Small Molecule
C30H34Cl2FN5O4
1398568-47-2
Beraprost is a synthetic, orally active small molecule drug and a pioneering analogue of prostacyclin ().[1] Its primary mechanism of action is as a potent agonist of the prostacyclin (IP) receptor, initiating an intracellular signaling cascade that results in significant vasodilation and potent inhibition of platelet aggregation.[3] This pharmacological profile has established its therapeutic role in several Asian countries, most notably Japan and South Korea, where it is approved for the treatment of peripheral arterial disease (PAD) and pulmonary arterial hypertension (PAH).[1] For patients with PAD, it offers symptomatic relief from ulcers, pain, and coldness associated with chronic arterial occlusion.[5]
Despite its long-standing use in Asia, Beraprost has faced significant challenges in securing global market access. A critical finding from pivotal clinical trials in PAH conducted in Western populations revealed a pattern of initial efficacy, with improvements in exercise capacity and disease progression observed within the first three to six months of therapy, followed by a notable attenuation of these benefits over longer-term treatment.[6] This lack of sustained efficacy, likely linked to the drug's short pharmacokinetic half-life, proved to be a major obstacle for regulatory approval. Consequently, despite being granted orphan drug designation for PAH in both the United States and Europe, these designations were ultimately withdrawn, and the drug has not received marketing authorization from the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA).[8]
Current research and development efforts are focused on overcoming the limitations of the original immediate-release formulation. The development of modified-release (MR) versions, including a single-isomer formulation, aims to provide more stable plasma concentrations, reduce dosing frequency, and potentially achieve a more durable clinical effect.[10] The success of these next-generation formulations will determine the future role of the Beraprost molecule in the global therapeutic landscape for vasospastic and proliferative vascular diseases.
Beraprost is a small molecule drug classified as a synthetic organic compound.[1] Chemically, it is a complex molecule characterized as a monocarboxylic acid, an organic heterotricyclic compound, a secondary alcohol, a secondary allylic alcohol, and an enyne.[1] The International Nonproprietary Name (INN) registered compound specifies a racemic mixture of two epimers and their enantiomers.[12]
A key structural feature of Beraprost is the replacement of the chemically unstable enol-ether moiety found in natural prostacyclin with a stable inter-m-phenylene benzofuran ether function.[14] This critical modification is responsible for the drug's enhanced stability and increased plasma half-life, which makes it resistant to gastric degradation and thus suitable for oral administration—a significant advantage over intravenously administered prostacyclins like epoprostenol.[14]
The molecular formula for the free acid form of Beraprost is , with a molar mass of 398.499 g·mol⁻¹.[1] It is often administered as its monosodium salt, Beraprost Sodium, which has the molecular formula and a molecular weight of approximately 420.47 g/mol.[17] The sodium salt is a crystalline solid with good solubility in organic solvents like DMF and DMSO and moderate solubility in ethanol and aqueous buffers.[14] The melting point of the free acid is reported to be in the range of 57-60 °C.[20] A comprehensive list of its chemical identifiers is provided in Table 1.
Table 1: Chemical Identifiers and Properties of Beraprost
| Identifier Type | Value (Beraprost - Free Acid) | Value (Beraprost Sodium) |
|---|---|---|
| Common Name | Beraprost | Beraprost Sodium |
| DrugBank ID | DB05229 | - |
| CAS Number | 88430-50-6 | 88475-69-8 16 |
| PubChem CID | 6917951 16 | 23663404 18 |
| IUPAC Name | 4--2,3,3a,8b-tetrahydro-1H-cyclopenta[b]benzofuran-5-yl]butanoic acid | sodium;4--2,3,3a,8b-tetrahydro-1H-cyclopenta[b]benzofuran-5-yl]butanoate 17 |
| Molecular Formula | 17 | |
| Molecular Weight | 398.50 g/mol 16 | 420.47 g/mol 17 |
| UNII | 35E3NJJ4O6 | 15K99VDU5F |
| ChEBI ID | CHEBI:135633 | - |
| ChEMBL ID | CHEMBL1207745 | - |
| InChIKey | CTPOHARTNNSRSR-APJZLKAGSA-N | JLHRLDZCMGGIKW-OQNJEIHKSA-M 18 |
| SMILES | CC#CCC(C)C@@HO | O[C@H]1C(/C=C/C(C)CC#CC)[C@@H]3C1.[Na+] |
| Synonyms & Codes | Dorner, Berasil, Careload LA, Procyclin, TRK 100, MDL 201229, ML 1229, ML 1129 | - |
Beraprost is a potent and stable synthetic analogue of prostacyclin (), a naturally occurring eicosanoid with critical roles in vascular homeostasis. The primary mechanism of action of Beraprost is its function as a direct agonist of the prostacyclin receptor, also known as the IP receptor. The IP receptor is a G protein-coupled receptor (GPCR) predominantly expressed on the surface of vascular smooth muscle cells and platelets.
