translation, emphasizing causal mechanisms like progesterone receptor agonism without the short half-life of oral or non-esterified alternatives. OHPC entered medical use in the mid-1950s, approved for indications including dysfunctional uterine bleeding, endometrial carcinoma palliation, and threatened abortion.[55] In the United States, it was marketed by E.R. Squibb & Sons as Delalutin, with initial dosing regimens of 250–500 mg intramuscularly for obstetric support based on empirical response rather than large-scale trials.[2] Early adoption reflected optimism for synthetic progestins in reproductive medicine, though outcomes varied due to heterogeneous patient populations and limited randomized data at the time.[55]Key Milestones in Clinical Adoption
Hydroxyprogesterone caproate (OHPC) received initial U.S. Food and Drug Administration (FDA) approval in 1956 under the brand name Delalutin for indications including the treatment of advanced endometrial adenocarcinoma, dysfunctional uterine bleeding, and support in pregnancy such as for habitual abortion or threatened miscarriage.[63] [33] Early clinical adoption focused on its progestogenic effects to maintain pregnancy in high-risk cases, with intramuscular injections administered weekly or as needed based on small observational studies from the 1950s and 1960s reporting anecdotal success in reducing miscarriage rates, though randomized evidence was limited and results mixed.[80] By the late 20th century, OHPC saw off-label use in obstetrics for preterm birth prevention among women with prior spontaneous preterm delivery, informed by meta-analyses of earlier trials suggesting potential benefit, such as a 1980s review indicating reduced recurrence risk.[27] Adoption remained niche due to inconsistent data and availability of alternative progestins, with compounding pharmacies providing customized formulations for select practitioners. The pivotal Meis et al. randomized controlled trial, published in 2003, demonstrated a 34% relative reduction in preterm birth before 37 weeks among 463 high-risk women receiving weekly 250 mg intramuscular OHPC compared to placebo (36.3% vs. 54.9% recurrence rate), prompting broader clinical uptake.[27] [81] This led to guidelines from the American College of Obstetricians and Gynecologists endorsing weekly OHPC starting at 16-20 weeks gestation for women with singleton pregnancies and prior preterm birth history, accelerating adoption from specialized centers to routine obstetric practice by the mid-2000s.[81] FDA accelerated approval of OHPC (as Makena) in February 2011 specifically for reducing recurrent preterm birth risk, based on the Meis trial under Subpart H provisions requiring post-approval confirmatory studies, further driving adoption.[63] [82] Quarterly prescription fills rose from fewer than 11,000 between 2010 and 2014 to over 30,000 by early 2019, reflecting widespread integration into U.S. prenatal care protocols for eligible patients despite ongoing debates over long-term efficacy.[75]Societal and Economic Dimensions
Nomenclature and Formulations
Hydroxyprogesterone caproate is the caproate (hexanoate) ester of the naturally occurring progestogen 17α-hydroxyprogesterone, formed by esterification at the 17α-hydroxyl group with caproic acid to enhance its duration of action via intramuscular depot formulation.[1] Its systematic IUPAC name is [(8R,9S,10R,13S,14S,17R)-17-acetyl-10,13-dimethyl-3-oxo-2,3,6,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H-cyclopentaphenanthren-17-yl] hexanoate.[1] The compound has the molecular formula C₂₇H₄₀O₄, a molecular weight of 428.6 g/mol, and CAS registry number 630-56-8.[1] Common synonyms include 17α-hydroxyprogesterone caproate, 17-OHPC, hydroxyprogesterone hexanoate, and trade names such as Delalutin (historical U.S. brand) and Primolut Depot (used internationally).[1] These names reflect its chemical derivation and pharmaceutical applications, with "caproate" denoting the six-carbon chain ester that imparts lipophilicity for sustained release.[1] The primary pharmaceutical formulation is an oil-based solution for intramuscular injection, containing 250 mg/mL hydroxyprogesterone caproate dissolved in a vehicle of castor oil (28.6% v/v) and benzyl benzoate (46% v/v), with benzyl alcohol (1.5% v/v) as a preservative.[83] It is supplied in 5 mL multiple-dose vials providing 1250 mg total active ingredient, designed for weekly dosing of 250 mg in clinical protocols for preterm birth prevention prior to regulatory changes.[3] This viscous, yellow solution ensures slow absorption from the injection site, achieving prolonged plasma levels over weeks.[83] Alternative routes, such as subcutaneous injection at similar doses (e.g., 275 mg weekly), have been investigated but are not standard formulations.[84] Experimental oral ester formulations have been patented but lack approved commercial availability.[85]Availability and Legal Status
