C43H67N11O12S2
90779-69-4
Premature Births
Atosiban is a synthetic nonapeptide analogue of the human hormone oxytocin, classified pharmacologically as a competitive oxytocin and vasopressin receptor antagonist.[1] Its principal clinical application is as a tocolytic agent, or labour repressant, administered intravenously for the acute management of preterm labor.[1] By selectively blocking the receptors responsible for initiating uterine contractions, Atosiban induces a state of uterine quiescence, thereby delaying imminent preterm birth in a well-defined patient population.[1] A key distinguishing feature of Atosiban is its highly specific mechanism of action, which translates to a significantly more favorable maternal safety and tolerability profile when compared to older, less specific tocolytic agents such as beta-adrenergic agonists.[2] This combination of targeted efficacy and enhanced safety has established Atosiban as a primary therapeutic option in many regions worldwide.
The development of Atosiban was pioneered by the Swedish pharmaceutical company Ferring Pharmaceuticals, with its first appearance in the scientific literature dating back to 1985.[1] As the originator firm, Ferring AB (Sweden) guided the molecule through clinical development, culminating in its approval and introduction as a novel tocolytic therapy.[5]
Atosiban received its marketing authorization from the European Medicines Agency (EMA) in January 2000, followed by its commercial launch within the European Union in April of the same year.[2] Its adoption expanded rapidly, and by June 2007, the drug was approved for use in 67 countries.[2]
A significant aspect of Atosiban's regulatory history is its conspicuous absence from the markets in the United States and Japan.[2] This was not the result of a regulatory rejection based on safety or efficacy concerns. Rather, it was a strategic commercial decision made by Ferring Pharmaceuticals. The company opted not to pursue licensing in these major markets, reportedly due to the short duration remaining on the drug's patent at the time, which would have limited its market exclusivity and return on investment.[2] This commercial decision has created a notable geopolitical divide in the standard of care for preterm labor. While clinicians in Europe and other approved regions have access to a tocolytic agent specifically designed for uterine relaxation with minimal systemic side effects, their counterparts in the United States must rely on off-label use of agents from other drug classes, such as calcium channel blockers (nifedipine) or magnesium sulfate. This divergence underscores how pharmaceutical market dynamics, patent law, and corporate strategy can directly influence the therapeutic options available to physicians and patients, leading to different clinical practice patterns for the same medical condition in different parts of the world.
To ensure unambiguous identification in clinical, research, and regulatory contexts, Atosiban is referenced by a variety of proprietary, generic, and chemical names, as well as unique database identifiers.
Table 1: Key Drug Identifiers
Identifier Type | Value | Source(s) |
---|---|---|
DrugBank ID | DB09059 | 1 |
CAS Number | 90779-69-4 | 2 |
PubChem CID | 5311010 | 13 |
ATC Code | G02CX01 | 6 |
FDA UNII | 081D12SI0Z | 3 |
KEGG Drug ID | D07475 (Atosiban acetate) | 16 |
InChIKey | VWXRQYYUEIYXCZ-OBIMUBPZSA-N | 9 |
Atosiban is a synthetic, cyclic nonapeptide, classified as a small molecule and an oligopeptide.[1] Its structure is derived from the natural hormone oxytocin through four key modifications designed to convert the native agonistic activity into a potent antagonistic effect.[1] These specific structural alterations are fundamental to its pharmacological profile:
Like oxytocin, the Atosiban peptide contains a disulfide bridge between the residue at position 1 (3-mercaptopropanoic acid) and the cysteine residue at position 6, forming a cyclic structure.[12]
Atosiban is typically supplied as a sterile, white to off-white lyophilized powder for reconstitution into a solution for intravenous administration.[5] Its key physicochemical properties, essential for its formulation, storage, and pharmacokinetic modeling, are summarized in Table 2.
Table 2: Physicochemical Properties of Atosiban
Property | Value | Source(s) |
---|---|---|
Molecular Formula | C43H67N11O12S2 | 7 |
Molecular Weight | 994.19 g/mol | 6 |
Appearance | White to off-white solid/powder | 5 |
Solubility (Water) | Soluble; up to 50-100 mg/mL | 5 |
Solubility (Other) | DMF: 30 mg/mL; DMSO: 14 mg/mL; Ethanol: 5 mg/mL | 9 |
Storage Conditions | Store desiccated at -20°C | 5 |
Melting Point | >165°C (decomposes) | 5 |
pKa (Predicted) | Strongest Acidic: 11.28; Strongest Basic: 9.59 | 17 |
logP (Predicted) | -0.17 (ALOGPS); -4.6 (Chemaxon) | 17 |
Purity (Typical) | ≥98% (by HPLC) | 11 |
The therapeutic effect of Atosiban is derived from its function as a competitive antagonist at neurohypophysial hormone receptors, primarily the oxytocin receptor (OTR) and, to a lesser extent, the vasopressin V1a receptor.