The activation of the IP receptor by Beraprost initiates a well-defined intracellular signaling cascade. Upon binding, the receptor couples to the stimulatory G protein, Gs. This activates the Gs alpha subunit, which in turn stimulates the membrane-bound enzyme adenylate cyclase. Adenylate cyclase then catalyzes the conversion of adenosine triphosphate (ATP) into the second messenger cyclic adenosine monophosphate (cAMP).
The subsequent elevation of intracellular cAMP levels is the pivotal step in mediating the drug's effects. In vascular smooth muscle cells, increased cAMP activates Protein Kinase A (PKA). PKA proceeds to phosphorylate several downstream targets, including myosin light-chain kinase, which leads to a decrease in intracellular calcium () concentrations by inhibiting its release from intracellular stores and reducing its influx across the cell membrane. This reduction in available intracellular calcium ultimately results in the relaxation of the smooth muscle cells and vasodilation. In platelets, a similar increase in cAMP inhibits platelet activation, adhesion, and aggregation.
While agonism at the IP receptor is its primary mechanism, the pharmacology of Beraprost is more complex. Research has shown that Beraprost also binds to other prostanoid receptors, notably the prostaglandin E receptor 4 (EP4). The EP4 receptor, like the IP receptor, also couples to Gs and stimulates adenylate cyclase activity, contributing to vasodilation. Studies on pulmonary arteries from both human patients with PAH and animal models have demonstrated that the vasodilatory response to Beraprost is partially attenuated by EP4 receptor blockade, confirming that this receptor plays a role in mediating the drug's overall effect. This dual agonism at both IP and EP4 receptors may provide a more robust and multifaceted mechanism for vasodilation, which could be particularly relevant in disease states like PAH where IP receptor expression is known to be downregulated.
The activation of the prostacyclin pathway by Beraprost translates into several key pharmacodynamic effects that form the basis of its therapeutic applications.
Vasodilation: As a direct consequence of its mechanism on vascular smooth muscle, Beraprost is a potent vasodilator, causing relaxation and widening of blood vessels in both the systemic and pulmonary circulations. This effect reduces peripheral and pulmonary vascular resistance, lowers blood pressure, and improves blood flow to tissues. This is the primary effect leveraged for the treatment of PAH, where it helps to reduce the strain on the right ventricle, and in PAD, where it improves perfusion to ischemic limbs.
Platelet Aggregation Inhibition: Beraprost is a powerful inhibitor of platelet aggregation. It effectively prevents platelet activation in response to various agonists like adenosine diphosphate (ADP) and reduces the expression of P-selectin, a key molecule in platelet adhesion. Comparative studies have shown that Beraprost is particularly effective at inhibiting the initial stages of aggregation elicited by thromboxane A₂, a potent pro-thrombotic agent. This antiplatelet action is fundamental to its use in chronic arterial occlusive diseases, where it helps to prevent the formation of microthrombi that can exacerbate ischemia.