In the United States, the Food and Drug Administration withdrew approval for hydroxyprogesterone caproate (17-OHPC), marketed as Makena and its generics, effective April 5, 2023, following confirmatory trials that failed to verify clinical benefit in reducing preterm birth risk under the accelerated approval pathway.[71] [70] The withdrawal applies to the drug's sole approved indication, rendering it unapproved for any use and prohibiting lawful interstate distribution, though limited pre-existing stockpiles may persist for off-label or investigational purposes.[6] In the European Union, the European Medicines Agency initiated suspension of all hydroxyprogesterone caproate-containing products in May 2024 after a pharmacovigilance review highlighted insufficient efficacy evidence across indications and potential risks, including congenital anomalies observed in animal studies and limited human data.[86] Previously authorized in select member states such as Austria, France, and Italy for obstetric uses, the medicines were required to be withdrawn from supply chains, with national authorities enforcing the measures.[76] Outside North America and the EU, hydroxyprogesterone caproate retains legal availability in various countries, often under brand names like Proluton Depot, for indications including preterm labor prevention and luteal phase support, with regulatory approval varying by jurisdiction. For instance, Pakistan suspended marketing authorizations in July 2025 citing ineffectiveness, aligning with FDA findings, but production and distribution continue in Asia-Pacific markets such as India and China, where market reports project sustained demand through 2030 despite global efficacy debates.[87] [88] In some regions, it is formulated in combination with estradiol valerate as a monthly injectable contraceptive, remaining accessible where not subject to equivalent restrictions.[89]Pricing, Market Dynamics, and Ethical Concerns
Prior to FDA approval in 2011, hydroxyprogesterone caproate was available as a compounded formulation at approximately $10 to $20 per 250 mg dose, equating to about $300 for a full 20-week treatment course.[90] [91] Following approval as the branded product Makena, the price surged to $1,500 per injection, resulting in costs exceeding $30,000 for the typical regimen of weekly doses over 20 weeks.[92] The manufacturer reduced the price by about 55% to $690 per injection (around $15,000 per course) amid public backlash, supplemented by rebates for certain payers.[93] [94] Branded versions remained over 5,200% more expensive than compounded equivalents, contributing to disparities in utilization patterns observed in insurance claims data.[95] Makena benefited from seven years of orphan drug exclusivity under the Orphan Drug Act, which deterred direct generic competition but prompted the FDA to permit compounding pharmacies to supply bioidentical versions, maintaining lower-cost access during that period.[92] [91] Market dynamics shifted after the 2019 PROLONG trial failed to confirm efficacy for preterm birth prevention, leading the FDA to propose withdrawal of approval in 2020 and finalize it in April 2023, citing insufficient evidence of benefit outweighing risks.[71] [96] Approvals for Makena and its generics were revoked, though limited pre-existing inventory persisted briefly; usage transitioned predominantly to compounded formulations, with state Medicaid programs reimbursing over $41 million for all forms in the third quarter of 2020 alone before full implementation of restrictions.[75] [71] As of 2025, the drug is no longer FDA-approved for preterm birth prevention, with supply limited to compounding pharmacies under section 503A, and market projections indicate further price erosion for available doses potentially below $250 amid reduced demand and regulatory scrutiny.[97] [98] The dramatic price escalation for Makena drew ethical criticism for leveraging orphan drug protections on a long-available compound with minimal new investment in innovation, effectively creating a barrier to access for vulnerable pregnant patients despite historical low-cost availability.[91] [99] This approach was argued to unjustifiably inflate healthcare expenditures without commensurate clinical advancements, prioritizing recoupment of confirmatory trial costs over equitable distribution.[100] Post-withdrawal, ethical debates intensified around continued off-label or compounded use, with proponents citing subgroup analyses (e.g., potential benefits in certain racial demographics) dismissed by regulators as unsubstantiated and potentially misleading, given overall trial failures and risks like injection-site complications.[101] Access inequities persisted for low-income and Medicaid-dependent populations, where high pre-withdrawal costs limited uptake even as efficacy evidence waned, raising questions about the moral imperative to promote unproven therapies under the guise of public health equity.[75]Ongoing Research and Future Directions