Atosiban is a potent and competitive antagonist of the OTR.[1] During late pregnancy and labor, the density of OTRs in the myometrium (the smooth muscle layer of the uterus) increases dramatically, making this tissue highly sensitive to circulating oxytocin.[5] Atosiban exerts its tocolytic effect by binding to these membrane-bound OTRs, thereby preventing the endogenous hormone oxytocin from activating them.[1]
The oxytocin receptor is a G-protein coupled receptor (GPCR) that primarily signals through the Gq protein pathway. Oxytocin binding normally activates phospholipase C, which catalyzes the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol triphosphate (IP3) and diacylglycerol (DAG). Atosiban's antagonism specifically blocks this oxytocin-stimulated increase in IP3 production.[1] The downstream consequence of inhibiting the
IP3 pathway is twofold: it prevents the release of stored calcium (Ca2+) from the sarcoplasmic reticulum and reduces the influx of extracellular Ca2+ through voltage-gated channels.[1] This comprehensive shutdown of the mechanisms that increase cytosolic
Ca2+ concentration is the ultimate molecular event that prevents the phosphorylation of myosin light chains, leading to uterine muscle relaxation. This results in a marked reduction in both the frequency and force of uterine contractions, inducing a state of uterine quiescence.[1] The onset of this therapeutic action is rapid, with a significant reduction in uterine contractions observed within 10 minutes of administration.[2]
In addition to its effects on the OTR, Atosiban is also a competitive antagonist at the vasopressin V1a receptor.[2] Some in vitro binding assays suggest its affinity may even be higher for the V1a receptor (
Ki = 3.5 nM) than for the OTR (Ki = 81 nM).[9] This dual antagonism is clinically relevant because vasopressin, which is structurally similar to oxytocin, can also stimulate myometrial contractions via V1a receptors. By blocking both receptor types, Atosiban provides a more comprehensive inhibition of the key hormonal pathways that drive uterine contractility during labor.
More recent and sophisticated pharmacological investigations have revealed a deeper complexity in Atosiban's mechanism of action. It does not act as a simple, neutral antagonist but rather as a "biased ligand" at the oxytocin receptor.[1] While it effectively blocks the Gq-coupled signaling pathway responsible for uterine contraction (its therapeutic effect), it simultaneously acts as an
agonist for the Gi-coupled signaling pathway at the very same receptor.[1]
This Gi agonism has been shown to activate distinct downstream signaling cascades, including the mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) pathways.[3] The activation of NF-κB, in particular, is a pivotal step in initiating pro-inflammatory responses. This creates a molecular paradox: Atosiban is concurrently sending a powerful "stop contraction" signal via Gq antagonism and a subtle "start inflammation" signal via Gi agonism. Given that inflammation is a potent and well-established trigger for the onset of labor, this opposing pro-inflammatory action may serve as an intrinsic pharmacological brake on the drug's own tocolytic efficacy. This molecular-level duality could provide a fundamental explanation for Atosiban's clinical profile—why it is highly effective and safe, yet does not demonstrate overwhelming superiority in prolonging gestation compared to less specific agents and was found to be no better than placebo for major neonatal outcomes in a major review.[25] The drug's maximum potential may be inherently capped by this paradoxical, self-limiting mechanism.
The pharmacokinetic profile of Atosiban has been characterized primarily in pregnant women, providing a clinically relevant understanding of its disposition. The drug's kinetics do not conform to a simple one- or two-compartment model.[1] A summary of key parameters is provided in Table 3.