Cytoprotective and Anti-proliferative Effects: Beyond its immediate hemodynamic actions, Beraprost exerts protective effects on the vasculature. It has demonstrated cytoprotective properties, helping to shield endothelial cells from damage caused by inflammation and oxidative stress, an effect that may be partially mediated by an enhanced production of nitric oxide (NO). Furthermore, Beraprost inhibits the proliferation of vascular smooth muscle cells. This anti-proliferative effect is highly relevant in PAH, a disease characterized by pathological remodeling and thickening of the pulmonary artery walls. By suppressing smooth muscle cell proliferation, Beraprost may help to slow or mitigate this adverse remodeling process. Additional research has also revealed that Beraprost can inhibit angiotensin II-induced proliferation of cardiac fibroblasts via the TGFβ-Smad signaling pathway, suggesting a potential ancillary benefit in preventing cardiac fibrosis.
The pharmacokinetic profile of Beraprost is characterized by rapid oral absorption, extensive metabolism, and a short elimination half-life, which has profound implications for its clinical use and dosing regimen.
Absorption: Beraprost is administered orally and is well-absorbed from the gastrointestinal tract, with an oral bioavailability reported to be between 50% and 70%. Following oral administration in healthy volunteers, it exhibits rapid absorption, with the time to reach maximum plasma concentration (Tmax) occurring at approximately 0.58 hours. The peak plasma concentration (Cmax) after a single dose was measured to be around 601.14 pg/mL.
Distribution: Specific data regarding the volume of distribution in humans are not widely available in the provided materials. However, as a small molecule targeting GPCRs on vascular endothelial and smooth muscle cells, it is expected to distribute throughout the circulatory system to reach its sites of action.
Metabolism: High-level drug information databases frequently state that the metabolism of Beraprost is "unknown". This statement, however, is an oversimplification that likely reflects a lack of complete metabolic profiling in human registration trials rather than a true absence of metabolic activity. In fact, detailed preclinical studies conducted in animal models (rats and dogs) have demonstrated that Beraprost undergoes extensive biotransformation. The identified metabolic pathways are diverse and include:
The major metabolites identified in these animal studies were 2,3-dinor-beraprost and 13,14-dihydro-2,3-dinor-15-oxo-beraprost. This discrepancy between preclinical data and clinical summaries highlights a knowledge gap that could have implications for predicting drug-drug interactions. While specific human CYP enzyme involvement is not detailed, interaction databases do predict numerous potential metabolic interactions, suggesting that some pathways are known or inferred.
Excretion: The primary route of elimination for Beraprost and its metabolites is through the feces. Animal studies in dogs showed that after an oral dose, approximately 70-74% of the radioactivity was recovered in the feces, while only 15-18% was found in the urine. This is consistent with human data, which show that the cumulative urinary excretion of the unchanged parent drug is very low (less than 1%), indicating that renal clearance is not a significant pathway for the elimination of the parent compound.
Elimination Half-Life: Beraprost has a very short elimination half-life, reported to be between 35 and 40 minutes. This pharmacokinetic property is arguably the most significant determinant of its clinical utility and limitations. The rapid clearance from the body necessitates frequent dosing, typically three to four times daily, to maintain plasma concentrations within the therapeutic range. This creates a high pill burden for patients and can affect adherence. More importantly, this short half-life results in significant fluctuations in plasma drug levels, with peaks that can contribute to dose-limiting side effects (e.g., headache, flushing) and troughs where drug levels may fall below the therapeutic threshold. This pulsatile, rather than continuous, stimulation of the prostacyclin pathway is the most probable reason for the observed attenuation of clinical efficacy in long-term studies of PAH, as the disease may progress during periods of sub-therapeutic drug exposure between doses. This stands in contrast to the continuous stimulation provided by intravenous prostacyclins, which have demonstrated more durable benefits.
The clinical utility of Beraprost has been extensively evaluated in two primary therapeutic areas: pulmonary arterial hypertension (PAH) and peripheral arterial disease (PAD). The evidence base reveals a complex picture of efficacy that varies by indication and duration of treatment.
Beraprost is approved for the treatment of PAH in Japan and South Korea and has been studied globally in patients with WHO Functional Class II and III idiopathic PAH, as well as PAH associated with collagen vascular disease or repaired congenital systemic-to-pulmonary shunts.