Current Investigations
Following the 2023 withdrawal of U.S. Food and Drug Administration approval for 17α-hydroxyprogesterone caproate (17-OHPC) for preterm birth prevention due to lack of efficacy demonstrated in the PROLONG trial, research has pivoted toward pharmacokinetic analyses, long-term offspring outcomes, and exploratory uses in non-standard indications.[71][102] A multicenter study (NCT03292731) completed in 2023 examined the correlation between maternal plasma 17-OHPC concentrations and preterm birth rates in high-risk singleton pregnancies, finding variable drug levels but no clear threshold predictive of efficacy, prompting further scrutiny of dosing adequacy in historical trials.[103] Similarly, a 2024 pharmacokinetic investigation reported that steady-state levels after weekly 250 mg intramuscular injections varied widely among women with prior preterm birth, with lower concentrations associated with treatment failure, though causality remains unestablished without randomized confirmation.[104] Safety-focused investigations have intensified, particularly regarding in utero exposure effects. A 2024 preclinical study in rodents exposed to 17-OHPC during gestation identified disruptions in hypothalamic-pituitary-adrenal axis programming and altered stress responses in offspring, raising hypotheses about potential neurobehavioral risks in humans despite the drug's historical use.[105] Epidemiologic data from a 2022 cohort analysis, updated in post-withdrawal reviews, indicated a 2.5-fold increased risk of childhood cancers (primarily acute lymphoblastic leukemia) among offspring exposed to 17-OHPC compared to unexposed controls, though sample sizes limited statistical power for rare events and confounding by maternal preterm risk was not fully adjusted.[55] These findings have spurred calls for longitudinal human follow-up, with no large-scale prospective studies yet initiated as of 2025.[106] Exploratory clinical trials persist in niche applications. An ongoing randomized trial (NCT02989025, last updated July 2025) evaluates adjunctive 17-OHPC in severe preeclampsia diagnosed before 34 weeks, hypothesizing prolongation of gestation via progesterone-mediated vascular stabilization, with primary endpoints of maternal hypertension resolution and perinatal survival; interim data suggest modest delays in delivery but elevated risks of hepatic complications.[107] Another study (NCT04183452) compares intramuscular versus subcutaneous 17-OHPC administration in women with prior preterm birth, aiming to optimize bioavailability and patient adherence post-market withdrawal, reporting equivalent plasma profiles but higher injection-site tolerability with subcutaneous routes.[108] Comparative effectiveness research, including a 2025 randomized trial in threatened miscarriage, found no superiority of intramuscular 17-OHPC over vaginal progesterone in preventing progression to preterm labor, reinforcing shifts toward localized progestins.[109] These efforts underscore unresolved mechanistic questions, such as 17-OHPC's limited anti-inflammatory effects at the cervicovaginal interface compared to alternatives.[110]Alternative Progestins and Comparative Studies
Vaginal micronized progesterone, administered daily at doses of 200 mg for women with a short cervix or 400 mg for those with prior preterm birth, serves as a primary alternative to intramuscular 17α-hydroxyprogesterone caproate (17-OHPC) for preventing recurrent spontaneous preterm birth in singleton gestations.[111] Unlike 17-OHPC, which faced regulatory withdrawal in 2023 following the negative PROLONG trial results showing no superiority over placebo, vaginal progesterone has demonstrated consistent efficacy in multiple randomized controlled trials and meta-analyses, reducing preterm birth risk by approximately 30-50% in high-risk populations without cervical shortening or with a cervical length below 25 mm.[112] [7] Comparative studies have yielded mixed findings on relative efficacy. A 2015 individual patient data meta-analysis of three trials involving women with prior preterm birth found vaginal progesterone superior to 17-OHPC, with a relative risk of 0.58 for preterm birth before 35 weeks versus 0.97 for 17-OHPC.[113] However, subsequent randomized trials, such as a 2021 multicenter study of 339 women, reported similar rates of preterm birth less than 37 weeks (18.5% for vaginal progesterone versus 22.3% for 17-OHPC), though vaginal administration was associated with longer latency to delivery.