Table 3: Summary of Pharmacokinetic Parameters
Parameter | Value | Source(s) |
---|---|---|
Administration Route | Intravenous | 1 |
Time to Steady State | ~1 hour (at 300 µg/min infusion) | 1 |
Volume of Distribution (Vd) | 41.8 L | 1 |
Plasma Protein Binding | 46-48% (in pregnant women) | 1 |
Metabolism | Enzymatic cleavage of peptide bond (Orn⁸-Pro⁷) | 1 |
Half-life (t½) | Biphasic: Initial (tα) = 0.21 h; Terminal (tβ) = 1.7 h | 1 |
Clearance | 41.8 L/h | 1 |
Placental Transfer | Yes; Fetal/Maternal concentration ratio = 0.12 | 1 |
As an intravenously administered peptide, Atosiban has 100% bioavailability. Following the initiation of an infusion at a rate of 300 µg/min, steady-state plasma concentrations of approximately 442 ng/mL are achieved within one hour.[1] The mean volume of distribution is 41.8 L.[1] Atosiban is moderately bound to plasma proteins (46-48%) in pregnant women and is not known to partition into red blood cells.[1]
A critical pharmacokinetic property is its ability to cross the placenta. Studies have shown that at a standard infusion rate, the fetal-to-maternal concentration ratio is approximately 0.12, indicating limited but definite fetal exposure.[1] The difference in the free fraction of the drug between maternal and fetal compartments remains unknown.[1]
Atosiban is subject to metabolic degradation. The primary metabolic pathway involves the enzymatic cleavage of the peptide bond between ornithine at position 8 and proline at position 7. This cleavage is thought to be facilitated by a prior reduction of the disulfide bridge.[1] This process yields two main metabolites. The larger fragment, known as des-(Orn⁸, Gly⁹-NH2)--oxytocin, retains pharmacological activity as an OTR antagonist, but its potency is approximately 10 times lower than that of the parent Atosiban molecule.[3] The plasma concentration ratio of the parent drug to this main metabolite changes over time during an infusion, being 1.4 at the second hour and increasing to 2.8 by the end of the infusion.[3]
Elimination of Atosiban from plasma is biphasic, characterized by a rapid initial half-life (tα) of 0.21 hours (approximately 13 minutes) and a slower terminal half-life (tβ) of 1.7 hours.[1] The mean plasma clearance rate is 41.8 L/h.[1] Renal excretion is a minor route of elimination for the parent drug; only small amounts are found in the urine. However, the main active metabolite is excreted renally in amounts approximately 50 times greater than the parent drug.[1] Information on fecal excretion is not available.[1]
The sole approved indication for Atosiban is the acute treatment of preterm labor, specifically for the purpose of delaying imminent preterm birth.[1] Its use is governed by a precise set of clinical criteria designed to identify the patient population most likely to benefit from therapy while minimizing unnecessary exposure. These criteria are:
The primary clinical objective of Atosiban therapy is to achieve uterine quiescence and delay delivery for a minimum of 48 hours. This critical time window allows for two crucial interventions: the administration of a full course of antenatal corticosteroids to promote fetal lung maturation, thereby reducing the risk of neonatal respiratory distress syndrome, and, if necessary, the safe transfer of the mother to a tertiary care center equipped with a neonatal intensive care unit (NICU).
Beyond its approved use in preterm labor, Atosiban has been investigated for its potential role in improving outcomes in assisted reproductive technology (ART). The underlying rationale for this application is a logical extension of its primary pharmacological effect. Successful embryo implantation requires a quiescent uterine environment, and even subtle, sub-clinical myometrial contractions can interfere with this delicate process. By inducing uterine relaxation, Atosiban may create a more receptive endometrium for the embryo, particularly in challenging cases.
Clinical research has focused on its use in patients undergoing in vitro fertilization-embryo transfer (IVF-ET), especially those with a history of repeated implantation failure.[2] A completed Phase 4 clinical trial (NCT01673399) specifically evaluated Atosiban for the treatment of implantation failure.[28] Another Phase 4 trial (NCT01501214) explored its use in the broader context of subfertility.[29] One study reported a remarkable improvement in the pregnancy rate from zero to 43.7% in a cohort of patients with repeated implantation failure treated with Atosiban, highlighting its potential utility in this difficult-to-treat population.[2] This demonstrates a plausible and promising repurposing of Atosiban's targeted uterine-relaxing properties from preventing birth to facilitating the very initiation of pregnancy.
The clinical value of a tocolytic agent is best understood through its comparison with other available therapies and placebo. Atosiban's position in the therapeutic landscape has been defined by numerous clinical trials and systematic reviews.