Pivotal clinical trials, including the Arterial Pulmonary Hypertension and Beraprost European Trial (ALPHABET) and a large 12-month U.S. Phase III study, have defined the drug's efficacy profile in this indication. These studies were typically randomized, double-blind, and placebo-controlled, with primary endpoints focused on exercise capacity (measured by the 6-minute walk test, 6MWT) or a composite of disease progression (including death, need for lung transplantation, or clinical worsening).
A consistent and critical finding emerged from this body of research: the therapeutic benefit of immediate-release Beraprost in PAH is transient. The U.S. trial, for instance, found that patients treated with Beraprost had a statistically significant improvement in 6MWT distance compared to placebo at 3 months (+22 meters) and 6 months (+31 meters). Similarly, the primary endpoint of disease progression was significantly lower in the Beraprost group at the 6-month mark (p=0.002). However, these advantages were not sustained; by the 9-month and 12-month assessments, the differences between the Beraprost and placebo groups were no longer statistically significant. This attenuation of effect over time has been a major factor limiting its adoption in Western countries and has led to its weak recommendation in international PAH treatment guidelines.
Despite these findings, Beraprost remains a frequently used therapy for PAH in Japan. This may be due to its oral availability, lower cost compared to other agents, and long-standing clinical experience. A retrospective Japanese study suggested that long-term, high-dose Beraprost therapy (>120 μg/day) was associated with a survival benefit compared to conventional therapy, particularly in the subgroup of patients with PAH related to connective tissue disease.
Table 2: Summary of Key Clinical Trials for Beraprost in Pulmonary Arterial Hypertension (PAH)
| Trial Name / Identifier | Patient Population | Design | Primary Endpoint | Key Secondary Endpoint | Results & Significance |
|---|---|---|---|---|---|
| Barst et al., JACC 2003 (U.S. Study) | 116 patients; WHO FC II/III PAH (idiopathic, collagen vascular, or congenital shunt) | 12-month, randomized, double-blind, placebo-controlled | Disease Progression (death, transplant, epoprostenol rescue, >25% decrease in peak ) | Change in 6-minute walk test (6MWT) from baseline | Disease Progression: Significant reduction vs. placebo at 6 months (p=0.002), but not significant at 12 months. 6MWT: Significant improvement vs. placebo at 3 months (+22m, p=0.010) and 6 months (+31m, p=0.016), but not significant at 9 or 12 months. |
| ALPHABET Study (Galiè et al.) | 130 patients; WHO FC II/III PPH or associated PAH | 12-week, randomized, double-blind, placebo-controlled | Change in 6MWT from baseline at week 12 | Cardiopulmonary hemodynamics, Borg dyspnea index | 6MWT: Significant improvement in the Beraprost group vs. placebo (treatment effect +25.1 m, p=0.04). Hemodynamics: No significant changes observed in hemodynamic parameters after 12 weeks. |
Beraprost is widely approved and used in Japan and other Asian countries for the treatment of chronic arterial occlusive diseases, including Buerger's disease and arteriosclerosis obliterans. Its therapeutic goal in PAD is to improve ischemic symptoms such as ulcers, rest pain, and coldness of the extremities.
The clinical evidence for Beraprost in PAD is more favorable than in PAH, although some conflicting results exist. A large, randomized, double-blind study demonstrated that Beraprost was as effective as the antiplatelet agent ticlopidine in treating patients with PAD, showing improvements in ulcer size and pain. The Beraprost et Claudication Intermittent (BERCI-2) study, a multicenter trial in Europe, found that Beraprost significantly increased both pain-free and absolute walking distances in patients suffering from intermittent claudication. Furthermore, a study focused specifically on diabetic patients with PAD reported significant improvements in a range of subjective leg symptoms, including burning, coldness, edema, and exertional pain, after 12 weeks of treatment.
However, the evidence is not uniformly positive. A large, Phase III, placebo-controlled trial conducted in the United States did not confirm the positive walking-distance results from the European trials and failed to show a statistically significant improvement in maximal walking distance among patients with intermittent claudication.