[114] [115] A 2021 non-inferiority trial further indicated that vaginal progesterone did not achieve predefined non-inferiority margins against 17-OHPC for recurrent preterm birth prevention but showed trends toward improved neonatal outcomes.[116] Oral dydrogesterone, a synthetic progestin, represents another alternative, typically dosed at 10-40 mg daily, but evidence supports its inferiority to 17-OHPC in threatened preterm labor. A 2022 randomized trial of 200 women found 17-OHPC extended pregnancy latency by a mean of 12.5 days compared to 7.2 days for dydrogesterone and improved neonatal respiratory outcomes, with preterm delivery rates of 28% versus 45%.[117] Broader systematic reviews, including the 2021 EPPI-IC collaborative analysis of 70 trials, reinforce vaginal progesterone's edge over intramuscular 17-OHPC for singleton preterm birth prevention, particularly in reducing early preterm delivery (<34 weeks), while noting no clear benefit for 17-OHPC in multifetal gestations and potential risks like increased preterm premature rupture of membranes.[118] These comparisons highlight formulation-specific pharmacokinetics—vaginal progesterone achieves targeted uterine effects with lower systemic exposure—though direct head-to-head trials remain limited, and guidelines increasingly favor vaginal over intramuscular routes post-17-OHPC withdrawal.[119][28]Veterinary and Non-Human Applications
Animal Health Uses
Hydroxyprogesterone caproate, a synthetic progestin, is utilized in veterinary medicine to address reproductive challenges in livestock, including cattle, equines, sheep, and goats. It functions by mimicking progesterone to support luteal phase maintenance, thereby aiding in the prevention of early embryonic mortality and habitual abortion in animals prone to pregnancy loss due to hormonal deficiencies.[120][121] Injections are typically administered intramuscularly, with formulations such as 750 mg doses employed to stabilize hormonal balance and reduce risks associated with conditions like repeat breeding and uterine prolapse.[120] Specific applications include pregnancy support in high-risk cases, where the compound helps sustain corpus luteum function and endometrial receptivity, potentially improving fertility outcomes in herds affected by luteal insufficiency or infertility.[121] In anestrous livestock, such as cattle or camels, regimens involving multiple 750 mg intramuscular injections at 72-hour intervals, often combined with equine chorionic gonadotropin (2000 IU) and prostaglandin F2α (25 mg), have been shown to induce estrus effectively, with subsequent fertility rates evaluated in controlled studies.[122] This approach targets true anestrus by prolonging progestational influence before ovulation induction, though efficacy varies by species and nutritional status.[122] In equines, hydroxyprogesterone caproate has been detected in plasma samples via gas chromatography-tandem mass spectrometry, indicating its off-label or targeted use for reproductive modulation, potentially for similar pregnancy maintenance or estrus control purposes.[123] Veterinary formulations are available through specialized pharmacies, underscoring its role as a progestin option for non-human applications where endogenous progesterone levels are inadequate.[124] Administration requires veterinary oversight to mitigate risks such as injection site reactions or disruptions to natural cyclicity, with dosing tailored to body weight and condition—typically 250–750 mg per treatment cycle in larger species.[120]Comparative Pharmacology in Species
Hydroxyprogesterone caproate (17-OHPC) exhibits species-dependent hepatic metabolism primarily mediated by cytochrome P450 3A (CYP3A) enzymes, with monohydroxylation as the major pathway forming metabolites such as M1, M3, M4, and M5 across tested species. In liver microsomes, the dominant monohydroxylated sub-metabolite varies: M3 predominates in rats, dogs, baboons, and humans; M4 in mice and rabbits; and M1 in pigs, with dogs uniquely producing additional M5. Inhibition studies confirm CYP3A involvement, though IC50 values for ketoconazole differ, ranging from 0.067 µM in rabbits to 8.5 µM in dogs, indicating varying enzyme sensitivity. Overall metabolic profiles in baboons and rats most closely resemble humans, while rabbits and rats show slower in vitro clearance, with microsomal half-lives of 128 minutes and 289 minutes, respectively, compared to 29 minutes in humans.[125]| Species | Dominant Metabolite | Microsomal Half-Life (min) | CYP3A IC50 (µM, ketoconazole) |
|---|---|---|---|
| Human | M3 | 29 | 0.20 |
| Baboon | M3 | 41 | 0.45 |
| Dog | M3 (plus M5) | 31 | 8.5 |
| Pig | M1 | 44 | 0.27 |
| Mouse | M4 | 55 | 0.23 |
| Rabbit | M4 | 128 | 0.067 |
| Rat | M3 | 289 | 1.34 |