Beta-adrenergic agonists (e.g., ritodrine, terbutaline, salbutamol) were once a mainstay of tocolytic therapy. Comparative assessments have consistently shown that Atosiban's tocolytic efficacy is comparable to that of beta-agonists.[2] Large-scale, randomized trials and meta-analyses have found no statistically significant differences in key efficacy endpoints, including the proportion of women remaining undelivered at 48 hours or 7 days, the mean gestational age at delivery, or the mean birthweight of the neonates.[4]
The decisive advantage of Atosiban over beta-agonists lies in its superior safety and tolerability profile. Beta-agonists are non-specific and stimulate beta-receptors throughout the body, leading to a high incidence of maternal side effects, particularly cardiovascular events. In a landmark comparative study, cardiovascular adverse events were reported in 81.2% of women receiving beta-agonists, compared to just 8.3% in the Atosiban group.[4] This profound difference in tolerability also translates to clinical practice, with significantly more patients discontinuing treatment due to adverse events in the beta-agonist arm (15.4%) compared to the Atosiban arm (1.1%).[4] This superior safety profile, coupled with equivalent efficacy, establishes a clear clinical advantage for Atosiban over beta-agonists.[2] Furthermore, economic analyses suggest that despite a higher acquisition cost for the drug itself, the overall cost of care with Atosiban may be lower due to the reduced need for monitoring and management of maternal side effects.[32]
The comparison between Atosiban and the calcium channel blocker nifedipine is more complex, revealing a nuanced trade-off between acute efficacy, long-term prolongation, and maternal safety.
The choice between Atosiban and nifedipine is therefore not one of clear superiority but rather a complex clinical decision that involves prioritizing different therapeutic goals. If the primary objective is to safely and reliably achieve uterine quiescence for the first 48 hours with minimal risk of maternal side effects, Atosiban appears to be the superior choice. If the clinical goal is to achieve the maximum possible prolongation of pregnancy, and the mother is able to tolerate the associated cardiovascular side effects, nifedipine may be considered a viable alternative.
Placebo-controlled trials are the gold standard for establishing efficacy. Such trials have demonstrated that Atosiban significantly increases the proportion of women who remain undelivered at 48 hours compared to placebo.[2] However, a comprehensive 2014 Cochrane systematic review provided a more sobering perspective on its overall impact. The review concluded that, when compared to placebo, Atosiban was not superior in major outcomes such as overall pregnancy prolongation or key neonatal outcomes.[2]
Most critically, this review highlighted a concerning finding from one large placebo-controlled trial: a statistically significant increase in infant deaths (up to 12 months of age) in the group treated with Atosiban (Relative Risk 6.13).[25] While this finding is alarming and cannot be dismissed, it requires careful interpretation. The authors of the review and subsequent analyses noted that this result may have been confounded by a baseline imbalance in the randomization process, where a greater number of women at extremely early gestational ages (less than 26 weeks) were allocated to the Atosiban arm.[25] Extreme prematurity is the single most powerful predictor of neonatal mortality, irrespective of tocolytic treatment. Therefore, it is highly plausible that the observed increase in mortality was driven by the inherently poorer prognosis of this subgroup of infants rather than a direct toxic effect of the drug. This highlights a fundamental challenge in tocolytic research: distinguishing the effects of the intervention from the severe risks of the underlying condition being treated.
Table 4: Comparative Summary of Tocolytic Agents
Feature | Atosiban | Beta-Agonists (e.g., Ritodrine) | Calcium Channel Blockers (Nifedipine) |
---|---|---|---|
Efficacy (Delay at 48h) | Effective; may be superior to nifedipine | Comparable to Atosiban | Effective; may be less reliable than Atosiban |
Efficacy (Delay >7 days) | Effective | Comparable to Atosiban | Effective; may be superior to Atosiban |
Maternal CV Side Effects | Low incidence (e.g., mild tachycardia) | Very high incidence (tachycardia, palpitations) | Moderate to high incidence (hypotension, flushing) |
Other Maternal Side Effects | Common but mild (nausea, headache) | Common (tremor, hyperglycemia) | Common (headache, dizziness) |
Neonatal Outcomes | Generally comparable to others; one trial vs. placebo raised concerns | Comparable to Atosiban | Generally comparable; may reduce RDS vs. Atosiban |
Overall Clinical Conclusion | Comparable efficacy to others with a superior safety profile, especially vs. beta-agonists. | Efficacious but largely superseded due to poor safety and tolerability. | Efficacious, but with a higher burden of maternal side effects than Atosiban. |
Atosiban is generally well-tolerated, with most adverse reactions being of mild severity.[24]
The use of Atosiban is contraindicated in several clinical situations where delaying delivery may be harmful to the mother or fetus, or where the drug's efficacy has not been established. These absolute contraindications are summarized in Table 5.