The differing outcomes between PAH and PAD may relate to the underlying pathophysiology of the diseases. In PAD, the primary issues are often related to flow limitation and microcirculatory dysfunction, which may be amenable to the potent but intermittent vasodilatory and antiplatelet effects of Beraprost. In contrast, PAH is a more aggressive and progressive disease characterized by severe, fixed vascular remodeling, which may require the more continuous and sustained pathway stimulation that immediate-release Beraprost fails to provide.
The safety profile of Beraprost is well-characterized and is largely predictable based on its pharmacology as a prostacyclin analogue. Adverse events are common and often dose-limiting, particularly during the initial dose-titration phase.
The most frequently reported adverse reactions are direct extensions of Beraprost's potent vasodilatory effects. These include headache, flushing (manifesting as hot flushes, facial flush, or a sensation of warmth), dizziness, and lightheadedness. Gastrointestinal disturbances, such as nausea, diarrhea, and abdominal discomfort, are also very common. Jaw pain, particularly upon chewing, is another recognized side effect of prostacyclin pathway agonists.
Due to its antiplatelet properties, Beraprost is associated with an increased risk of bleeding. Common hemorrhagic events include a general bleeding tendency, subcutaneous bleeding (bruising), and epistaxis (nasal bleeding).
While most side effects are mild to moderate, several rare but serious adverse reactions have been reported, necessitating careful patient monitoring and immediate medical intervention if they occur. These include major hemorrhagic events (cerebral, gastrointestinal, pulmonary, or ocular), shock or syncope (often secondary to severe hypotension), interstitial pneumonia, severe hepatic dysfunction with jaundice, and major adverse cardiovascular events like angina pectoris and myocardial infarction.
Table 3: Common and Serious Adverse Drug Reactions Associated with Beraprost
| System Organ Class | Common Adverse Reactions | Serious (Rare) Adverse Reactions |
|---|---|---|
| Nervous system disorders | Headache, dizziness, lightheadedness | Cerebral hemorrhage, syncope, loss of consciousness |
| Vascular disorders | Flushing, hot flushes, hypotension | Shock |
| Gastrointestinal disorders | Nausea, diarrhea, abdominal pain, GI upset | Gastrointestinal hemorrhage |
| Blood and lymphatic system disorders | Bleeding tendency, subcutaneous bleeding, nasal bleeding, anemia | Pulmonary hemorrhage, bleeding of the ocular fundus |
| Cardiac disorders | Palpitations | Angina pectoris, myocardial infarction |
| Hepatobiliary disorders | Elevated liver enzymes, elevated bilirubin | Hepatic dysfunction, jaundice |
| Respiratory, thoracic and mediastinal disorders | - | Interstitial pneumonia |
| Skin and subcutaneous tissue disorders | Rash, eczema, itch, hives | Severe allergic reactions |
There are specific patient populations in whom Beraprost should not be used or should be administered with significant caution.
Absolute Contraindications:
Precautions and Populations Requiring Careful Administration:
The most clinically significant drug interactions with Beraprost are pharmacodynamic in nature and relate to its antiplatelet effects.
Beraprost is administered orally, and to minimize gastrointestinal side effects, it is typically taken in divided doses after meals. The dosage varies depending on the indication and patient tolerability.
Formulations:
The regulatory history of Beraprost is marked by a significant divergence between its acceptance in Asia and its status in Western markets, primarily driven by differences in the interpretation of its clinical trial data and risk-benefit profile.
Table 4: Global Regulatory Status of Beraprost
| Regulatory Agency | Status | Approved Indications | Key Dates & Notes |
|---|---|---|---|
| PMDA (Japan) | Approved | Peripheral Arterial Disease; Pulmonary Arterial Hypertension | Launched for PAD in 1992; for PAH in 2000. Remains a widely used therapy. |
| FDA (USA) | Not Approved | N/A | Orphan Designation granted Dec 22, 2011; Designation withdrawn Jan 17, 2020. |
| EMA (Europe) | Not Approved | N/A | Orphan Designation granted Sep 18, 2001; Designation withdrawn Feb 2004. |
| TGA (Australia) | Status Unknown | N/A | No evidence of submission or approval in the provided materials. |
The primary focus of current and future research on Beraprost is to address the well-documented limitations of its original immediate-release formulation. The central challenges to overcome are its very short pharmacokinetic half-life, which necessitates an inconvenient, frequent dosing schedule, and the resulting attenuation of clinical effect observed in long-term PAH trials.