Table 5: Contraindications for Atosiban Use
Contraindication Category | Specific Condition | Source(s) |
---|---|---|
Gestational Age | <24 or >33 completed weeks | 24 |
Membrane Status | Premature rupture of membranes at >30 weeks gestation | 24 |
Maternal/Fetal Health | Antepartum uterine hemorrhage requiring immediate delivery | 24 |
Eclampsia and severe pre-eclampsia requiring delivery | 24 | |
Intrauterine fetal death | 24 | |
Suspected intrauterine infection | 24 | |
Placental Issues | Placenta previa or placental abruption | 27 |
Hypersensitivity | Known hypersensitivity to Atosiban or excipients | 37 |
In addition to these contraindications, several warnings and precautions must be observed:
Treatment with Atosiban should be initiated and managed by a physician experienced in the treatment of preterm labor.[27] The standard, EMA-approved protocol is a 48-hour intravenous regimen administered in three distinct, successive stages [26]:
The total duration of a single treatment course should not exceed 48 hours, and the total dose of Atosiban administered should preferably not exceed 330.75 mg.[27] If uterine contractions recur after a successful course of treatment, Atosiban therapy can be re-initiated. It is recommended that no more than three re-treatment cycles be used during a single pregnancy.[38]
While the 48-hour protocol is the regulatory standard, its requirement for prolonged hospitalization carries significant cost and resource utilization implications.[45] This has driven a pragmatic evolution in clinical practice and research toward developing shorter, more cost-effective, and convenient dosing regimens. These alternative protocols aim to provide sufficient tocolysis to administer corticosteroids while reducing the burden on both the patient and the healthcare system.
Studies have explored abbreviated regimens, including a 14-hour course and even a single-bolus-dose approach.[44] Research on single-bolus administration (6.75 mg) has demonstrated efficacy in delaying delivery for up to 48 hours in a significant proportion of patients, achieving the primary clinical goal with a dramatically simplified protocol and no reported adverse effects.[45] The development and validation of these shorter protocols could substantially enhance the utility and accessibility of Atosiban, particularly in healthcare settings where cost and bed occupancy are major constraints.
Atosiban represents a significant and targeted pharmacological advancement in the management of preterm labor. Its primary contribution to obstetrical practice is not rooted in a claim of overwhelmingly superior efficacy in prolonging gestation, but rather in its highly specific mechanism of action, which translates directly into a superior safety and tolerability profile.
Its positioning in therapy is clear: Atosiban is a modern alternative to older, non-specific tocolytics like beta-adrenergic agonists. It offers comparable efficacy in delaying delivery but does so with a drastically reduced burden of maternal cardiovascular and systemic side effects, making it a safer first-line option where available. The comparison with nifedipine is more nuanced and does not support a claim of universal superiority for either agent. Instead, the evidence points to a clinical trade-off. Atosiban appears more reliable for achieving acute tocolysis within the critical 48-hour window with minimal maternal risk, making it an excellent choice for facilitating antenatal corticosteroid administration. Nifedipine, conversely, may offer longer-term pregnancy prolongation but at the cost of a higher incidence of maternal side effects.
The drug's clinical profile may be explained, at least in part, by its complex molecular pharmacology. The discovery of its function as a biased ligand—simultaneously antagonizing the pro-contractile Gq pathway while agonizing the pro-inflammatory Gi pathway—suggests an intrinsic molecular mechanism that may limit its maximum potential efficacy. This could explain why a drug with such high receptor specificity is effective and safe, but not a definitive solution for preventing preterm birth.
Looking forward, the development of shorter, more cost-effective dosing regimens is a promising avenue of research that could broaden Atosiban's clinical utility and accessibility. The continued lack of approval in major markets like the United States remains a significant issue, highlighting how commercial and regulatory factors, rather than purely clinical evidence, can shape the therapeutic landscape and create disparities in the standard of care. In conclusion, Atosiban is a valuable, highly specific, and safe tool in the obstetrician's armamentarium for the acute management of preterm labor, whose principal strength lies in its ability to achieve effective tocolysis with an unparalleled level of maternal safety.
Published at: August 19, 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.