The main strategy being pursued is the development of advanced, modified-release (MR) formulations. A key candidate in late-stage clinical development is BPS-314d-MR, which represents a sophisticated, dual-pronged approach to enhancing the molecule's therapeutic potential. First, it is a single-isomer formulation. The original Beraprost is a racemic mixture, and it is a common principle in pharmacology that one isomer (enantiomer) may be responsible for the majority of the therapeutic activity while the other may be inactive or contribute to side effects. Isolating the pharmacologically active isomer is a classic strategy to create a more potent and potentially better-tolerated drug. Second, it incorporates modified-release technology. This is designed to slow the dissolution and absorption of the drug, thereby prolonging its presence in the circulation.
The goals of this MR formulation are twofold. From a patient perspective, it is designed to reduce the dosing frequency from three or four times daily to a more convenient twice-daily regimen, which is expected to significantly improve treatment adherence. From a pharmacological perspective, the more critical goal is to achieve more stable, sustained plasma concentrations. By smoothing out the sharp peaks and deep troughs associated with the immediate-release version, the MR formulation aims to provide more continuous stimulation of the prostacyclin pathway. The underlying hypothesis is that this sustained action may lead to a more durable clinical benefit, potentially overcoming the attenuation of efficacy that has so far limited Beraprost's use in PAH globally.
These next-generation formulations are currently being evaluated in Phase III clinical trials, often as an add-on therapy to other established PAH treatments like treprostinil, reflecting the modern paradigm of combination therapy for this complex disease. The outcomes of these trials will be decisive in determining whether a reformulated Beraprost can finally secure a place in the global therapeutic armamentarium for PAH.
Beraprost holds a significant place in pharmaceutical history as the first chemically stable and orally active prostacyclin analogue, a landmark achievement that offered a convenient alternative to the cumbersome continuous intravenous infusions required for earlier prostacyclins. Its pharmacology is well-understood, centered on potent agonism of the IP and EP4 receptors, which translates into powerful vasodilation and antiplatelet effects.
However, the clinical and regulatory story of Beraprost is one of profound regional divergence. In Japan and several other Asian nations, it has been a therapeutic mainstay for decades, providing valuable symptomatic relief for patients with peripheral arterial disease and pulmonary arterial hypertension. In stark contrast, its development in the West was ultimately unsuccessful. Rigorous, long-term clinical trials in PAH, while demonstrating clear short-term benefits, consistently revealed an attenuation of efficacy over time. This failure to provide a durable, long-term benefit—a critical requirement for a chronic and progressive disease—led to the withdrawal of its orphan drug designations and prevented its approval by the FDA and EMA.
The core of this clinical limitation appears to be inextricably linked to the drug's fundamental pharmacokinetic profile. Its very short elimination half-life of 35-40 minutes dictates a frequent dosing regimen that produces pulsatile, rather than continuous, stimulation of the prostacyclin pathway. This intermittent effect, while sufficient to provide symptomatic relief in some forms of PAD, proved inadequate to halt the relentless progression of severe vascular remodeling in PAH over the long term.
The future of the Beraprost molecule now rests on the success of its second-generation, modified-release formulations. These advanced products, which combine a refined single isomer with controlled-release technology, are a direct and scientifically sound attempt to correct the pharmacokinetic deficiencies of the original drug. If ongoing Phase III trials can demonstrate that these new formulations provide the sustained efficacy that their predecessor lacked, Beraprost may yet be repositioned as a valuable component of combination therapy for PAH on a global scale. Ultimately, the journey of Beraprost serves as a compelling case study in drug development, powerfully illustrating that a drug's ultimate success is defined not by its mechanism of action alone, but by the crucial and complex interplay between its pharmacokinetics, the specific pathophysiology of the disease it is intended to treat, and the rigorous, evolving evidence standards of global regulatory bodies.
Published at: October 8, 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.